1
|
Wu S, Zhang YF, Gui Y, Jiang T, Zhou CM, Li JY, Suo JL, Li YN, Jin RL, Li SL, Cui JY, Tan BH, Li YC. A detection method for neuronal death indicates abnormalities in intracellular membranous components in neuronal cells that underwent delayed death. Prog Neurobiol 2023; 226:102461. [PMID: 37179048 DOI: 10.1016/j.pneurobio.2023.102461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/20/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023]
Abstract
Acute neuronal degeneration is always preceded under the light and electron microscopes by a stage called microvacuolation, which is characterized by a finely vacuolar alteration in the cytoplasm of the neurons destined to death. In this study, we reported a method for detecting neuronal death using two membrane-bound dyes, rhodamine R6 and DiOC6(3), which may be associated with the so-called microvacuolation. This new method produced a spatiotemporally similar staining pattern to Fluoro-Jade B in kainic acid-damaged brains in mice. Further experiments showed that increased staining of rhodamine R6 and DiOC6(3) was observed only in degenerated neurons, but not in glia, erythrocytes, or meninges. Different from Fluoro-Jade-related dyes, rhodamine R6 and DiOC6(3) staining is highly sensitive to solvent extraction and detergent exposure. Staining with Nile red for phospholipids and filipin III for non-esterified cholesterol supports that the increased staining of rhodamine R6 and DiOC6(3) might be associated with increased levels of phospholipids and free cholesterol in the perinuclear cytoplasm of damaged neurons. In addition to kainic acid-injected neuronal death, rhodamine R6 and DiOC6(3) were similarly useful for detecting neuronal death in ischemic models either in vivo or in vitro. As far as we know, the staining with rhodamine R6 or DiOC6(3) is one of a few histochemical methods for detecting neuronal death whose target molecules have been well defined and therefore may be useful for explaining experimental results as well as exploring the mechanisms of neuronal death. (250 words).
Collapse
Affiliation(s)
- Shuang Wu
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Yan-Feng Zhang
- Department of Pediatric Neurology, First Hospital of Jilin University, Changchun, Jilin Province 130021, PR China
| | - Yue Gui
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Tian Jiang
- Department of Emergency and Critical Care Medicine, The Second Hospital of Jilin University, Jilin Province 130041, PR China
| | - Cheng-Mei Zhou
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Jing-Yi Li
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Jia-Le Suo
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Yong-Nan Li
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, PR China
| | - Rui-Lin Jin
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Shu-Lei Li
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Jia-Yue Cui
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Bai-Hong Tan
- Laboratory Teaching Center of Basic Medicine, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China
| | - Yan-Chao Li
- Department of Histology and Embryology, College of Basic Medical Sciences, Norman Bethune Health Science Center of Jilin University, Jilin Province 130021, PR China.
| |
Collapse
|
2
|
Fujikawa DG. Programmed Mechanisms of Status Epilepticus-induced Neuronal Necrosis. Epilepsia Open 2022; 8 Suppl 1:S25-S34. [PMID: 35278284 PMCID: PMC10173844 DOI: 10.1002/epi4.12593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/05/2022] [Indexed: 11/11/2022] Open
Abstract
Excitotoxicity is the underlying mechanism for all acute neuronal injury, from cerebral ischemia, status epilepticus, traumatic CNS injury and hypoglycemia. It causes morphological neuronal necrosis, and it triggers a programmed cell death program. Excessive calcium entry through the NMDA-receptor-operated cation channel activates two key enzymes-calpain I and neuronal nitric oxide synthase (nNOS). Calpain I, a cytosolic enzyme, translocates to mitochondrial and lysosomal membranes, causing release of cytochrome c, endonuclease G and apoptosis-inducing factor (AIF) from mitochondria and DNase II and cathepsins B and D from lysosomes. These all translocate to neuronal nuclei, creating DNA damage, which activates poly(ADP) ribose polymerase-1 (PARP-1) to form excessive amounts of poly(ADP) ribose (PAR) polymers, which translocate to mitochondrial membranes, causing release of truncated AIF (tAIF). The free radicals that are released from mitochondria and peroxynitrite, formed from nitric oxide (NO) from nNOS catalysis of L-arginine to L-citrulline, damage mitochondrial and lysosomal membranes and DNA. The end result is the necrotic death of neurons. Another programmed necrotic pathway, necroptosis, occurs through a parallel pathway. As investigators of necroptosis do not recognize the excitotoxic pathway, it is unclear to what extent each contributes to programmed neuronal necrosis. We are studying the extent to which each contributes to acute neuronal necrosis and the extent of cross-talk between these pathways.
