101
|
Antioxidant Properties of Fucoidan Alleviate Acceleration and Exacerbation of Hippocampal Neuronal Death Following Transient Global Cerebral Ischemia in High-Fat Diet-Induced Obese Gerbils. Int J Mol Sci 2019; 20:ijms20030554. [PMID: 30696078 PMCID: PMC6387260 DOI: 10.3390/ijms20030554] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/18/2019] [Accepted: 01/27/2019] [Indexed: 01/01/2023] Open
Abstract
Fucoidan, a natural sulfated polysaccharide, displays various biological activities including antioxidant properties. We examined the neuroprotective effect of fucoidan against transient global cerebral ischemia (tGCI) in high-fat diet (HFD)-induced obese gerbils and its related mechanisms. Gerbils received HFD for 12 weeks and fucoidan (50 mg/kg) daily for the last 5 days during HFD exposure, and they were subjected to 5-min tGCI. Pyramidal cell death was observed only in the CA 1 area (CA1) of the hippocampus in non-obese gerbils 5 days after tGCI. However, in obese gerbils, pyramidal cell death in the CA1 and CA2/3 occurred at 2 days and 5 days, respectively, after tGCI. In the obese gerbils, oxidative stress indicators (dihydroethidium, 8-hydroxyguanine and 4-hydroxy-2-nonenal) were significantly enhanced and antioxidant enzymes (SOD1 and SOD2) were significantly reduced in pre- and post-ischemic phases compared to the non-obese gerbils. Fucoidan treatment attenuated acceleration and exacerbation of tGCI-induced neuronal death in the CA1–3, showing that oxidative stress was significantly reduced, and antioxidant enzymes were significantly increased in pre- and post-ischemic phases. These findings indicate that pretreated fucoidan can relieve the acceleration and exacerbation of ischemic brain injury in an obese state via the attenuation of obesity-induced severe oxidative damage.
Collapse
|
102
|
Shih EK, Robinson MB. Role of Astrocytic Mitochondria in Limiting Ischemic Brain Injury? Physiology (Bethesda) 2019; 33:99-112. [PMID: 29412059 DOI: 10.1152/physiol.00038.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Until recently, astrocyte processes were thought to be too small to contain mitochondria. However, it is now clear that mitochondria are found throughout fine astrocyte processes and are mobile with neuronal activity resulting in positioning near synapses. In this review, we discuss evidence that astrocytic mitochondria confer selective resiliency to astrocytes during ischemic insults and the functional significance of these mitochondria for normal brain function.
Collapse
Affiliation(s)
- Evelyn K Shih
- Children's Hospital of Philadelphia Research Institute , Philadelphia, Pennsylvania.,Children's Hospital of Philadelphia, Division of Neurology , Philadelphia, Pennsylvania.,Department of Pediatrics, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Michael B Robinson
- Children's Hospital of Philadelphia Research Institute , Philadelphia, Pennsylvania.,Department of Pediatrics, University of Pennsylvania , Philadelphia, Pennsylvania.,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania , Philadelphia, Pennsylvania
| |
Collapse
|
103
|
Cengiz P, Zafer D, Chandrashekhar JH, Chanana V, Bogost J, Waldman A, Novak B, Kintner DB, Ferrazzano PA. Developmental differences in microglia morphology and gene expression during normal brain development and in response to hypoxia-ischemia. Neurochem Int 2019; 127:137-147. [PMID: 30639264 DOI: 10.1016/j.neuint.2018.12.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/27/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Neuroinflammation plays an important role in ischemic brain injury and recovery, however the interplay between brain development and the neuroinflammatory response is poorly understood. We previously described age-dependent differences in the microglial response and the effect of microglial inhibition. Here we investigate whether age-dependent microglial responses may be related to pre-injury developmental differences in microglial phenotype. METHODS Measures of microglia morphology were quantified using semi-automated software analysis of immunostained sections from postnatal day 2 (P2), P9, P30 and P60 mice using IMARIS. Microglia were isolated from P2, P9, P30 and P60 mice, and expression of markers of classical and alternative microglial activation was assessed, as well as transforming growth factor beta (TGF-β) receptor, Serpine1, Mer Tyrosine Kinase (MerTK), and the suppressor of cytokine signaling (SOCS3). Hypoxia-ischemia (HI) was induced in P9 and P30 mice using unilateral carotid artery ligation and exposure to 10% oxygen for 50 min. Microglia morphology and microglial expression of genes in the TGF-β and MerTK pathways were determined in ipsilateral and contralateral hippocampus. RESULTS A progressive and significant increase in microglia branching morphology was seen in all brain regions from P2 to P30. No consistent classical or alternative activation profile was seen in isolated microglia. A clear transition to increased expression of TGF-β and its downstream effector serpine1 was seen between P9 and P30. A similar increase in expression was seen in MerTK and its downstream effector SOCS3. HI resulted in a significant decrease in branching morphology only in the P9 mice, and expression of TGF-β receptor, Serpine1, MerTK, and SOCS3 were elevated in P30 mice compared to P9 post-HI. CONCLUSION Microglia maturation is associated with changes in morphology and gene expression, and microglial responses to ischemia in the developing brain differ based on the age at which injury occurs.
Collapse
Affiliation(s)
- Pelin Cengiz
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Dila Zafer
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jayadevi H Chandrashekhar
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; University of Illinois at Urbana-Champaign, IL, USA
| | - Vishal Chanana
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacob Bogost
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Alex Waldman
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Emory University School of Medicine, Atlanta, GA, USA
| | - Becca Novak
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Douglas B Kintner
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peter A Ferrazzano
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
104
|
Lee TK, Kim H, Song M, Lee JC, Park JH, Ahn JH, Yang GE, Kim H, Ohk TG, Shin MC, Cho JH, Won MH. Time-course pattern of neuronal loss and gliosis in gerbil hippocampi following mild, severe, or lethal transient global cerebral ischemia. Neural Regen Res 2019; 14:1394-1403. [PMID: 30964065 PMCID: PMC6524495 DOI: 10.4103/1673-5374.253524] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Transient ischemia in the whole brain leads to neuronal loss/death in vulnerable brain regions. The striatum, neocortex and hippocampus selectively loose specific neurons after transient ischemia. Just 5 minutes of transient ischemia can cause pyramidal neuronal death in the hippocampal cornu ammonis (CA) 1 field at 4 days after transient ischemia. In this study, we investigated the effects of 5-minute (mild), 15-minute (severe), and 20-minute (lethal) transient ischemia by bilateral common carotid artery occlusion (BCCAO) on behavioral change and neuronal death and gliosis (astrocytosis and microgliosis) in gerbil hippocampal subregions (CA1–3 region and dentate gyrus). We performed spontaneous motor activity test to evaluate gerbil locomotor activity, cresyl violet staining to detect cellular distribution, neuronal nuclei immunohistochemistry to detect neuronal distribution, and Fluoro-Jade B histofluorescence to evaluate neuronal death. We also conducted immunohistochemical staining for glial fibrillary acidic protein and ionized calcium-binding adapter molecule 1 (Iba1) to evaluate astrocytosis and microgliosis, respectively. Animals subjected to 20-minute BCCAO died in at least 2 days. BCCAO for 15 minutes led to pyramidal cell death in hippocampal CA1–3 region 2 days later and granule cell death in hippocampal dentate gyrus 5 days later. Similar results were not found in animals subjected to 5-minute BCCAO. Gliosis was much more rapidly and severely progressed in animals subjected to 15-minute BCCAO than in those subjected to 5-minute BCCAO. Our results indicate that neuronal loss in the hippocampal formation following transient ischemia is significantly different according to regions and severity of transient ischemia. The experimental protocol was approved by Institutional Animal Care and Use Committee (AICUC) of Kangwon National University (approval No. KW-180124-1) on May 22, 2018.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon, Republic of Korea
| | - Go Eun Yang
- Department of Radiology, Kangwon National University Hospital, Chuncheon, Gangwon, Republic of Korea
| | - Hyeyoung Kim
- Department of Anesthesiology and Pain Medicine, Chungju Hospital, Konkuk University School of Medicine, Chungju Chungcheongbuk; Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| |
Collapse
|
105
|
Petschner P, Balogh N, Adori C, Tamasi V, Kumar S, Juhasz G, Bagdy G. Downregulation of the Vitamin D Receptor Regulated Gene Set in the Hippocampus After MDMA Treatment. Front Pharmacol 2018; 9:1373. [PMID: 30559663 PMCID: PMC6287013 DOI: 10.3389/fphar.2018.01373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/08/2018] [Indexed: 11/13/2022] Open
Abstract
The active ingredient of ecstasy, ±3,4-methylenedioxymethamphetamine (MDMA), in addition to its initial reinforcing effects, induces selective and non-selective brain damage. Evidences suggest that the hippocampus (HC), a central region for cognition, may be especially vulnerable to impairments on the long-run, nevertheless, transcription factors that may precede and regulate such chronic changes remained uninvestigated in this region. In the current study, we used gene-set enrichment analysis (GSEA) to reveal possible transcription factor candidates responsible for enhanced vulnerability of HC after MDMA administration. Dark Agouti rats were intraperitoneally injected with saline or 15 mg/kg MDMA. Three weeks later HC gene expression was measured by Illumina whole-genome beadarrays and GSEA was performed with MSigDB transcription factor sets. The number of significantly altered genes on the genome level (significance < 0.001) in up/downregulated sets was also counted. MDMA upregulated one, and downregulated 13 gene sets in the HC of rats, compared to controls, including Pax4, Pitx2, FoxJ2, FoxO1, Oct1, Sp3, AP3, FoxO4, and vitamin D receptor (VDR)-regulated sets (q-value <0.05). VDR-regulated set contained the second highest number of significantly altered genes, including among others, Camk2n2, Gria3, and Grin2a. Most identified transcription factors are implicated in the response to ischemia confirming that serious hypoxia/ischemia occurs in the HC after MDMA administration, which may contribute to the selective vulnerability of this brain region. Moreover, our results also raise the possibility that vitamin D supplementation, in addition to the commonly used antioxidants, could be a potential alternative method to attenuate MDMA-induced chronic hippocampal impairments.
Collapse
Affiliation(s)
- Peter Petschner
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary.,MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - Noemi Balogh
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | - Csaba Adori
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | - Viola Tamasi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Sahel Kumar
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary.,SE-NAP 2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary.,NAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary.,MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary.,NAP-2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| |
Collapse
|
106
|
Marchese E, Corvino V, Di Maria V, Furno A, Giannetti S, Cesari E, Lulli P, Michetti F, Geloso MC. The Neuroprotective Effects of 17β-Estradiol Pretreatment in a Model of Neonatal Hippocampal Injury Induced by Trimethyltin. Front Cell Neurosci 2018; 12:385. [PMID: 30416427 PMCID: PMC6213803 DOI: 10.3389/fncel.2018.00385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Hippocampal dysfunction plays a central role in neurodevelopmental disorders, resulting in severe impairment of cognitive abilities, including memory and learning. On this basis, developmental studies represent an important tool both to understanding the cellular and molecular phenomena underlying early hippocampal damage and to study possible therapeutic interventions, that may modify the progression of neuronal death. Given the modulatory role played by 17β-estradiol (E2) on hippocampal functions and its neuroprotective properties, the present study investigates the effects of pretreatment with E2 in a model of neonatal hippocampal injury obtained by trimethyltin (TMT) administration, characterized by neuronal loss in CA1 and CA3 subfields and astroglial and microglial activation. At post-natal days (P)5 and P6 animals received E2 administration (0.2 mg/kg/die i.p.) or vehicle. At P7 they received a single dose of TMT (6.5 mg/kg i.p.) and were sacrificed 72 h (P10) or 7 days after TMT treatment (P14). Our findings indicate that pretreatment with E2 exerts a protective effect against hippocampal damage induced by TMT administration early in development, reducing the extent of neuronal death in the CA1 subfield, inducing the activation of genes involved in neuroprotection, lowering the neuroinflammatory response and restoring neuropeptide Y- and parvalbumin- expression, which is impaired in the early phases of TMT-induced damage. Our data support the efficacy of estrogen-based neuroprotective approaches to counteract early occurring hippocampal damage in the developing hippocampus.
Collapse
Affiliation(s)
- Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alfredo Furno
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Giannetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Cesari
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Paola Lulli
- Laboratorio di Biochimica Clinica e Biologia Molecolare, IRCCS Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Facoltà di Medicina e Chirurgia - IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
107
|
Inhibiting Succinate Dehydrogenase by Dimethyl Malonate Alleviates Brain Damage in a Rat Model of Cardiac Arrest. Neuroscience 2018; 393:24-32. [PMID: 30300703 DOI: 10.1016/j.neuroscience.2018.09.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 01/08/2023]
Abstract
Brain damage is a leading cause of death in patients with cardiac arrest (CA). The accumulation of succinate during ischemia by succinate dehydrogenase (SDH) is an important mechanism of ischemia-reperfusion injury. It was unclear whether inhibiting the oxidation of accumulated succinate could also mitigate brain damage after CA. In this study, rats were subjected to a 6 min of CA, and cardiopulmonary resuscitation (CPR) was performed with administration of normal saline or dimethyl malonate (DMM, a competitive inhibitor of SDH). After the return of spontaneous circulation, neurological function of the rats was assessed by a tape removal test for 3 days. The rats were then sacrificed, and their brains were used to assess neuronal apoptosis by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Hippocampal tissues were used for Western blotting analysis and biochemical detection. In addition, hippocampal mitochondria during CA and CPR were isolated. The relative mitochondrial membrane potential (MMP) and cytochrome C in the cytosol were detected. Our results show that DMM promoted ROSC and neurological performance in rats after CA. The TUNEL assay showed that DMM reduced neuronal apoptosis. Western blotting analysis showed that DMM inhibited the activation of caspase-3 and enhanced the expression of HIF-1α. Moreover, DMM inhibited excessive hyperpolarization of MMP after CPR, and prevented the release of cytochrome C. Therefore, inhibiting SDH by DMM alleviated brain damage after CA, and the main mechanisms included inhibiting the excessive hyperpolarization of MMP, reducing the generation of mtROS and stabilizing the structure of HIF-1α.
Collapse
|
108
|
Wu Y, Chen C, Yang Q, Jiao M, Qiu S. Endocytosis of GluN2B-containing NMDA receptors mediates NMDA-induced excitotoxicity. Mol Pain 2018; 13:1744806917701921. [PMID: 28326942 PMCID: PMC5391130 DOI: 10.1177/1744806917701921] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract N-methyl-D-aspartate (NMDA) receptor overactivation is involved in neuronal damage after stroke. However, the mechanism underlying NMDA receptor-mediated excitotoxicity remains unclear. In this study, we confirmed that excessive activation of NMDARs led to cell apoptosis in PC12 cells and in primary cultured cortical neurons, which was mediated predominantly by the GluN2B-containing, but not the GluN2A-containing NMDARs. In addition, Clathrin-dependent endocytosis participated in NMDA-induced excitotoxicity. Furthermore, we identified that GluN2B-containing NMDARs underwent endocytosis during excessive NMDA treatment. Peptides specifically disrupting the interaction between GluN2B and AP-2 complex not only blocked endocytosis of GluN2B induced by NMDA treatment but also abolished NMDA-induced excitotoxicity. These results demonstrate that Clathrin-dependent endocytosis of GluN2B-containing NMDARs is critical to NMDA-induced excitotoxicity in PC12 cells and in primary cultured cortical neurons, and therefore provide a novel target for blocking NMDAR-mediated excitotoxicity.
Collapse
Affiliation(s)
- Yu Wu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Changwan Chen
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qian Yang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mingfei Jiao
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shuang Qiu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Shuang Qiu, Department of Neurobiology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
109
|
[Neurogenesis and gliogenesis modulation in cerebral ischemia by CDK5 RNAi-based therapy]. BIOMEDICA 2018; 38:388-397. [PMID: 30335244 DOI: 10.7705/biomedica.v38i4.3800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/15/2018] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Cerebral ischemia is the third cause of death risk in Colombia and the first cause of physical disability worldwide. Different studies on the silencing of the cyclin-dependent kinase 5 (CDK5) have shown that reducing its activity is beneficial in ischemic contexts. However, its effect on neural cell production after cerebral ischemia has not been well studied yet. OBJECTIVE To evaluate CDK5 silencing on the production of neurons and astrocytes after a focal cerebral ischemia in rats. MATERIALS AND METHODS We used 40 eight-week-old male Wistar rats. Both sham and ischemia groups were transduced at CA1 hippocampal region with an adeno-associated viral vector using a noninterfering (shSCRmiR) and an interfering sequence for CDK5 (shCDK5miR). We injected 50 mg/kg of bromodeoxyuridine intraperitoneally from hour 24 to day 7 post-ischemia. We assessed the neurological abilities during the next 15 days and we measured the immunoreactivity of bromodeoxyuridine (BrdU), doublecortin (DCX), NeuN, and glial fibrillary acid protein (GFAP) from day 15 to day 30 post-ischemia. RESULTS Our findings showed that CDK5miR-treated ischemic animals improved their neurological score and presented increased BrdU+ cells 15 days after ischemia, which correlated with higher DCX and lower GFAP fluorescence intensities, and, although mature neurons populations did not change, GFAP immunoreactivity was still significantly reduced at 30 days post-ischemia in comparison with untreated ischemic groups. CONCLUSION CDK5miR therapy generated the neurological recovery of ischemic rats associated with the induction of immature neurons proliferation and the reduction of GFAP reactivity at short and longterm post-ischemia.
Collapse
|
110
|
Kirschen GW, Kéry R, Ge S. The Hippocampal Neuro-Glio-Vascular Network: Metabolic Vulnerability and Potential Neurogenic Regeneration in Disease. Brain Plast 2018; 3:129-144. [PMID: 30151338 PMCID: PMC6091038 DOI: 10.3233/bpl-170055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Brain metabolism is a fragile balance between nutrient/oxygen supply provided by the blood and neuronal/glial demand. Small perturbations in these parameters are necessary for proper homeostatic functioning and information processing, but can also cause significant damage and cell death if dysregulated. During embryonic and early post-natal development, massive neurogenesis occurs, a process that continues at a limited rate in adulthood in two neurogenic niches, one in the lateral ventricle and the other in the hippocampal dentate gyrus. When metabolic demand does not correspond with supply, which can occur dramatically in the case of hypoxia or ischemia, or more subtly in the case of neuropsychiatric or neurodegenerative disorders, both of these neurogenic niches can respond—either in a beneficial manner, to regenerate damaged or lost tissue, or in a detrimental fashion—creating aberrant synaptic connections. In this review, we focus on the complex relationship that exists between the cerebral vasculature and neurogenesis across development and in disease states including hypoxic-ischemic injury, hypertension, diabetes mellitus, and Alzheimer’s disease. Although there is still much to be elucidated, we are beginning to appreciate how neurogenesis may help or harm the metabolically-injured brain, in the hopes that these insights can be used to tailor novel therapeutics to regenerate damaged tissue after injury.
Collapse
Affiliation(s)
- Gregory W Kirschen
- Medical Scientist Training Program (MSTP), Stony Brook Medicine, Stony Brook, NY, USA.,Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Rachel Kéry
- Medical Scientist Training Program (MSTP), Stony Brook Medicine, Stony Brook, NY, USA.,Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Shaoyu Ge
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
111
|
Huang YJ, Yuan YJ, Liu YX, Zhang MY, Zhang JG, Wang TC, Zhang LN, Hu YY, Li L, Xian XH, Qi J, Zhang M. Nitric Oxide Participates in the Brain Ischemic Tolerance Induced by Intermittent Hypobaric Hypoxia in the Hippocampal CA1 Subfield in Rats. Neurochem Res 2018; 43:1779-1790. [PMID: 29995175 DOI: 10.1007/s11064-018-2593-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/23/2018] [Accepted: 07/05/2018] [Indexed: 12/30/2022]
Abstract
Previous studies have shown that intermittent hypobaric hypoxia (IH) preconditioning protected neurons survival from brain ischemia. However, the mechanism remains to be elucidated. The present study explored the role of nitric oxide (NO) in the process by measuring the expression of NO synthase (NOS) and NO levels. Male Wistar rats (100) were randomly assigned into four groups: sham group, IH + sham group, ischemia group and IH + ischemia group. Rats for IH preconditioning were exposed to hypobaric hypoxia mimicking 5000 m high-altitude (PB = 404 mmHg, PO2 = 84 mmHg) 6 h/day, once daily for 28 days. Global brain ischemia was established by four-vessel occlusion that has been created by Pulsinelli. Rats were sacrificed at 7th day after the ischemia for neuropathological evaluation by thionin stain. In addition, the expression of neuronal NOS (nNOS), inducible NOS (iNOS), and NO content in the hippocampal CA1 subfield were measured at 2nd day and 7th day after the ischemia. Results revealed that global brain ischemia engendered delayed neuronal death (DND), both nNOS and iNOS expression up-regulated, and NO content increased in the hippocampal CA1 subfield. IH preconditioning reduced neuronal injury induced by the ischemia, and prevented the up-regulation of NOS expression and NO production. In addition, L-NAME + ischemia group was designed to detect whether depressing NO production could alleviate the DND. Pre-administration of L-NAME alleviated DND induced by the ischemia. These results suggest that IH preconditioning plays a protective role by inhibiting the over expression of NOS and NO content after brain ischemia.
Collapse
Affiliation(s)
- Ya-Jie Huang
- Undergraduate of Clinical Medicine, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Yu-Jia Yuan
- Undergraduate of Clinical Medicine, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Yi-Xian Liu
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Meng-Yue Zhang
- Undergraduate of Clinical Medicine, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Jing-Ge Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China.
| | - Tian-Ci Wang
- Undergraduate of Clinical Medicine, Hebei Medical University, Shijiazhuang, 050017, People's Republic of China
| | - Li-Nan Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Yu-Yan Hu
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Li Li
- Department of Science and Technology, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, People's Republic of China
| | - Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Jie Qi
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China. .,Aging and Cognition Neuroscience Laboratory of Hebei Province, Shijiazhuang, 050017, People's Republic of China.
| |
Collapse
|
112
|
Fleischman DA, Arfanakis K, Leurgans S, Keating SM, Lamar M, Bennett DA, Adeyemi OM, Barnes LL. Neopterin is associated with hippocampal subfield volumes and cognition in HIV. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2018; 5:e467. [PMID: 29904644 PMCID: PMC5999345 DOI: 10.1212/nxi.0000000000000467] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 04/04/2018] [Indexed: 11/27/2022]
Abstract
Objective HIV infection sets off an immediate immune response and inflammatory cascade that can lead to neuronal injury and cognitive impairment, but the relationship between immune markers, regional brain volumes, and cognition remains understudied in HIV-infected adults. Methods Cross-sectional associations were examined between serum immune markers of activation (neopterin) and inflammation (interleukin [IL]-1β, IL-6, tumor necrosis factor alpha, and C-reactive protein) with regional brain volumes (cortical, subcortical, total gray matter, hippocampus, and subfields) and cognition in 66 HIV-infected, virally suppressed, adults who underwent 3.0-T MRI as part of the Research Core of the Rush Center of Excellence on Disparities in HIV and Aging. Immune markers were assayed from frozen plasma, values were entered into linear regression models as predictors of regional brain volumes, and interactive effects of immune response and regional brain volumes on cognition were examined. Results No inflammatory marker was associated with any regional brain volume. Higher neopterin level was associated with lower total hippocampal, presubiculum, and cornu ammonis (CA) subfield volumes. Higher neopterin level and lower total hippocampal volume were independently associated with lower episodic memory, and neopterin level fully mediated the effect of hippocampal atrophy on episodic memory. Higher neopterin levels were associated with lower presubiculum, CA1, and CA4/dentate volumes and lower semantic memory, working memory, and global cognition. Conclusion Immune activation in response to HIV infection, measured by neopterin, has a deleterious and targeted effect on regional brain structure, which can be visualized with clinically available MRI measures of hippocampus and its subfields, and this effect is associated with lower cognitive function.