Collapse
Affiliation(s)
- Denson G Fujikawa
- VA Greater Los Angeles Healthcare System, CA and Department of Neurology and Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
3
|
Demyanenko SV, Uzdensky A. LIM kinase inhibitor T56-LIMKi protects mouse brain from photothrombotic stroke. Brain Inj 2021; 35:490-500. [PMID: 33523710 DOI: 10.1080/02699052.2021.1879397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Primary Objective: In an ischemic stroke, the damage spreads from the infarction core to surrounding tissues. The present work was aimed at the search of effective neuroprotectors that restrict injury propagation. Research Design: We studied possible protective effects of inhibitors of protein kinases LIMK2 (T56-LIMKi), DYRK1A (harmine), and tryptophan hydroxylase (4-chlorophenylalanine) on infarction size and morphology of peri-infarct area after photothrombotic stroke (a model of ischemic stroke) in mouse brain. Methods and Procedures: Photothrombotic stroke was induced by laser irradiation of mouse cortex after administration of photosensitizer Bengal Rose, which does not penetrate cells and remains in blood vessels. Under light exposure, it induces vessel occlusion. Infarct volume and histological changes in the cerebral cortex were evaluated 3, 7 and 14 days after photothrombotic impact. Main Outcomes and Results: Harmine and 4-chlorophenylalanine did not influence infarct volume and morphology of peri-infarct area in the mouse brain cortex after photothrombotic stroke. However, LIMK2 inhibitor T56-LIMKi significantly reduced infarct volume 7 and 14 days after photothrombotic stroke. It also increased the percent of normochromic neurons and decreased the fraction of altered cortical cells (hypochromic, hyperchromic and pyknotic neurons). Conclusions: T56-LIMK2i may be considered as a promising anti-stroke agent.
Collapse
Affiliation(s)
- Svetlana V Demyanenko
- Laboratory of Molecular Neuroscience, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Anatoly Uzdensky
- Laboratory of Molecular Neuroscience, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| |
Collapse
|
4
|
Unal DB, Caliari SR, Lampe KJ. Engineering biomaterial microenvironments to promote myelination in the central nervous system. Brain Res Bull 2019; 152:159-174. [PMID: 31306690 DOI: 10.1016/j.brainresbull.2019.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 01/01/2023]
Abstract
Promoting remyelination and/or minimizing demyelination are key therapeutic strategies under investigation for diseases and injuries like multiple sclerosis (MS), spinal cord injury, stroke, and virus-induced encephalopathy. Myelination is essential for efficacious neuronal signaling. This myelination process is originated by oligodendrocyte progenitor cells (OPCs) in the central nervous system (CNS). Resident OPCs are capable of both proliferation and differentiation, and also migration to demyelinated injury sites. OPCs can then engage with these unmyelinated or demyelinated axons and differentiate into myelin-forming oligodendrocytes (OLs). However this process is frequently incomplete and often does not occur at all. Biomaterial strategies can now be used to guide OPC and OL development with the goal of regenerating healthy myelin sheaths in formerly damaged CNS tissue. Growth and neurotrophic factors delivered from such materials can promote proliferation of OPCs or differentiation into OLs. While cell transplantation techniques have been used to replace damaged cells in wound sites, they have also resulted in poor transplant cell viability, uncontrollable differentiation, and poor integration into the host. Biomaterial scaffolds made from extracellular matrix (ECM) mimics that are naturally or synthetically derived can improve transplanted cell survival, support both transplanted and endogenous cell populations, and direct their fate. In particular, stiffness and degradability of these scaffolds are two parameters that can influence the fate of OPCs and OLs. The future outlook for biomaterials research includes 3D in vitro models of myelination / remyelination / demyelination to better mimic and study these processes. These models should provide simple relationships of myelination to microenvironmental biophysical and biochemical properties to inform improved therapeutic approaches.
Collapse
Affiliation(s)
- Deniz B Unal
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Steven R Caliari
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, United States.