Collapse
Affiliation(s)
- Debra A Fleischman
- Rush Alzheimer's Disease Center (D.A.F., K.A., S.L., M.L., D.A.B., L.L.B.), Rush University Medical Center; the Department of Neurological Sciences (D.A.F., S.L., M.L., D.A.B., L.L.B.), the Department of Behavioral Sciences (D.A.F., M.L., L.L.B.), the Department of Preventive Medicine (S.L.), the Department of Diagnostic Radiology and Nuclear Medicine (K.A.), Rush University Medical Center; Ruth M. Rothstein CORE Center (O.M.A.); the Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago; the Blood Systems Research Institute (S.M.K.), San Francisco, CA; and the University of California at San Francisco (S.M.K.), Laboratory Medicine
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease Center (D.A.F., K.A., S.L., M.L., D.A.B., L.L.B.), Rush University Medical Center; the Department of Neurological Sciences (D.A.F., S.L., M.L., D.A.B., L.L.B.), the Department of Behavioral Sciences (D.A.F., M.L., L.L.B.), the Department of Preventive Medicine (S.L.), the Department of Diagnostic Radiology and Nuclear Medicine (K.A.), Rush University Medical Center; Ruth M. Rothstein CORE Center (O.M.A.); the Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago; the Blood Systems Research Institute (S.M.K.), San Francisco, CA; and the University of California at San Francisco (S.M.K.), Laboratory Medicine
| | - Sue Leurgans
- Rush Alzheimer's Disease Center (D.A.F., K.A., S.L., M.L., D.A.B., L.L.B.), Rush University Medical Center; the Department of Neurological Sciences (D.A.F., S.L., M.L., D.A.B., L.L.B.), the Department of Behavioral Sciences (D.A.F., M.L., L.L.B.), the Department of Preventive Medicine (S.L.), the Department of Diagnostic Radiology and Nuclear Medicine (K.A.), Rush University Medical Center; Ruth M. Rothstein CORE Center (O.M.A.); the Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago; the Blood Systems Research Institute (S.M.K.), San Francisco, CA; and the University of California at San Francisco (S.M.K.), Laboratory Medicine
| | - Sheila M Keating
- Rush Alzheimer's Disease Center (D.A.F., K.A., S.L., M.L., D.A.B., L.L.B.), Rush University Medical Center; the Department of Neurological Sciences (D.A.F., S.L., M.L., D.A.B., L.L.B.), the Department of Behavioral Sciences (D.A.F., M.L., L.L.B.), the Department of Preventive Medicine (S.L.), the Department of Diagnostic Radiology and Nuclear Medicine (K.A.), Rush University Medical Center; Ruth M. Rothstein CORE Center (O.M.A.); the Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago; the Blood Systems Research Institute (S.M.K.), San Francisco, CA; and the University of California at San Francisco (S.M.K.), Laboratory Medicine
| | - Melissa Lamar
- Rush Alzheimer's Disease Center (D.A.F., K.A., S.L., M.L., D.A.B., L.L.B.), Rush University Medical Center; the Department of Neurological Sciences (D.A.F., S.L., M.L., D.A.B., L.L.B.), the Department of Behavioral Sciences (D.A.F., M.L., L.L.B.), the Department of Preventive Medicine (S.L.), the Department of Diagnostic Radiology and Nuclear Medicine (K.A.), Rush University Medical Center; Ruth M. Rothstein CORE Center (O.M.A.); the Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago; the Blood Systems Research Institute (S.M.K.), San Francisco, CA; and the University of California at San Francisco (S.M.K.), Laboratory Medicine
| | - David A Bennett
- Rush Alzheimer's Disease Center (D.A.F., K.A., S.L., M.L., D.A.B., L.L.B.), Rush University Medical Center; the Department of Neurological Sciences (D.A.F., S.L., M.L., D.A.B., L.L.B.), the Department of Behavioral Sciences (D.A.F., M.L., L.L.B.), the Department of Preventive Medicine (S.L.), the Department of Diagnostic Radiology and Nuclear Medicine (K.A.), Rush University Medical Center; Ruth M. Rothstein CORE Center (O.M.A.); the Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago; the Blood Systems Research Institute (S.M.K.), San Francisco, CA; and the University of California at San Francisco (S.M.K.), Laboratory Medicine
| | - Oluwatoyin M Adeyemi
- Rush Alzheimer's Disease Center (D.A.F., K.A., S.L., M.L., D.A.B., L.L.B.), Rush University Medical Center; the Department of Neurological Sciences (D.A.F., S.L., M.L., D.A.B., L.L.B.), the Department of Behavioral Sciences (D.A.F., M.L., L.L.B.), the Department of Preventive Medicine (S.L.), the Department of Diagnostic Radiology and Nuclear Medicine (K.A.), Rush University Medical Center; Ruth M. Rothstein CORE Center (O.M.A.); the Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago; the Blood Systems Research Institute (S.M.K.), San Francisco, CA; and the University of California at San Francisco (S.M.K.), Laboratory Medicine
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center (D.A.F., K.A., S.L., M.L., D.A.B., L.L.B.), Rush University Medical Center; the Department of Neurological Sciences (D.A.F., S.L., M.L., D.A.B., L.L.B.), the Department of Behavioral Sciences (D.A.F., M.L., L.L.B.), the Department of Preventive Medicine (S.L.), the Department of Diagnostic Radiology and Nuclear Medicine (K.A.), Rush University Medical Center; Ruth M. Rothstein CORE Center (O.M.A.); the Biomedical Engineering (K.A.), Illinois Institute of Technology, Chicago; the Blood Systems Research Institute (S.M.K.), San Francisco, CA; and the University of California at San Francisco (S.M.K.), Laboratory Medicine
| |
Collapse
|
113
|
Syngeneic Transplantation of Olfactory Ectomesenchymal Stem Cells Restores Learning and Memory Abilities in a Rat Model of Global Cerebral Ischemia. Stem Cells Int 2018; 2018:2683969. [PMID: 29861741 PMCID: PMC5971302 DOI: 10.1155/2018/2683969] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/16/2018] [Accepted: 03/25/2018] [Indexed: 01/01/2023] Open
Abstract
Stem cells are considered as promising tools to repair diverse tissue injuries. Among the different stem cell types, the “olfactory ectomesenchymal stem cells” (OE-MSCs) located in the adult olfactory mucosa stand as one of the best candidates. Here, we evaluated if OE-MSC grafts could decrease memory impairments due to ischemic injury. OE-MSCs were collected from syngeneic F344 rats. After a two-step global cerebral ischemia, inducing hippocampal lesions, learning abilities were evaluated using an olfactory associative discrimination task. Cells were grafted into the hippocampus 5 weeks after injury and animal's learning abilities reassessed. Rats were then sacrificed and the brains collected for immunohistochemical analyses. We observed significant impairments in learning and memory abilities following ischemia. However, 4 weeks after OE-MSC grafts, animals displayed learning and memory performances similar to those of controls, while sham rats did not improve them. Immunohistochemical analyses revealed that grafts promoted neuroblast and glial cell proliferation, which could permit to restore cognitive functions. These results demonstrated, for the first time, that syngeneic transplantations of OE-MSCs in rats can restore cognitive abilities impaired after brain injuries and provide support for the development of clinical studies based on grafts of OE-MSCs in amnesic patients following brain injuries.
Collapse
|
114
|
Hypermetabolism in the hippocampal formation of cognitively impaired patients indicates detrimental maladaptation. Neurobiol Aging 2018; 65:41-50. [DOI: 10.1016/j.neurobiolaging.2018.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/27/2017] [Accepted: 01/07/2018] [Indexed: 11/22/2022]
|
115
|
Tecuatl C, Herrrera-López G, Martín-Ávila A, Yin B, Weber S, Barrionuevo G, Galván EJ. TrkB-mediated activation of the phosphatidylinositol-3-kinase/Akt cascade reduces the damage inflicted by oxygen-glucose deprivation in area CA3 of the rat hippocampus. Eur J Neurosci 2018; 47:1096-1109. [PMID: 29480936 PMCID: PMC5938095 DOI: 10.1111/ejn.13880] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 12/31/2022]
Abstract
The selective vulnerability of hippocampal area CA1 to ischemia-induced injury is a well-known phenomenon. However, the cellular mechanisms that confer resistance to area CA3 against ischemic damage remain elusive. Here, we show that oxygen-glucose deprivation-reperfusion (OGD-RP), an in vitro model that mimic the pathological conditions of the ischemic stroke, increases the phosphorylation level of tropomyosin receptor kinase B (TrkB) in area CA3. Slices preincubated with brain-derived neurotrophic factor (BDNF) or 7,8-dihydroxyflavone (7,8-DHF) exhibited reduced depression of the electrical activity triggered by OGD-RP. Consistently, blockade of TrkB suppressed the resistance of area CA3 to OGD-RP. The protective effect of TrkB activation was limited to area CA3, as OGD-RP caused permanent suppression of CA1 responses. At the cellular level, TrkB activation leads to phosphorylation of the accessory proteins SHC and Gab as well as the serine/threonine kinase Akt, members of the phosphoinositide 3-kinase/Akt (PI-3-K/Akt) pathway, a cascade involved in cell survival. Hence, acute slices pretreated with the Akt antagonist MK2206 in combination with BDNF lost the capability to resist the damage inflicted with OGD-RP. Consistently, with these results, CA3 pyramidal cells exhibited reduced propidium iodide uptake and caspase-3 activity in slices pretreated with BDNF and exposed to OGD-RP. We propose that PI-3-K/Akt downstream activation mediated by TrkB represents an endogenous mechanism responsible for the resistance of area CA3 to ischemic damage.
Collapse
Affiliation(s)
- Carolina Tecuatl
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Gabriel Herrrera-López
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Alejandro Martín-Ávila
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Bocheng Yin
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen Weber
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Germán Barrionuevo
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Emilio J. Galván
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| |
Collapse
|
116
|
Ou Y, Weber SG. Higher Aminopeptidase Activity Determined by Electroosmotic Push-Pull Perfusion Contributes to Selective Vulnerability of the Hippocampal CA1 Region to Oxygen Glucose Deprivation. ACS Chem Neurosci 2018; 9:535-544. [PMID: 29078045 DOI: 10.1021/acschemneuro.7b00326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
It has been known for over a century that the hippocampus, the center for learning and memory in the brain, is selectively vulnerable to ischemic damage, with the CA1 being more vulnerable than the CA3. It is also known that leucine enkephalin, or YGGFL, is neuroprotective. We hypothesized that the extracellular hydrolysis of YGGFL may be greater in the CA1 than the CA3, which would lead to the observed difference in susceptibility to ischemia. In rat organotypic hippocampal slice cultures, we estimated the Michaelis constant and the maximum velocity for membrane-bound aminopeptidase activity in the CA1 and CA3 regions. Using electroosmotic push-pull perfusion and offline capillary liquid chromatography, we inferred enzyme activity based on the production rate of GGFL, a natural and inactive product of the enzymatic hydrolysis of YGGFL. We found nearly 3-fold higher aminopeptidase activity in the CA1 than the CA3. The aminopeptidase inhibitor bestatin significantly reduced hydrolysis of YGGFL in both regions by increasing apparent Km. Based on propidium iodide cell death measurements 24 h after oxygen-glucose deprivation, we demonstrate that inhibition of aminopeptidase activity using bestatin selectively protected CA1 against delayed cell death due to oxygen-glucose deprivation and that this neuroprotection occurs through enkephalin-dependent pathways.
Collapse
Affiliation(s)
- Yangguang Ou
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen G. Weber
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
117
|
Rehni AK, Shukla V, Perez-Pinzon MA, Dave KR. Acidosis mediates recurrent hypoglycemia-induced increase in ischemic brain injury in treated diabetic rats. Neuropharmacology 2018; 135:192-201. [PMID: 29551689 DOI: 10.1016/j.neuropharm.2018.03.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Cerebral ischemia is a serious possible manifestation of diabetic vascular disease. Recurrent hypoglycemia (RH) enhances ischemic brain injury in insulin-treated diabetic (ITD) rats. In the present study, we determined the role of ischemic acidosis in enhanced ischemic brain damage in RH-exposed ITD rats. METHODS Diabetic rats were treated with insulin and mild/moderate RH was induced for 5 days. Three sets of experiments were performed. The first set evaluated the effects of RH exposure on global cerebral ischemia-induced acidosis in ITD rats. The second set evaluated the effect of an alkalizing agent (Tris-(hydroxymethyl)-aminomethane: THAM) on ischemic acidosis-induced brain injury in RH-exposed ITD rats. The third experiment evaluated the effect of the glucose transporter (GLUT) inhibitor on ischemic acidosis-induced brain injury in RH-exposed ITD rats. Hippocampal pH and lactate were measured during ischemia and early reperfusion for all three experiments. Neuronal survival in Cornu Ammonis 1 (CA1) hippocampus served as a measure of ischemic brain injury. FINDINGS Prior RH exposure increases lactate concentration and decreases pH during ischemia and early reperfusion when compared to controls. THAM and GLUT inhibitor treatments attenuated RH-induced increase in ischemic acidosis. GLUT inhibitor treatment reduced the RH-induced increase in lactate levels. Both THAM and GLUT inhibitor treatments significantly decreased ischemic damage in RH-exposed ITD rats. CONCLUSIONS Ischemia causes increased acidosis in RH-exposed ITD rats via a GLUT-sensitive mechanism. Exploring downstream pathways may help understand mechanisms by which prior exposure to RH increases cerebral ischemic damage.
Collapse
Affiliation(s)
- Ashish K Rehni
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Vibha Shukla
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
118
|
Lee SW, Yoo JH, Kim KW, Kim D, Park H, Choi J, Teicher MH, Jeong B. Hippocampal Subfields Volume Reduction in High Schoolers with Previous Verbal Abuse Experiences. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2018; 16:46-56. [PMID: 29397666 PMCID: PMC5810448 DOI: 10.9758/cpn.2018.16.1.46] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 01/31/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
Objective Reduced hippocampal volume and alterations in white matter tracts have been frequently reported in adults having the history of emotional maltreatment. We investigated whether these structural change occur in adolescents with previous verbal abuse (VA) experiences. Methods Hippocampal subfield volume and white matter structural connectivity measures were assessed in 31 first year male high school students with various degrees of exposure to parental and peer VA. Results The high VA group showed significant volume reduction in the left cornu ammonis (CA) 1 and left subiculum compared to the low VA group (p<0.05). Volumes of left hippocampal subfields CA1 and subiculum were negatively correlated with previous VA experiences (p<0.05). Increased mean diffusivity (MD) of the splenium of the corpus callosum was related to high VA score across all subjects (p<0.05). There was an inverse relationship between volume of the CA1 and subiculum and MD of the splenium (p<0.05). Conclusion Exposure to parental and peer VA may affect development of the left hippocampal subfields and the splenium of corpus callosum. These structural alterations can be discernible during adolescence.
Collapse
Affiliation(s)
- Sang Won Lee
- Computational Affective Neuroscience and Development Laboratory, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,Department of Psychiatry, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jae Hyun Yoo
- Computational Affective Neuroscience and Development Laboratory, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Ko Woon Kim
- Computational Affective Neuroscience and Development Laboratory, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Dongchan Kim
- Department of Electrical Engineering, KAIST, Daejeon, Korea
| | - HyunWook Park
- Department of Electrical Engineering, KAIST, Daejeon, Korea
| | - Jeewook Choi
- Department of Psychiatry, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - Martin H Teicher
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.,Developmental Biopsychiatry Research Program, McLean Hospital, Belmont, MA, USA
| | - Bumseok Jeong
- Computational Affective Neuroscience and Development Laboratory, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,KAIST Institute for Health Science and Technology, KAIST, Daejeon, Korea
| |
Collapse
|
119
|
Long-term effects of brief hypoxia due to cardiac arrest: Hippocampal reductions and memory deficits. Resuscitation 2018; 126:65-71. [PMID: 29474878 DOI: 10.1016/j.resuscitation.2018.02.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/01/2018] [Accepted: 02/15/2018] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To examine the effects of brief hypoxia (<7 min) due to cardiac arrest on the integrity of the brain and performance on memory and executive functions tasks. METHODS Patients after out-of-hospital cardiac arrest (CA) (n = 9), who were deemed neurologically intact on discharge, were compared to matched patients with myocardial infarction (MI) (n = 9). A battery of clinical and experimental memory and executive functions neuropsychological tests were administered and MRI scans for all patients were collected. Measures of subcortical and cortical volumes and cortical thickness were obtained using FreeSurfer. Manual segmentations of the hippocampus were also performed. APACHE-II scores were calculated based on metrics collected at admission to ICCU for all patients. RESULTS Significant differences between the two groups were observed on several verbal memory tests. Both hippocampi were significantly reduced (p < 0.05) in the CA patients, relative to MI patients. Hippocampal subfields segmentation showed significantly reduced presubiculum volumes bilaterally. CA patients had on average 10% reduction in volumes bilaterally across hippocampal subfields. No cortical thickness differences survived correction. Significant correlations were observed in the CA group only between the hippocampal volumes and performance on verbal memory tasks, including recollection. Hippocampal volumes and several memory measures (but not other cognitive domains) were strongly correlated with APACHE-II scores on admission in the CA group, but not in the MI group CONCLUSIONS: Chronic patients with cardiac arrest who were discharged from hospital in "good neurological condition" showed an average of 10% reduction in hippocampal volume bilaterally and significant verbal memory deficits relative to matched controls with myocardial infarction, suggesting even brief hypoxic periods suffice to lead to specific hippocampal damage.
Collapse
|
120
|
Treit S, Steve T, Gross DW, Beaulieu C. High resolution in-vivo diffusion imaging of the human hippocampus. Neuroimage 2018; 182:479-487. [PMID: 29395905 DOI: 10.1016/j.neuroimage.2018.01.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/09/2018] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
The human hippocampus is a key target of many imaging studies given its capacity for neurogenesis, role in long term potentiation and memory, and nearly ubiquitous involvement in neurological and psychiatric conditions. Diffusion tensor imaging (DTI) has detected microstructural abnormalities of the human hippocampus associated with various disorders, but acquisitions have typically been limited to low spatial resolution protocols designed for whole brain (e.g. > 2 mm isotropic, >8 mm3 voxels), limiting regional specificity and worsening partial volume effects. The purpose here was to develop a simple DTI protocol using readily available standard single-shot EPI at 3T, capable of yielding much higher spatial resolution images (1 x 1 x 1 mm3) of the human hippocampus in a 'clinically feasible' scan time of ~6 min. A thin slab of twenty 1 mm slices oriented along the long axis of the hippocampus enabled efficient coverage and a shorter repetition time, allowing more diffusion weighted images (DWIs) per slice per unit time. In combination with this strategy, a low b value of 500 s/mm2 was chosen to help overcome the very low SNR of a 1 x 1 x 1 mm3 EPI acquisition. 1 mm isotropic mean DWIs (averaged over 120-128 DWIs) showed excellent detail of the hippocampal architecture (e.g. morphology and digitations, sub-regions, stratum lacunosum moleculare - SLM) that was not readily visible on 2 mm isotropic diffusion images. Diffusion parameters within the hippocampus were consistent across subjects and fairly homogenous across sub-regions of the hippocampus (with the exception of the SLM and tail). However, it is expected that DTI parameters will be sensitive to microstructural changes associated with a number of clinical disorders (e.g. epilepsy, dementia) and that this practical, translatable approach for high resolution acquisition will facilitate localized detection of hippocampal pathology.
Collapse
Affiliation(s)
- Sarah Treit
- Department of Biomedical Engineering, Faculty of Medicine & Dentistry, University of Alberta, Canada
| | - Trevor Steve
- Division of Neurology, Faculty of Medicine & Dentistry, University of Alberta, Canada
| | - Donald W Gross
- Division of Neurology, Faculty of Medicine & Dentistry, University of Alberta, Canada
| | - Christian Beaulieu
- Department of Biomedical Engineering, Faculty of Medicine & Dentistry, University of Alberta, Canada.
| |
Collapse
|
121
|
Hefter D, Marti HH, Gass P, Inta D. Perinatal Hypoxia and Ischemia in Animal Models of Schizophrenia. Front Psychiatry 2018; 9:106. [PMID: 29651259 PMCID: PMC5884869 DOI: 10.3389/fpsyt.2018.00106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/16/2018] [Indexed: 12/12/2022] Open
Abstract
Intrauterine or perinatal complications constitute a major risk for psychiatric diseases. Infants who suffered from hypoxia-ischemia (HI) are at twofold risk to develop schizophrenia in later life. Several animal models attempt to reproduce these complications to study the yet unknown steps between an insult in early life and outbreak of the disease decades later. However, it is very challenging to find the right type and severity of insult leading to a disease-like phenotype in the animal, but not causing necrosis and focal neurological deficits. By contrast, too mild, repetitive insults may even be protective via conditioning effects. Thus, it is not surprising that animal models of hypoxia lead to mixed results. To achieve clinically translatable findings, better protocols are urgently needed. Therefore, we compare widely used models of hypoxia and HI and propose future directions for the field.
Collapse
Affiliation(s)
- Dimitri Hefter
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany.,RG Neuro- and Sensory Physiology, Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Hugo H Marti
- RG Neurovascular Research, Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Peter Gass
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany
| | - Dragos Inta
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Heidelberg, Germany.,Department of Psychiatry, University of Basel, Basel, Switzerland
| |
Collapse
|
122
|
Recording Field Potentials and Synaptic Plasticity From Freely Behaving Rodents. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2018. [DOI: 10.1016/b978-0-12-812028-6.00001-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
123
|
Happ DF, Tasker RA. Effects of α7 Nicotinic Receptor Activation on Cell Survival in Rat Organotypic Hippocampal Slice Cultures. Neurotox Res 2017; 33:887-895. [DOI: 10.1007/s12640-017-9854-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/13/2017] [Indexed: 11/28/2022]
|
124
|
Hassanzadeh P, Atyabi F, Dinarvand R, Dehpour AR, Azhdarzadeh M, Dinarvand M. Application of nanostructured lipid carriers: the prolonged protective effects for sesamol in in vitro and in vivo models of ischemic stroke via activation of PI3K signalling pathway. Daru 2017; 25:25. [PMID: 29262855 PMCID: PMC5738862 DOI: 10.1186/s40199-017-0191-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/01/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Treatment of the ischemic stroke has remained a major healthcare challenge. The phenolic compound, sesamol, has shown promising antioxidant and neuroprotective effects, however, fast clearance may negatively affect its efficiency. This, prompted us to incorporate sesamol into the nanostructured lipid carriers (S-NLCs) and evaluate its therapeutic potential in in vitro and in vivo models of ischemic stroke. METHODS S-NLCs formulations were prepared by high-pressure homogenization followed by physicochemical characterization, evaluation of the bioactivity of the optimal formulation in oxygen-glucose deprivation (OGD) and global cerebral ischemia/reperfusion (I/R) injury and implication of phosphatidylinositol 3-kinase (PI3K) pathway in this regard. Two- or three-way ANOVA, Mann-Whitney U test, and Student's t-test were used for data analysis. RESULTS Formation of S-NLCs which exhibited a controlled release profile, was confirmed by scanning electron microscope and differential scanning calorimetry. 1- and 8-h OGD followed by 24 h re-oxygenation significantly reduced PC12 cell viability, increased lactate dehydrogenase activity and the number of condensed nuclei, and induced oxidative stress as revealed by increased malondialdehyde level and decreased glutathione content and superoxide dismutase and catalase activities. Sesamol (80 and 100 μM) reduced the cytotoxicity, oxidative stress, and cellular damage only after 1-h OGD, while, S-NLCs (containing 80 and 100 μM of sesamol) were effective at both time points. Intravenous injections of S-NLCs (20 and 25 mg/kg) into rats markedly attenuated I/R-induced neurobehavioural deficits, cellular damage, and oxidative stress, while, free sesamol failed. Pre-treatment with PI3K inhibitor, LY294002, abolished the protective effects against OGD or I/R. CONCLUSIONS S-NLCs improve the pharmacological profile of sesamol and provide longer lasting protective effects for this phenolic phytochemical. This nanoformulation by activating PI3K pathway may serve as a promising candidate for neuroprotection against the cerebral stroke or other neurodegenerative disorders. Sesamol-loaded NLCs, a promising nanoformulation against the ischemic stroke.
Collapse
Affiliation(s)
- Parichehr Hassanzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad-Reza Dehpour
- Department of Pharmacology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Azhdarzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Meshkat Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
125
|
Pastuszko P, Schears GJ, Kubin J, Wilson DF, Pastuszko A. Granulocyte colony-stimulating factor significantly decreases density of hippocampal caspase 3-positive nuclei, thus ameliorating apoptosis-mediated damage, in a model of ischaemic neonatal brain injury. Interact Cardiovasc Thorac Surg 2017; 25:600-605. [PMID: 28962511 DOI: 10.1093/icvts/ivx047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 01/06/2017] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Ischaemic brain injury is a major complication in patients undergoing surgery for congenital heart disease, with the hippocampus being a particularly vulnerable region. We hypothesized that neuronal injury resulting from cardiopulmonary bypass and associated circulatory arrest is ameliorated by pretreatment with granulocyte colony-stimulating factor (G-CSF), a cytokine and an anti-apoptotic neurotrophic factor. METHODS In a model of ischaemic brain injury, 4 male newborn piglets were anaesthetized and subjected to deep hypothermic circulatory arrest (DHCA) (cooled to 18°C, DHCA maintained for 60 min, rewarmed and recovered for 8-9 h), while 4 animals received G-CSF (34 µg/kg, intravenously) 2 h prior to the DHCA procedure. At the end of each experiment, the animals were perfused with a fixative, the hippocampus was extracted, cryoprotected, cut and the brain sections were immunoprocessed for activated caspase 3, a pro-apoptotic factor. Immunopositive neuronal nuclei were counted in multiple counting boxes (440 × 330 µm) centred on the CA1 or CA3 hippocampal regions and their mean numbers compared between the different treatment groups and regions. RESULTS G-CSF pretreatment resulted in significantly lower counts of caspase 3-positive nuclei per counting box in both the CA1 [52.2 ± 9.3 (SD) vs 61.6 ± 8.4, P < 0.001] and CA3 (41.2 ± 6.9 vs 60.4 ± 16.4, P < 0.00002) regions of the hippocampus as compared to DHCA groups. The effects of G-CSF were significant for pyramidal cells of both regions and for interneurons in the CA3 region. CONCLUSIONS In an animal model of ischaemic brain injury, G-CSF reduces neuronal injury in the hippocampus, thus potentially having beneficial effect on neurologic outcomes.