| |
Collapse
|
5
|
Chen X, Nakada S, Donahue JE, Chen RH, Tucker R, Qiu J, Lim YP, Stopa EG, Stonestreet BS. Neuroprotective effects of inter-alpha inhibitor proteins after hypoxic-ischemic brain injury in neonatal rats. Exp Neurol 2019; 317:244-259. [PMID: 30914159 DOI: 10.1016/j.expneurol.2019.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/08/2019] [Accepted: 03/22/2019] [Indexed: 11/30/2022]
Abstract
Hypoxic-ischemic (HI) brain injury is one of the most common neurological problems occurring in the perinatal period. Hypothermia is the only approved intervention for neonatal HI encephalopathy. However, this treatment is only partially protective, has a narrow therapeutic time window after birth and only can be used to treat full-term infants. Consequently, additional therapies are critically needed. Inflammation is an important contributing factor to the evolution of HI brain injury in neonates. Inter-alpha Inhibitor Proteins (IAIPs) are immunomodulatory proteins with anti-inflammatory properties. We have previously shown that IAIPs reduce neuronal cell death and improve behavioral outcomes when given after carotid artery ligation, but before hypoxia in male neonatal rats. The objective of the current study was to investigate the neuroprotective effects of treatment with IAIPs given immediately or 6 h after HI in both male and female neonatal rats. HI was induced with the Rice-Vannucci method in postnatal (P) day 7 rats. After ligation of the right common carotid artery, P7 rats were exposed to 90 min of hypoxia (8% oxygen). Human plasma-derived IAIPs or placebo (phosphate buffered saline) was given at zero, 24, and 48 h after HI. Brains were perfused, weighed and fixed 72 h after HI at P10. In a second, delayed treatment group, the same procedure was followed except that IAIPs or placebo were given at 6, 24 and 48 h after HI. Separate sham-operated, placebo-treated groups were exposed to identical protocols but were not exposed to carotid artery ligation and remained in room air. Rat sex was recorded. The effects of IAIPs on HI brain injury were examined using histopathological scoring and immunohistochemical analyses of the brain and by using infarct volume measurements on frozen tissue of the entire brain hemispheres ipsilateral and contralateral to HI injury. IAIPs given immediately after HI improved (P < 0.050) histopathological brain injury across and within the cingulate, caudate/putamen, thalamus, hippocampus and parietal cortex in males, but not in females. In contrast, IAIPs given immediately after HI reduced (P < 0.050) infarct volumes of the hemispheres ipsilateral to HI injury in similarly both the males and females. Treatment with IAIPs also resulted in higher (P < 0.050) brain weights compared with the placebo-treated HI group, reduced (P < 0.050) neuronal and non-neuronal cell death in the cortex and total hemisphere, and also increased the total area of oligodendrocytes determined by CNPase in the ipsilateral hemisphere and corpus callosum (P < 0.050) of male, but not female subjects exposed to HI. Delayed treatment with IAIPs 6 h after HI did not improve histopathological brain injury in males or females, but resulted in higher (P < 0.050) brain weights compared with the placebo-treated HI males. Therefore, treatment with IAIPs immediately after HI improved brain weights and reduced neuropathological brain injury and cell death in male rats, and reduced infarct volume in both male and female neonatal rats. We conclude that IAIPs exert neuroprotective effects after exposure to HI in neonatal rats and may exhibit some sex-related differential effects.
Collapse
Affiliation(s)
- Xiaodi Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Warren Alpert Medical School of Brown University, USA
| | - Sakura Nakada
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Warren Alpert Medical School of Brown University, USA
| | - John E Donahue
- The Warren Alpert Medical School of Brown University, USA; Department of Pathology and Neurosurgery, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, USA
| | - Ray H Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Warren Alpert Medical School of Brown University, USA
| | - Richard Tucker
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA
| | - Joseph Qiu
- ProThera Biologics, Inc, Providence, RI, USA
| | - Yow-Pin Lim
- The Warren Alpert Medical School of Brown University, USA; ProThera Biologics, Inc, Providence, RI, USA; Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Edward G Stopa
- The Warren Alpert Medical School of Brown University, USA; Department of Pathology and Neurosurgery, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Warren Alpert Medical School of Brown University, USA.
| |
Collapse
|
6
|
Prineas JW, Lee S. Multiple Sclerosis: Destruction and Regeneration of Astrocytes in Acute Lesions. J Neuropathol Exp Neurol 2019; 78:140-156. [PMID: 30605525 PMCID: PMC6330170 DOI: 10.1093/jnen/nly121] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
There are reports that astrocyte perivascular endfeet are damaged in some cases of multiple sclerosis (MS). This study was designed to determine the origin and outcome of astrocyte damage in acute, resolving, and inactive plaques. Ten acute plaques from 10 early MS cases and 14 plaques of differing histological age from 9 subacute and chronic cases were examined immunohistochemically. Also examined were nonnecrotic early lesions in 3 patients with neuromyelitis optica (NMO). Plaques from 3 MS cases were examined electron microscopically. The edge zones in each of the 10 acute MS lesions revealed a complete loss of astrocyte cell bodies and their pericapillary, perineuronal, and perivascular foot processes. Dendrophagocytosis of degenerate astrocytes was observed. Astrocyte precursors, similar to those that replace destroyed astrocytes in nonnecrotic NMO lesions, were present in areas depleted of astrocytes. Resolving plaques were repopulated initially by stellate astrocytes that stained negatively for the water channel molecule aquaporin4 (AQP4). In older lesions, astrocytes were predominantly AQP4-positive. Loss and recovery of astrocytes in new MS lesions may be as important as myelin loss as a cause of conduction block responsible for symptoms in patients with relapsing and remitting and secondary progressive MS.