Collapse
Affiliation(s)
- Peter Pastuszko
- Section of Cardiac Surgery, Children's Mercy Hospital and Clinics, The Ward Family Heart Center, Kansas City, MO, USA
| | | | - Joanna Kubin
- Department of Biochemistry and Biophysics, University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
| | - David F Wilson
- Department of Biochemistry and Biophysics, University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
| | - Anna Pastuszko
- Department of Biochemistry and Biophysics, University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
126
|
Hassanzadeh P, Arbabi E, Atyabi F, Dinarvand R. Ferulic acid-loaded nanostructured lipid carriers: A promising nanoformulation against the ischemic neural injuries. Life Sci 2017; 193:64-76. [PMID: 29196052 DOI: 10.1016/j.lfs.2017.11.046] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 11/07/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023]
Abstract
AIMS Treatment of the ischemic stroke has remained a major healthcare challenge. The phenolic compound, ferulic acid (FA), has shown promising antioxidant and neuroprotective effects, however, low bioavailability may negatively affect its efficiency. This, prompted us to incorporate FA into the nanostructured lipid carriers (FA-NLCs) and evaluate its therapeutic potential in in vitro and in vivo models of ischemic stroke. MAIN METHODS FA-NLCs were prepared by high-pressure homogenization followed by physicochemical characterization, evaluation of the bioactivity of FA-NLCs in oxygen-glucose deprivation (OGD) and global cerebral ischemia/reperfusion (I/R) injury and implication of phosphatidylinositol 3-kinase (PI3K) pathway in this regard. KEY FINDINGS Formation of FA-NLCs which exhibited a controlled release profile, was confirmed by scanning electron microscope and differential scanning calorimetry. 1- and 8-h OGD followed by 24h re-oxygenation significantly reduced PC12 cell viability, increased lactate dehydrogenase activity and number of condensed nuclei, and induced oxidative stress as revealed by increased malondialdehyde and decreased glutathione content and superoxide dismutase and catalase activities. FA (80 and 100μM) reduced the cytotoxicity, oxidative stress, and cellular damage only after 1-h OGD, while, FA-NLCs (containing 80 and 100μM of FA) were effective at both time points. Intravenous injections of FA-NLCs (20 and 25mg/kg) into rats significantly attenuated I/R-induced neurobehavioural deficits, cellular damage, and oxidative stress, while, FA failed. Pre-treatment with PI3K inhibitor, LY294002, abolished the protective effects against OGD or I/R. SIGNIFICANCE FA-NLCs by improving the pharmacological profile of FA and activating PI3K pathway might be of therapeutic value in cerebral stroke.
Collapse
Affiliation(s)
- Parichehr Hassanzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Elham Arbabi
- Research Center for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
127
|
Smith KR, Rajgor D, Hanley JG. Differential regulation of the Rac1 GTPase-activating protein (GAP) BCR during oxygen/glucose deprivation in hippocampal and cortical neurons. J Biol Chem 2017; 292:20173-20183. [PMID: 29046349 PMCID: PMC5724004 DOI: 10.1074/jbc.m117.796292] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/28/2017] [Indexed: 12/22/2022] Open
Abstract
Brain ischemia causes oxygen and glucose deprivation (OGD) in neurons, triggering a cascade of events leading to synaptic accumulation of glutamate. Excessive activation of glutamate receptors causes excitotoxicity and delayed cell death in vulnerable neurons. Following global cerebral ischemia, hippocampal CA1 pyramidal neurons are more vulnerable to injury than their cortical counterparts, but the mechanisms that underlie this difference are unclear. Signaling via Rho-family small GTPases, their upstream guanine nucleotide exchange factors, and GTPase-activating proteins (GAPs) is differentially dysregulated in response to OGD/ischemia in hippocampal and cortical neurons. Increased Rac1 activity caused by OGD/ischemia contributes to neuronal death in hippocampal neurons via diverse effects on NADPH oxidase activity and dendritic spine morphology. The Rac1 guanine nucleotide exchange factor Tiam1 mediates an OGD-induced increase in Rac1 activity in hippocampal neurons; however, the identity of an antagonistic GAP remains elusive. Here we show that the Rac1 GAP breakpoint cluster region (BCR) associates with NMDA receptors (NMDARs) along with Tiam1 and that this protein complex is more abundant in hippocampal compared with cortical neurons. Although total BCR is similar in the two neuronal types, BCR is more active in hippocampal compared with cortical neurons. OGD causes an NMDAR- and Ca2+-permeable AMPAR-dependent deactivation of BCR in hippocampal but not cortical neurons. BCR knockdown occludes OGD-induced Rac1 activation in hippocampal neurons. Furthermore, disrupting the Tiam1–NMDAR interaction with a fragment of Tiam1 blocks OGD-induced Tiam1 activation but has no effect on the deactivation of BCR. This work identifies BCR as a critical player in Rac1 regulation during OGD in hippocampal neurons.
Collapse
Affiliation(s)
- Katharine R Smith
- Centre for Synaptic Plasticity and School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom; Department of Pharmacology, University of Colorado Denver School of Medicine, Aurora, Colorado 80045.
| | - Dipen Rajgor
- Centre for Synaptic Plasticity and School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Jonathan G Hanley
- Centre for Synaptic Plasticity and School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
128
|
Fujimura M, Usuki F. In situ different antioxidative systems contribute to the site-specific methylmercury neurotoxicity in mice. Toxicology 2017; 392:55-63. [PMID: 29030019 DOI: 10.1016/j.tox.2017.10.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/28/2017] [Accepted: 10/06/2017] [Indexed: 01/17/2023]
Abstract
Methylmercury (MeHg), an environmental toxicant, induces site-specific neurotoxicity in adult human and animal models. In this study, we demonstrated that MeHg-induced neuropathological changes of the brain in mice were remarkable in the cerebrocortical neurons of deeper layers (dl-CCNs), but not in the CCNs of shallow layers (sl-CCNs) and the hippocampal neurons of cornu ammonis 1 (CA1-HNs). Total mercury concentration was not corresponded to the pathological changes. Here, we investigated the cause of such site-specific MeHg neurotoxicity with a focus on in situ antioxidative systems due to its critical role in MeHg intoxication. We performed in situ analyses of antioxidative enzymes expression using RT-qPCR analyses from laser microdissected sl-CCNs, dl-CCNs, and CA1-HNs samples, and immunohistochemistry. The results of antioxidative enzymes expression analyses demonstrated the lowest basal expression levels of mRNA and proteins, especially manganese superoxide dismutase (Mn-SOD) and glutathione peroxidase 1 (GPx1) in dl-CCNs. In addition, the Mn-SOD expression showed a lowest response to MeHg in dl-CCNs. We also performed enzymatic activity analyses for antioxidative enzymes using separated cerebral cortex and hippocampus. The results of enzymatic activity analyses indicate that the expression levels of antioxidative enzymes reflect their enzymatic activities. Immunostaining of thymidine glycerol, a sensitive oxidative stress marker, showed selectively increased expression in dl-CCNs after the exposure to MeHg but not in sl-CCNs and CA1-HNs, suggesting the occurrence of MeHg-induced oxidative stress in dl-CCNs. The differences in MeHg-induced occurrence of oxidative stress and pathological changes in sl-CCNs, dl-CCNs, and CA1-HNs corresponded to the basal level of Mn-SOD and GPx1 expression and the different protective response of Mn-SOD expression to MeHg. These findings suggest that the in situ different antioxidative systems play a role in the site-specific neurotoxicity of MeHg.
Collapse
Affiliation(s)
- Masatake Fujimura
- Department of Basic Medical Sciences, National Institute for Minamata Disease, Kumamoto, Japan.
| | - Fusako Usuki
- Department of Clinical Medicine, National Institute for Minamata Disease, Kumamoto, Japan
| |
Collapse
|
129
|
Hadem IKH, Majaw T, Kharbuli B, Sharma R. Beneficial effects of dietary restriction in aging brain. J Chem Neuroanat 2017; 95:123-133. [PMID: 29031555 DOI: 10.1016/j.jchemneu.2017.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/14/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022]
Abstract
Aging is a multifactorial complex process that leads to the deterioration of biological functions wherein its underlying mechanism is not fully elucidated. It affects the organism at the molecular and cellular level that contributes to the deterioration of structural integrity of the organs. The central nervous system is the most vulnerable organ affected by aging and its effect is highly heterogeneous. Aging causes alteration in the structure, metabolism and physiology of the brain leading to impaired cognitive and motor-neural functions. Dietary restriction (DR), a robust mechanism that extends lifespan in various organisms, ameliorates brain aging by reducing oxidative stress, improving mitochondrial function, activating anti-inflammatory responses, promoting neurogenesis and increasing synaptic plasticity. It also protects and prevents age-related structural changes. DR alleviates many age-associated diseases including neurodegeneration and improves cognitive functions. DR inhibits/activates nutrient signaling cascades such as insulin/IGF-1, mTOR, AMPK and sirtuins. Because of its sensitivity to energy status and hormones, AMPK is considered as the global nutrient sensor. This review will present an elucidative potential role of dietary restriction in the prevention of phenotypic features during aging in brain and its diverse mechanisms.
Collapse
Affiliation(s)
| | - Teikur Majaw
- Department of Biochemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India
| | - Babiangshisha Kharbuli
- Department of Biochemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India
| | - Ramesh Sharma
- Department of Biochemistry, North-Eastern Hill University, Shillong 793022, Meghalaya, India.
| |
Collapse
|
130
|
Endocytosis and lysosomal degradation of GluA2/3 AMPARs in response to oxygen/glucose deprivation in hippocampal but not cortical neurons. Sci Rep 2017; 7:12318. [PMID: 28951554 PMCID: PMC5614987 DOI: 10.1038/s41598-017-12534-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 09/06/2017] [Indexed: 02/04/2023] Open
Abstract
Global cerebral ischemia results in oxygen and glucose deprivation (OGD) and consequent delayed cell death of vulnerable neurons, with hippocampal CA1 neurons more vulnerable than cortical neurons. Most AMPA receptors (AMPARs) are heteromeric complexes of subunits GluA1/GluA2 or GluA2/GluA3, and the presence of GluA2 renders AMPARs Ca2+-impermeable. In hippocampal CA1 neurons, OGD causes the synaptic expression of GluA2-lacking Ca2+-permeable AMPARs, contributing to toxic Ca2+ influx. The loss of synaptic GluA2 is caused by rapid trafficking of GluA2-containing AMPARs from the cell surface, followed by a delayed reduction in GluA2 mRNA expression. We show here that OGD causes endocytosis, lysosomal targeting and consequent degradation of GluA2- and GluA3-containing AMPARs, and that PICK1 is required for both OGD-induced GluA2 endocytosis and lysosomal sorting. Our results further suggest that GluA1-containing AMPARs resist OGD-induced endocytosis. OGD does not cause GluA2 endocytosis in cortical neurons, and we show that PICK1 binding to the endocytic adaptor AP2 is enhanced by OGD in hippocampal, but not cortical neurons. We propose that endocytosis of GluA2/3, caused by a hippocampal-specific increase in PICK1-AP2 interactions, followed by PICK1-dependent lysosomal targeting, are critical events in determining changes in AMPAR subunit composition in the response to ischaemia.
Collapse
|
131
|
Amyloid Precursor Protein Protects Neuronal Network Function after Hypoxia via Control of Voltage-Gated Calcium Channels. J Neurosci 2017; 36:8356-71. [PMID: 27511009 DOI: 10.1523/jneurosci.4130-15.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/19/2016] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Acute cerebral ischemia and chronic neurovascular diseases share various common mechanisms with neurodegenerative diseases, such as disturbed cellular calcium and energy homeostasis and accumulation of toxic metabolites. A link between these conditions may be constituted by amyloid precursor protein (APP), which plays a pivotal role in the pathogenesis of Alzheimer's disease, but has also been associated with the response to acute hypoxia and regulation of calcium homeostasis. We therefore studied hypoxia-induced loss of function and recovery upon reoxygenation in hippocampal slices of mice lacking APP (APP(-/-)) or selectively expressing its soluble extracellular domain (APPsα-KI). Transient hypoxia disrupted electrical activity at the network and cellular level. In mice lacking APP, these impairments were significantly more severe, showing increased rise of intracellular calcium, faster loss of function, and higher incidence of spreading depression. Likewise, functional recovery upon reoxygenation was much slower and less complete than in controls. Most of these deficits were rescued by selective expression of the soluble extracellular fragment APPsα, or by pharmacological block of L-type calcium channels. We conclude that APP supports neuronal resistance toward acute hypoxia. This effect is mediated by the secreted APPsα-domain and involves L-type calcium channels. SIGNIFICANCE STATEMENT Amyloid precursor protein (APP) is involved in the pathophysiology of Alzheimer's disease, but its normal function in the brain remains elusive. Here, we describe a neuroprotective role of the protein in acute hypoxia. Functional recovery of mouse hippocampal networks after transient reduction of oxygen supply was strongly impaired in animals lacking APP. Most protective effects are mediated by the soluble extracellular fragment APPsα and involve L-type calcium channels. Thus, APP contributes to calcium homeostasis in situations of metabolic stress. This finding may shed light on the physiological function of APP and may be important for understanding mechanisms of neurodegenerative diseases.