Collapse
Affiliation(s)
- John W Prineas
- Department of Medicine, The University of Sydney, Camperdown, NSW, Australia
| | - Sandra Lee
- Department of Medicine, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
7
|
Chen X, Hovanesian V, Naqvi S, Lim YP, Tucker R, Donahue JE, Stopa EG, Stonestreet BS. Systemic infusions of anti-interleukin-1β neutralizing antibodies reduce short-term brain injury after cerebral ischemia in the ovine fetus. Brain Behav Immun 2018; 67:24-35. [PMID: 28780000 PMCID: PMC5696097 DOI: 10.1016/j.bbi.2017.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/14/2017] [Accepted: 08/02/2017] [Indexed: 01/27/2023] Open
Abstract
Perinatal hypoxic-ischemic reperfusion (I/R)-related brain injury is a leading cause of neurologic morbidity and life-long disability in children. Infants exposed to I/R brain injury develop long-term cognitive and behavioral deficits, placing a large burden on parents and society. Therapeutic strategies are currently not available for infants with I/R brain damage, except for hypothermia, which can only be used in full term infants with hypoxic-ischemic encephalopathy (HIE). Moreover, hypothermia is only partially protective. Pro-inflammatory cytokines are key contributors to the pathogenesis of perinatal I/R brain injury. Interleukin-1β (IL-1β) is a critical pro-inflammatory cytokine, which has been shown to predict the severity of HIE in infants. We have previously shown that systemic infusions of mouse anti-ovine IL-1β monoclonal antibody (mAb) into fetal sheep resulted in anti-IL-1β mAb penetration into brain, reduced I/R-related increases in IL-1β expression and blood-brain barrier (BBB) dysfunction in fetal brain. The purpose of the current study was to examine the effects of systemic infusions of anti-IL-1β mAb on short-term I/R-related parenchymal brain injury in the fetus by examining: 1) histopathological changes, 2) apoptosis and caspase-3 activity, 3) neuronal degeneration 4) reactive gliosis and 5) myelin basic protein (MBP) immunohistochemical staining. The study groups included non-ischemic controls, placebo-treated ischemic, and anti-IL-1β mAb treated ischemic fetal sheep at 127days of gestation. The systemic intravenous infusions of anti-IL-1β mAb were administered at fifteen minutes and four hours after in utero brain ischemia. The duration of each infusion was two hours. Parenchymal brain injury was evaluated by determining pathological injury scores, ApopTag® positive cells/mm2, caspase-3 activity, Fluoro-Jade B positive cells/mm2, glial fibrillary acidic protein (GFAP) and MBP staining in the brains of fetal sheep 24h after 30min of ischemia. Treatment with anti-IL-1β mAb reduced (P<0.05) the global pathological injury scores, number of apoptotic positive cells/mm2, and caspase-3 activity after ischemia in fetal sheep. The regional pathological scores and Fluoro-Jade B positive cells/mm2 did not differ between the placebo- and anti-IL-1β mAb treated ischemic fetal sheep. The percent of the cortical area stained for GFAP was lower (P<0.05) in the placebo ischemic treated than in the non-ischemic group, but did not differ between the placebo- and anti-IL-1β mAb treated ischemic groups. MBP immunohistochemical expression did not differ among the groups. In conclusion, infusions of anti-IL-1β mAb attenuate short-term I/R-related histopathological tissue injury, apoptosis, and reduce I/R-related increases in caspase-3 activity in ovine fetal brain. Therefore, systemic infusions of anti-IL-1β mAb attenuate short-term I/R-related parenchymal brain injury in the fetus.