Collapse
|
132
|
Lee JC, Kim YH, Lee TK, Kim IH, Cho JH, Cho GS, Shin BN, Park JH, Ahn JH, Shin MC, Cho JH, Kang IJ, Won MH, Seo JY. Effects of ischemic preconditioning on PDGF-BB expression in the gerbil hippocampal CA1 region following transient cerebral ischemia. Mol Med Rep 2017. [PMID: 28627606 PMCID: PMC5562056 DOI: 10.3892/mmr.2017.6799] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Ischemic preconditioning (IPC) is induced by exposure to brief durations of transient ischemia, which results in ischemic tolerance to a subsequent longer or lethal period of ischemia. In the present study, the effects of IPC (2 min of transient cerebral ischemia) were examined on immunoreactivity of platelet‑derived growth factor (PDGF)‑BB and on neuroprotection in the gerbil hippocampal CA1 region following lethal transient cerebral ischemia (LTCI; 5 min of transient cerebral ischemia). IPC was subjected to a 2‑min sublethal ischemia and a LTCI was given 5‑min transient ischemia. The animals in all of the groups were given recovery times of 1, 2 and 5 days and change in PDGF‑BB immunoreactivity was examined as was the neuronal damage/death in the hippocampus induced by LTCI. LTCI induced a significant loss of pyramidal neurons in the hippocampal CA1 region 5 days after LTCI, and significantly decreased PDGF‑BB immunoreactivity in the CA1 pyramidal neurons from day 1 after LTCI. Conversely, IPC effectively protected the CA1 pyramidal neurons from LTCI and increased PDGF‑BB immunoreactivity in the CA1 pyramidal neurons post‑LTCI. In conclusion, the results demonstrated that LTCI significantly altered PDGF‑BB immunoreactivity in pyramidal neurons in the hippocampal CA1 region, whereas IPC increased the immunoreactivity. These findings indicated that PDGF‑BB may be associated with IPC‑mediated neuroprotection.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yang Hee Kim
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Geum-Sil Cho
- Pharmacology and Toxicology Department, Shinpoong Pharmaceutical Co., Ltd., Ansan, Gyeonggi 15610, Republic of Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jeong Yeol Seo
- Department of Emergency Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| |
Collapse
|
133
|
Lana D, Ugolini F, Melani A, Nosi D, Pedata F, Giovannini MG. The neuron-astrocyte-microglia triad in CA3 after chronic cerebral hypoperfusion in the rat: Protective effect of dipyridamole. Exp Gerontol 2017; 96:46-62. [PMID: 28606482 DOI: 10.1016/j.exger.2017.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 12/01/2022]
Abstract
We investigated the quantitative and morphofunctional alterations of neuron-astrocyte-microglia triads in CA3 hippocampus, in comparison to CA1, after 2 Vessel Occlusion (2VO) and the protective effect of dipyridamole. We evaluated 3 experimental groups: sham-operated rats (sham, n=15), 2VO-operated rats treated with vehicle (2VO-vehicle, n=15), and 2VO-operated rats treated with dipyridamole from day 0 to day 7 (2VO-dipyridamole, n=15), 90days after 2VO. We analyzed Stratum Pyramidalis (SP), Stratum Lucidum (SL) and Stratum Radiatum (SR) of CA3. 1) ectopic neurons increased in SL and SR of 2VO-vehicle, and 2VO-dipyridamole rats; 2) apoptotic neurons increased in SP of 2VO-vehicle rats and dipyridamole reverted this effect; 3) astrocytes increased in SP, SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 4) TNF-α expression increased in astrocytes, blocked by dipyridamole, and in dendrites in SR of 2VO-vehicle rats; 5) total microglia increased in SL and SR of 2VO-vehicle and 2VO-dipyridamole rats; 6) triads increased in SR of 2VO-vehicle rats and dipyridamole reverted this effect. Microglia cooperated with astrocytes to phagocytosis of apoptotic neurons and debris, and engulfed ectopic non-fragmented neurons in SL of 2VO-vehicle and 2VO-dipyridamole rats, through a new mechanism called phagoptosis. CA3 showed a better adaptive capacity than CA1 to the ischemic insult, possibly due to the different behaviour of astrocytes and microglial cells. Dipyridamole had neuroprotective effects.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Filippo Ugolini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Alessia Melani
- Department of NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla, 50139 Firenze, Italy.
| | - Felicita Pedata
- Department of NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
134
|
A Comparison of Neuroimaging Abnormalities in Multiple Sclerosis, Major Depression and Chronic Fatigue Syndrome (Myalgic Encephalomyelitis): is There a Common Cause? Mol Neurobiol 2017; 55:3592-3609. [PMID: 28516431 PMCID: PMC5842501 DOI: 10.1007/s12035-017-0598-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/03/2017] [Indexed: 01/23/2023]
Abstract
There is copious evidence of abnormalities in resting-state functional network connectivity states, grey and white matter pathology and impaired cerebral perfusion in patients afforded a diagnosis of multiple sclerosis, major depression or chronic fatigue syndrome (CFS) (myalgic encephalomyelitis). Systemic inflammation may well be a major element explaining such findings. Inter-patient and inter-illness variations in neuroimaging findings may arise at least in part from regional genetic, epigenetic and environmental variations in the functions of microglia and astrocytes. Regional differences in neuronal resistance to oxidative and inflammatory insults and in the performance of antioxidant defences in the central nervous system may also play a role. Importantly, replicated experimental findings suggest that the use of high-resolution SPECT imaging may have the capacity to differentiate patients afforded a diagnosis of CFS from those with a diagnosis of depression. Further research involving this form of neuroimaging appears warranted in an attempt to overcome the problem of aetiologically heterogeneous cohorts which probably explain conflicting findings produced by investigative teams active in this field. However, the ionising radiation and relative lack of sensitivity involved probably preclude its use as a routine diagnostic tool.
Collapse
|
135
|
Sadelli K, Stamegna JC, Girard SD, Baril N, Escoffier G, Brus M, Véron AD, Khrestchatisky M, Roman FS. Global cerebral ischemia in rats leads to amnesia due to selective neuronal death followed by astroglial scar formation in the CA1 layer. Neurobiol Learn Mem 2017; 141:168-178. [PMID: 28438578 DOI: 10.1016/j.nlm.2017.04.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/12/2017] [Indexed: 12/23/2022]
Abstract
Global Cerebral Ischemia (GCI) occurs following cardiac arrest or neonatal asphyxia and leads to harmful neurological consequences. In most cases, patients who survive cardiac arrest develop severe cognitive and motor impairments. This study focused on learning and memory deficits associated with brain neuroanatomical reorganization that appears after GCI. The four-vessel occlusion (4VO) model was performed to produce a transient GCI. Hippocampal lesions in ischemic rats were visualized using anatomical Magnetic Resonance Imaging (aMRI). Then, the learning and memory abilities of control and ischemic (bilaterally or unilaterally) rats were assessed through the olfactory associated learning task. Finally, a "longitudinal" histological study was carried out to highlight the cellular reorganizations occurring after GCI. We demonstrated that the imaging, behavioral and histological results are closely related. In fact, aMRI revealed the appearance of hyper-intense signals in the dorsal hippocampus at day 3 post-GCI. Consequently, we showed a rise in cell proliferation (Ki 67+ cells) and endogenous neurogenesis especially in the dentate gyrus (DG) at day 3 post-GCI. Then, hyper-intense signals in the dorsal hippocampus were confirmed by strong neuronal losses in the CA1 layer at day 7 post-GCI. These results were linked with severe learning and memory impairments only in bilaterally ischemic rats at day 14 post-GCI. This amnesia was accompanied by huge astroglial and microglial hyperactivity at day 30 post-GCI. Finally, Nestin+ cells and astrocytes gave rise to astroglial scars, which persisted 60days post-GCI. In the light of these results, the 4VO model appears a reliable method to produce amnesia in order to study and develop new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Nathalie Baril
- Aix Marseille Univ, Fédération de recherche 3C, FR 3512, Marseille, France
| | | | - Maïna Brus
- Aix Marseille Univ, CNRS, NICN, Marseille, France
| | - Antoine D Véron
- Aix Marseille Univ, CNRS, NICN, Marseille, France; IRSEA, Research Institute in Semiochemistry and Applied Ethology, 84400 Apt, France
| | | | | |
Collapse
|
136
|
Neuroprotective Role of Exogenous Brain-Derived Neurotrophic Factor in Hypoxia-Hypoglycemia-Induced Hippocampal Neuron Injury via Regulating Trkb/MiR134 Signaling. J Mol Neurosci 2017; 62:35-42. [PMID: 28343294 DOI: 10.1007/s12031-017-0907-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 02/27/2017] [Indexed: 01/19/2023]
Abstract
Hypoxic-ischemic brain injury is an important cause of neonatal mortality and morbidity. Brain-derived neurotrophic factor (BDNF) has been reported to play a neuroprotective role in hypoxic-ischemic brain injury; however, the specific effects and mechanism of BDNF on hypoxic-hypoglycemic hippocampal neuron injury remains unknown. The current study investigated the action of BDNF in regulating cerebral hypoxic-ischemic injury by simulating hippocampal neuron ischemia and hypoxia. We found that BDNF, p-Trkb, and miR-134 expression levels decreased, and that exogenous BDNF increased survival and reduced apoptosis in hypoxic-hypoglycemic hippocampal neurons. The results also show that BDNF inhibits MiR-134 expression by activating the TrkB pathway. Transfection with TrkB siRNA and pre-miR-134 abrogated the neuroprotective role of BDNF in hypoxic-hypoglycemic hippocampal neurons. Our results suggest that exogenous BDNF alleviates hypoxic-ischemic brain injury through the Trkb/MiR-134 pathway. These findings may help to identify a potential therapeutic agent for the treatment of hypoxic-ischemic brain injury.
Collapse
|
137
|
Kuyumcu G, Byrne RW, Dawe RJ, Kocak M. Incomplete Circle of Willis: A risk factor for mesial temporal sclerosis? Epilepsy Res 2017; 132:29-33. [PMID: 28284050 DOI: 10.1016/j.eplepsyres.2017.02.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/15/2017] [Accepted: 02/27/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVE The aim of this study was to investigate whether there may be a correlation between the anatomical variants of Circle of Willis (CoW) and presence/laterality of mesial temporal sclerosis (MTS). METHODS We retrospectively identified the CoW variants on Wada angiograms in 71 patients with pathologically proven MTS. Angiograms were interpreted by two radiologists independently and blinded to clinical data. We divided the anterior and posterior components of the CoW into functionally complete and functionally incomplete groups. We then sought its correlation with the presence and laterality of MTS. RESULTS No statistically significant relationship was found between the functional status of the anterior circulation and the laterality of the MTS (p=0.657). Relationship of the posterior incomplete circle to MTS was statistically significant on both sides (p=0.023 for the left, p=0.04 for the right), with an effect size moderate to large for the left side and moderate for the right side. Although the fetal variant appeared to be related to the ipsilateral MTS, it did not reach to a level of statistical significance (p=0.15). SIGNIFICANCE The study demonstrates a statistically significant association of the incomplete posterior circulation of the CoW to the presence of ipsilateral MTS. Further studies in larger patient populations may be needed to seek whether an incomplete circulation may facilitate development of MTS, especially affecting the watershed zones.
Collapse
Affiliation(s)
- Gokhan Kuyumcu
- Rush University Medical Center, Department of Radiology & Nuclear Medicine, 1725 West Harrison Street, Suite 437, Chicago, IL 60612, USA.
| | - Richard W Byrne
- Rush University Medical Center, Department of Neurosurgery, 1725 West Harrison Street, Suite 855, Chicago, IL 60612, USA.
| | - Robert J Dawe
- Rush University Medical Center, Department of Radiology & Nuclear Medicine, 1725 West Harrison Street, Suite 437, Chicago, IL 60612, USA; Rush Alzheimer's Disease Center, Johnston R Bowman Health Center, 600 S. Paulina Street, Chicago, IL, 60612, USA.
| | - Mehmet Kocak
- Rush University Medical Center, Department of Radiology & Nuclear Medicine, 1725 West Harrison Street, Suite 437, Chicago, IL 60612, USA.
| |
Collapse
|
138
|
Lee JC, Cho JH, Lee TK, Kim IH, Won MH, Cho GS, Shin BN, Hwang IK, Park JH, Ahn JH, Kang IJ, Lee YJ, Kim YH. Effect of hyperthermia on calbindin-D 28k immunoreactivity in the hippocampal formation following transient global cerebral ischemia in gerbils. Neural Regen Res 2017; 12:1458-1464. [PMID: 29089991 PMCID: PMC5649466 DOI: 10.4103/1673-5374.215256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Calbindin D-28K (CB), a Ca2+-binding protein, maintains Ca2+ homeostasis and protects neurons against various insults. Hyperthermia can exacerbate brain damage produced by ischemic insults. However, little is reported about the role of CB in the brain under hyperthermic condition during ischemic insults. We investigated the effects of transient global cerebral ischemia on CB immunoreactivity as well as neuronal damage in the hippocampal formation under hyperthermic condition using immunohistochemistry for neuronal nuclei (NeuN) and CB, and Fluoro-Jade B histofluorescence staining in gerbils. Hyperthermia (39.5 ± 0.2°C) was induced for 30 minutes before and during transient ischemia. Hyperthermic ischemia resulted in neuronal damage/death in the pyramidal layer of CA1–3 area and in the polymorphic layer of the dentate gyrus at 1, 2, 5 days after ischemia. In addition, hyperthermic ischemia significantly decreaced CB immunoreactivity in damaged or dying neurons at 1, 2, 5 days after ischemia. In brief, hyperthermic condition produced more extensive and severer neuronal damage/death, and reduced CB immunoreactivity in the hippocampus following transient global cerebral ischemia. Present findings indicate that the degree of reduced CB immunoreactivity might be related with various neuronal damage/death overtime and corresponding areas after ischemic insults.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jeong-Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Geum-Sil Cho
- Department of Pharmacology & Toxicology, Shinpoong Pharmaceutical Co., Ltd., Ansan, South Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, South Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, South Korea
| | - Young Joo Lee
- Department of Emergency Medicine, Seoul Hospital, College of Medicine, Sooncheonhyang University, Seoul, South Korea
| | - Yang Hee Kim
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
139
|
Sanganalmath SK, Gopal P, Parker JR, Downs RK, Parker JC, Dawn B. Global cerebral ischemia due to circulatory arrest: insights into cellular pathophysiology and diagnostic modalities. Mol Cell Biochem 2016; 426:111-127. [PMID: 27896594 DOI: 10.1007/s11010-016-2885-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/08/2016] [Indexed: 02/07/2023]
Abstract
Circulatory arrest (CA) remains a major unresolved public health problem in the United States; the annual incidence of which is ~0.50 to 0.55 per 1000 population. Despite seminal advances in therapeutic approaches over the past several decades, brain injury continues to be the leading cause of morbidity and mortality after CA. In brief, CA typically results in global cerebral ischemia leading to delayed neuronal death in the hippocampal pyramidal cells as well as in the cortical layers. The dynamic changes occurring in neurons after CA are still unclear, and predicting these neurological changes in the brain still remains a difficult issue. It is hypothesized that the "no-flow" period produces a cytotoxic cascade of membrane depolarization, Ca2+ ion influx, glutamate release, acidosis, and resultant activation of lipases, nucleases, and proteases. Furthermore, during reperfusion injury, neuronal death occurs due to the generation of free radicals by interfering with the mitochondrial respiratory chain. The efficacy of many pharmacological agents for CA patients has often been disappointing, reflecting our incomplete understanding of this enigmatic disease. The primary obstacles to the development of a neuroprotective therapy in CA include uncertainties with regard to the precise cause(s) of neuronal dysfunction and what to target. In this review, we summarize our knowledge of the pathophysiology as well as specific cellular changes in brain after CA and revisit the most important neurofunctional, neuroimaging techniques, and serum biomarkers as potent predictors of neurologic outcome in CA patients.
Collapse
Affiliation(s)
- Santosh K Sanganalmath
- Division of Cardiovascular Diseases, Department of Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - Purva Gopal
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - John R Parker
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, KY, USA
| | - Richard K Downs
- Division of Neuroradiology, Department of Radiology, University of Louisville, Louisville, KY, USA
| | - Joseph C Parker
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, KY, USA
| | - Buddhadeb Dawn
- Division of Cardiovascular Diseases, Department of Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| |
Collapse
|
140
|
Karunasinghe RN, Grey AC, Telang R, Vlajkovic SM, Lipski J. Differential spread of anoxic depolarization contributes to the pattern of neuronal injury after oxygen and glucose deprivation (OGD) in the Substantia Nigra in rat brain slices. Neuroscience 2016; 340:359-372. [PMID: 27826106 DOI: 10.1016/j.neuroscience.2016.10.067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/14/2016] [Accepted: 10/29/2016] [Indexed: 12/21/2022]
Abstract
Anoxic depolarization (AD) is an acute event evoked by brain ischemia, involving a profound loss of cell membrane potential and swelling that spreads over susceptible parts of the gray matter. Its occurrence is a strong predictor of the severity of neuronal injury. Little is known about this event in the Substantia Nigra, a midbrain nucleus critical for motor control. We tested the effects of oxygen and glucose deprivation (OGD), an in vitro model of brain ischemia, in rat midbrain slices. AD developed within 4min from OGD onset and spread in the Substantia Nigra pars reticulata (SNr), but not through the Substantia Nigra pars compacta (SNc). This differential effect involved a contrasting pattern of changes in membrane potential between dopamine-producing SNc and non-dopaminergic SNr neurons. A fast depolarization in SNr neurons was not followed by repolarization after the end of OGD, and was associated with swollen somata and beaded dendrites. In contrast, slowly developing depolarization of SNc neurons led to repolarization after OGD ended, and no changes in neuronal morphology were observed. The AD-resistance of the SNc involved smaller dysregulations of K+ and Ca2+ ions, and a slower loss of energy metabolites. Our results show that acute ischemia profoundly impairs the function and morphology of SNr neurons but not adjacent SNc neurons, and that the surprising higher tolerance of SNc neurons correlates with the resistance of the SNc region to AD. This differential response may affect the pattern of early neuronal injury that develops in the brainstem after acute ischemic insults.
Collapse
Affiliation(s)
- Rashika N Karunasinghe
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Angus C Grey
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Ravindra Telang
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Srdjan M Vlajkovic
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand
| | - Janusz Lipski
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Auckland 1023, New Zealand.
| |
Collapse
|
141
|
Ulbrich F, Kaufmann KB, Meske A, Lagrèze WA, Augustynik M, Buerkle H, Ramao CC, Biermann J, Goebel U. The CORM ALF-186 Mediates Anti-Apoptotic Signaling via an Activation of the p38 MAPK after Ischemia and Reperfusion Injury in Retinal Ganglion Cells. PLoS One 2016; 11:e0165182. [PMID: 27764224 PMCID: PMC5072679 DOI: 10.1371/journal.pone.0165182] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/08/2016] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Ischemia and reperfusion injury may induce apoptosis and lead to sustained tissue damage and loss of function, especially in neuronal organs. While carbon monoxide is known to exert protective effects after various harmful events, the mechanism of carbon monoxide releasing molecules in neuronal tissue has not been investigated yet. We hypothesize that the carbon monoxide releasing molecule (CORM) ALF-186, administered after neuronal ischemia-reperfusion injury (IRI), counteracts retinal apoptosis and its involved signaling pathways and consecutively reduces neuronal tissue damage. METHODS IRI was performed in rat´s retinae for 1 hour. The water-soluble CORM ALF-186 (10 mg/kg) was administered intravenously via a tail vein after reperfusion. After 24 and 48 hours, retinal tissue was harvested to analyze mRNA and protein expression of Bcl-2, Bax, Caspase-3, ERK1/2, p38 and JNK. Densities of fluorogold pre-labeled retinal ganglion cells (RGC) were analyzed 7 days after IRI. Immunohistochemistry was performed on retinal cross sections. RESULTS ALF-186 significantly reduced IRI mediated loss of RGC. ALF-186 treatment differentially affected mitogen-activated protein kinases (MAPK) phosphorylation: ALF-186 activated p38 and suppressed ERK1/2 phosphorylation, while JNK remained unchanged. Furthermore, ALF-186 treatment affected mitochondrial apoptosis, decreasing pro-apoptotic Bax and Caspase-3-cleavage, but increasing anti-apoptotic Bcl-2. Inhibition of p38-MAPK using SB203580 reduced ALF-186 mediated anti-apoptotic effects. CONCLUSION In this study, ALF-186 mediated substantial neuroprotection, affecting intracellular apoptotic signaling, mainly via MAPK p38. CORMs may thus represent a promising therapeutic alternative treating neuronal IRI.
Collapse
Affiliation(s)
- Felix Ulbrich
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kai B. Kaufmann
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Meske
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolf A. Lagrèze
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Augustynik
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hartmut Buerkle
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carlos C. Ramao
- Instituto de Tecnologia Química e Biológica-António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
- Alfama Ltd., Instituto de Biologia Experimental e Tecnológica, IBET, Oeiras, Portugal
| | - Julia Biermann
- Eye Center, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ulrich Goebel
- Department of Anesthesiology and Critical Care, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- * E-mail:
| |
Collapse
|
142
|
Dzieciol AM, Bachevalier J, Saleem KS, Gadian DG, Saunders R, Chong WKK, Banks T, Mishkin M, Vargha-Khadem F. Hippocampal and diencephalic pathology in developmental amnesia. Cortex 2016; 86:33-44. [PMID: 27880886 PMCID: PMC5264402 DOI: 10.1016/j.cortex.2016.09.016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/10/2016] [Accepted: 09/18/2016] [Indexed: 01/01/2023]
Abstract
Developmental amnesia (DA) is a selective episodic memory disorder associated with hypoxia-induced bilateral hippocampal atrophy of early onset. Despite the systemic impact of hypoxia-ischaemia, the resulting brain damage was previously reported to be largely limited to the hippocampus. However, the thalamus and the mammillary bodies are parts of the hippocampal-diencephalic network and are therefore also at risk of injury following hypoxic-ischaemic events. Here, we report a neuroimaging investigation of diencephalic damage in a group of 18 patients with DA (age range 11-35 years), and an equal number of controls. Importantly, we uncovered a marked degree of atrophy in the mammillary bodies in two thirds of our patients. In addition, as a group, patients had mildly reduced thalamic volumes. The size of the anterior-mid thalamic (AMT) segment was correlated with patients' visual memory performance. Thus, in addition to the hippocampus, the diencephalic structures also appear to play a role in the patients' memory deficit.
Collapse
Affiliation(s)
- Anna M Dzieciol
- University College London Great Ormond Street Institute of Child Health, London, UK.
| | | | | | - David G Gadian
- University College London Great Ormond Street Institute of Child Health, London, UK
| | | | - W K Kling Chong
- Department of Radiology, Great Ormond Street Hospital for Children, London, UK
| | - Tina Banks
- Department of Radiology, Great Ormond Street Hospital for Children, London, UK
| | | | | |
Collapse
|
143
|
Park SM, Park CW, Lee TK, Cho JH, Park JH, Lee JC, Chen BH, Shin BN, Ahn JH, Tae HJ, Shin MC, Ohk TG, Cho JH, Won MH, Choi SY, Kim IH. Effect of ischemic preconditioning on antioxidant status in the gerbil hippocampal CA1 region after transient forebrain ischemia. Neural Regen Res 2016; 11:1081-9. [PMID: 27630689 PMCID: PMC4994448 DOI: 10.4103/1673-5374.187039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Ischemic preconditioning (IPC) is a condition of sublethal transient global ischemia and exhibits neuroprotective effects against subsequent lethal ischemic insult. We, in this study, examined the neuroprotective effects of IPC and its effects on immunoreactive changes of antioxidant enzymes including superoxide dismutase (SOD) 1 and SOD2, catalase (CAT) and glutathione peroxidase (GPX) in the gerbil hippocampal CA1 region after transient forebrain ischemia. Pyramidal neurons of the stratum pyramidale (SP) in the hippocampal CA1 region of animals died 5 days after lethal transient ischemia without IPC (8.6% (ratio of remanent neurons) of the sham-operated group); however, IPC prevented the pyramidal neurons from subsequent lethal ischemic injury (92.3% (ratio of remanent neurons) of the sham-operated group). SOD1, SOD2, CAT and GPX immunoreactivities in the sham-operated animals were easily detected in pyramidal neurons in the stratum pyramidale (SP) of the hippocampal CA1 region, while all of these immunoreactivities were rarely detected in the stratum pyramidale at 5 days after lethal transient ischemia without IPC. Meanwhile, their immunoreactivities in the sham-operated animals with IPC were similar to (SOD1, SOD2 and CAT) or higher (GPX) than those in the sham-operated animals without IPC. Furthermore, their immunoreactivities in the stratum pyramidale of the ischemia-operated animals with IPC were steadily maintained after lethal ischemia/reperfusion. Results of western blot analysis for SOD1, SOD2, CAT and GPX were similar to immunohistochemical data. In conclusion, IPC maintained or increased the expression of antioxidant enzymes in the stratum pyramidale of the hippocampal CA1 region after subsequent lethal transient forebrain ischemia and IPC exhibited neuroprotective effects in the hippocampal CA1 region against transient forebrain ischemia.
Collapse
Affiliation(s)
- Seung Min Park
- Department of Emergency Medicine, Sacred Heart Hospital, College of Medicine, Hallym University, Anyang, South Korea; Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Chan Woo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Bai Hui Chen
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Bich-Na Shin
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Hyun-Jin Tae
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Taek Geun Ohk
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Soo Young Choi
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
144
|
Ahmed ME, Dong Y, Lu Y, Tucker D, Wang R, Zhang Q. Beneficial Effects of a CaMKIIα Inhibitor TatCN21 Peptide in Global Cerebral Ischemia. J Mol Neurosci 2016; 61:42-51. [PMID: 27604243 DOI: 10.1007/s12031-016-0830-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/30/2016] [Indexed: 12/16/2022]
Abstract
Aberrant calcium influx is a common feature following ischemic reperfusion (I/R) in transient global cerebral ischemia (GCI) and causes delayed neuronal cell death in the CA1 region of the hippocampus. Activation of calcium-calmodulin (CaM)-dependent protein kinase IIα (CaMKIIα) is a key event in calcium signaling in ischemic injury. The present study examined the effects of intracerebroventricular (icv) injection of tatCN21 in ischemic rats 3 h after GCI reperfusion. Cresyl violet and NeuN staining revealed that tatCN21 exerted neuroprotective effects against delayed neuronal cell death of hippocampal CA1 pyramidal neurons 10 days post-GCI. In addition, TatCN21 administration ameliorated GCI-induced spatial memory deficits in the Barnes maze task as well as anxiety-like behaviors and spontaneous motor activity in the elevated plus maze and open field test, respectively. Mechanistic studies showed that the administration of tatCN21 decreased GCI-induced phosphorylation, translocation, and membrane targeting of CaMKIIα. Treatment with tatCN21 also inhibited the level of CaMKIIα-NR2B interaction and NR2B phosphorylation. Our results revealed an important role of tatCN21 in inhibiting CaMKIIα activation and its beneficial effects in neuroprotection and memory preservation in an ischemic brain injury model.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yujiao Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Donovan Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Ruimin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
145
|
Zhang L, Chopp M, Zhang Y, Xiong Y, Li C, Sadry N, Rhaleb I, Lu M, Zhang ZG. Diabetes Mellitus Impairs Cognitive Function in Middle-Aged Rats and Neurological Recovery in Middle-Aged Rats After Stroke. Stroke 2016; 47:2112-8. [PMID: 27387991 DOI: 10.1161/strokeaha.115.012578] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 06/10/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Diabetes mellitus (DM) is a common metabolic disease among the middle-aged and older population, which leads to an increase of stroke incidence and poor stroke recovery. The present study was designed to investigate the impact of DM on brain damage and on ischemic brain repair after stroke in aging animals. METHODS DM was induced in middle-aged rats (13 months) by administration of nicotinamide and streptozotocin. Rats with confirmed hyperglycemia status 30 days after nicotinamide-streptozotocin injection and age-matched non-DM rats were subjected to embolic middle cerebral artery occlusion. RESULTS Middle-aged rats subjected to nicotinamide-streptozotocin injection became hyperglycemic and developed cognitive deficits 2 months after induction of DM. Histopathologic analysis revealed that there was sporadic vascular disruption, including cerebral microvascular thrombosis, blood-brain barrier leakage, and loss of paravascular aquaporin-4 in the hippocampi. Importantly, middle-aged DM rats subjected to stroke had exacerbated sensorimotor and cognitive deficits compared with age-matched non-DM ischemic rats during stroke recovery. Compared with age-matched non-DM ischemic rats, DM ischemic rats exhibited aggravated neurovascular disruption in the bilateral hippocampi and white matter, suppressed stroke-induced neurogenesis and oligodendrogenesis, and impaired dendritic/spine plasticity. However, DM did not enlarge infarct volume. CONCLUSIONS Our data suggest that DM exacerbates neurovascular damage and hinders brain repair processes, which likely contribute to the impairment of stroke recovery.
Collapse
Affiliation(s)
- Li Zhang
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.).
| | - Michael Chopp
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Yanlu Zhang
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Ye Xiong
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Chao Li
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Neema Sadry
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Imane Rhaleb
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Mei Lu
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Zheng Gang Zhang
- From the Department of Neurology (L.Z., M.C., C.L., N.S., I.R., Z.G.Z.), Department of Neurosurgery (Y.Z., Y.X.), and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| |
Collapse
|
146
|
Yi JH, Cho SY, Jeon SJ, Jung JW, Park MS, Kim DH, Ryu JH. Early immature neuronal death is partially involved in memory impairment induced by cerebral ischemia. Behav Brain Res 2016; 308:75-82. [DOI: 10.1016/j.bbr.2016.04.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 04/09/2016] [Accepted: 04/12/2016] [Indexed: 12/27/2022]
|
147
|
Morrison JH, Siegel SJ, Gazzaley AH, Huntley GW. ■ REVIEW : Glutamate Receptors: Emerging Links Between Subunit Proteins and Specific Excitatory Circuits in Primate Hippocampus and Neocortex. Neuroscientist 2016. [DOI: 10.1177/107385849600200513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Glutamate receptors (GluRs) are the primary mediators of excitatory neurotransmission in the CNS and play an indispensable role in brain function. Recent molecular advances have revealed an increasingly elaborate panel of GluR subunits that combine to form a variety of heteromeric GluR complexes with distinct functional characteristics determined by the stoichiometry of the subunit composition. Excitatory circuits in hippocampus and neocortex exhibit a complex and highly ordered array of termination patterns that reflect both segregation and convergence at specific target sites. We hypothesize that the molecular diversity of the GluRs will be manifested as circuit-specific profiles that will generate extensive functional diversity in cortical excitatory circuits. To elucidate the link between GluR diversity and neuroanatomical circuitry, immunocytochemical techniques employing subunit-specific antibodies have been used to localize various subunit proteins at the cellular and synaptic level. Such studies have revealed differential subunit parcellation between neocortical neuronal populations, as well as within defined dendritic compartments of hippocampal pyramidal cells. Additionally, the intradendritic parcellation of a specific GluR subunit is modifiable in an age-related and circuit-specific manner. NEUROSCIENTIST 2:272-283, 1996
Collapse
Affiliation(s)
- John H. Morrison
- Fishberg Research Center for Neurobiology, Department of Geriatrics and Adult Development, Mount
Sinai School of Medicine New York, New York
| | | | | | | |
Collapse
|
148
|
Tóth Z, Györimolnár I, Abrahám H, Hevesi A. Cannulation and Cardiopulmonary Bypass Produce Selective Brain Lesions in Pigs. Asian Cardiovasc Thorac Ann 2016; 14:273-8. [PMID: 16868098 DOI: 10.1177/021849230601400402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Whether cardiopulmonary bypass alone or together with manipulation of the aorta produces neurologic complications remains controversial. Using a pig model, the immediate effects of aortic cannulation and cardiopulmonary bypass on neural injury in different brain regions were investigated in 3 experimental groups: non-operated controls; operated controls with aortic cannulation without cardiopulmonary bypass; and operated animals undergoing cardiopulmonary bypass. Immunohistochemistry using a monoclonal antibody against calretinin was used to show possible ischemic damage in the hippocampal formation which is one of the most vulnerable regions to ischemia. Both cannulation of the aorta alone and cardiopulmonary bypass resulted in numerous argyrophilic neurons in discrete regions of the prefrontal and cerebellar cortex. Decreased calretinin immunoreaction and a reduced number of calretinin-positive neurons were observed following aortic cannulation or cardiopulmonary bypass compared to the non-operated controls. This suggests that both cannulation of the aorta alone and cardiopulmonary bypass affect a selected population of neurons. Therefore, off-pump, aorta no-touch technique may prevent neurologic complications.
Collapse
Affiliation(s)
- Zsolt Tóth
- Division of Cardiac Surgery, Heart Institute, Faculty of Medicine, University of Pécs, 7624 Pécs, Ifjuság út 13, Hungary.
| | | | | | | |
Collapse
|
149
|
Neuroprotective effects of syringic acid against OGD/R-induced injury in cultured hippocampal neuronal cells. Int J Mol Med 2016; 38:567-73. [PMID: 27278454 DOI: 10.3892/ijmm.2016.2623] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 05/13/2016] [Indexed: 11/05/2022] Open
Abstract
Cerebral ischemic injury and treatment are important topics in neurological science. In the present study, an in vitro model of cerebral ischemia was established by subjecting primary cultures of hippocampal neuronal cells to oxygen-glucose deprivation followed by reperfusion (OGD/R), in order to evaluate the possible neuroprotective role of syringic acid (SA). The results of 3-(4,5-dimethylthiazol‑2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) assays showed that pre-treatment with SA (0.1, 1, 10, and 20 µM) attenuated OGD/R-induced neuronal injury in a dose-dependent manner, with evidence of increased cell viability and decreased LDH leakage. In addition, oxidative stress markers were evaluated using commercial kits, and the results demonstrated that OGD/R exposure induced distinct oxidative stress, accompanied by elevated levels of intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) production, and reduced activity of the antioxidant enzyme superoxide dismutase (SOD), which were dose-dependently restored by pre-treatment with SA. In addition, the concentration of intracellular free calcium [Ca2+]i and mitochondrial membrane potential (MMP or Δψm) were determined in order to evaluate the degree of neuronal damage by performing flow cytometric analysis and observing the cells under a fluorescence microscope, respectively. We demonstrated that pre-treatment with SA inhibited elevations in [Ca2+]i, whereas it increased the MMP dose-dependently following exposure to OGD/R. Western blot analysis revealed that OGD/R promoted cell apoptosis with concomitant increases in Bax and caspase-3 expression, and reduced Bcl-2 expression, which was reversed by pre‑treatment with SA in a dose-dependent manner. Moreover, these effects were mediated through the JNK and p38 pathways, as pre‑treatment with SA inhibited the OGD/R-induced increase in phosphorylated (p-)JNK and p-p38 expression. Taken together, these results suggested that SA exerted strong neuroprotective effects in hippocampal neuronal cells, which may be attributed to the attenuation of OGD/R-induced cell injury through the JNK and p38 signaling pathways.
Collapse
|
150
|
Park JH, Cho JH, Kim IH, Ahn JH, Lee JC, Chen BH, Shin BN, Tae HJ, Yoo KY, Hong S, Kang IJ, Won MH, Kim JD. Oenanthe Javanica Extract Protects Against Experimentally Induced Ischemic Neuronal Damage via its Antioxidant Effects. Chin Med J (Engl) 2016; 128:2932-7. [PMID: 26521793 PMCID: PMC4756874 DOI: 10.4103/0366-6999.168063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Water dropwort (Oenanthe javanica) as a popular traditional medicine in Asia shows various biological properties including antioxidant activity. In this study, we firstly examined the neuroprotective effect of Oenanthe javanica extract (OJE) in the hippocampal cornus ammonis 1 region (CA1 region) of the gerbil subjected to transient cerebral ischemia. METHODS Gerbils were established by the occlusion of common carotid arteries for 5 min. The neuroprotective effect of OJE was estimated by cresyl violet staining. In addition, 4 antioxidants (copper, zinc superoxide dismutase [SOD], manganese SOD, catalase, and glutathione peroxidase) immunoreactivities were investigated by immunohistochemistry. RESULTS Pyramidal neurons in the CA1 region showed neuronal death at 5 days postischemia; at this point in time, all antioxidants immunoreactivities disappeared in CA1 pyramidal neurons and showed in many nonpyramidal cells. Treatment with 200 mg/kg, not 100 mg/kg, OJE protected CA1 pyramidal neurons from ischemic damage. In addition, 200 mg/kg OJE treatment increased or maintained antioxidants immunoreactivities. Especially, among the antioxidants, glutathione peroxidase immunoreactivity was effectively increased in the CA1 pyramidal neurons of the OJE-treated sham-operated and ischemia-operated groups. CONCLUSION Our present results indicate that treatment with OJE can protect neurons from transient ischemic damage and that the neuroprotective effect may be closely associated with increased or maintained intracellular antioxidant enzymes by OJE.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 200-701, South Korea
| | - Jong-Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon 200-701, South Korea
| |
Collapse
|