Collapse
Affiliation(s)
- Xiaodi Chen
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI
| | - Virginia Hovanesian
- Core Research Laboratories, the Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Syed Naqvi
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI
| | | | - Richard Tucker
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI
| | - John E. Donahue
- Department of Pathology and Neurosurgery, the Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Edward G. Stopa
- Department of Pathology and Neurosurgery, the Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI
| | - Barbara S. Stonestreet
- Department of Pediatrics, the Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI
| |
Collapse
|
8
|
Fujikawa DG. The role of excitotoxic programmed necrosis in acute brain injury. Comput Struct Biotechnol J 2015; 13:212-21. [PMID: 25893083 PMCID: PMC4398818 DOI: 10.1016/j.csbj.2015.03.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 03/19/2015] [Accepted: 03/21/2015] [Indexed: 12/20/2022] Open
Abstract
Excitotoxicity involves the excessive release of glutamate from presynaptic nerve terminals and from reversal of astrocytic glutamate uptake, when there is excessive neuronal depolarization. N-methyl-d-aspartate (NMDA) receptors, a subtype of glutamate receptor, are activated in postsynaptic neurons, opening their receptor-operated cation channels to allow Ca2 + influx. The Ca2 + influx activates two enzymes, calpain I and neuronal nitric oxide synthase (nNOS). Calpain I activation produces mitochondrial release of cytochrome c (cyt c), truncated apoptosis-inducing factor (tAIF) and endonuclease G (endoG), the lysosomal release of cathepsins B and D and DNase II, and inactivation of the plasma membrane Na+–Ca2 + exchanger, which add to the buildup of intracellular Ca2 +. tAIF is involved in large-scale DNA cleavage and cyt c may be involved in chromatin condensation; endoG produces internucleosomal DNA cleavage. The nuclear actions of the other proteins have not been determined. nNOS forms nitric oxide (NO), which reacts with superoxide (O2−) to form peroxynitrite (ONOO−). These free radicals damage cellular membranes, intracellular proteins and DNA. DNA damage activates poly(ADP-ribose) polymerase-1 (PARP-1), which produces poly(ADP-ribose) (PAR) polymers that exit nuclei and translocate to mitochondrial membranes, also releasing AIF. Poly(ADP-ribose) glycohydrolase hydrolyzes PAR polymers into ADP-ribose molecules, which translocate to plasma membranes, activating melastatin-like transient receptor potential 2 (TRPM-2) channels, which open, allowing Ca2 + influx into neurons. NADPH oxidase (NOX1) transfers electrons across cellular membranes, producing O2−. The result of these processes is neuronal necrosis, which is a programmed cell death that is the basis of all acute neuronal injury in the adult brain.
Collapse
|
9
|
Shimada JI, Taniguchi J, Mori M, Sato Y, Takuwa H, Ito H, Kuwabara S. Retinol palmitate prevents ischemia-induced cell changes in hippocampal neurons through the Notch1 signaling pathway in mice. Exp Neurol 2013; 247:182-7. [PMID: 23651513 DOI: 10.1016/j.expneurol.2013.04.014] [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: 12/12/2012] [Revised: 04/23/2013] [Accepted: 04/29/2013] [Indexed: 02/05/2023]
Abstract
Retinol palmitate, an analog of vitamin A, plays multiple roles in the nervous system, including neural differentiation, axon outgrowth, and neural patterning, and is also an antioxidative agent and thereby potential neuroprotectant for brain ischemia. The present study aimed at investigating the protective effects of retinol palmitate against ischemia-induced brain injury in a bilateral common carotid artery occlusion (BCCAO) model in mice. Ischemia induced by 20-min BCCAO resulted in significant neuronal morphological changes and reactive astrocyte proliferation in the hippocampus, particularly in the CA1 region, and these changes were accompanied by increased Notch1 expression. Intraperitoneal retinol palmitate administration before ischemia reduced ischemic neurons with Notch1 expression; the differences were statistically significant in both the 1.2mg/kg group and 12 mg/kg group. These results show that retinol palmitate prevents brain ischemia-induced neuronal injury with Notch1 expression and that Notch1 signaling could be involved in the neuroprotective mechanism. Retinol palmitate could be a treatment option for human brain infarction.
Collapse
Affiliation(s)
- Jun-Ichiro Shimada
- Department of Neurology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | | | | | | | | | | | | |
Collapse
|
10
|
Mathai S, Gunn AJ, Backhaus RA, Guan J. Window of opportunity for neuroprotection with an antioxidant, allene oxide synthase, after hypoxia-ischemia in adult male rats. CNS Neurosci Ther 2012; 18:887-94. [PMID: 22998294 PMCID: PMC6493396 DOI: 10.1111/cns.12004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 06/18/2012] [Accepted: 08/14/2012] [Indexed: 01/20/2023] Open
Abstract
AIMS Oxidative stress is an early event in the cascade leading in neuronal damage after hypoxic-ischemic (HI) brain injury. In the present study, we examined the dose response and window of opportunity for neuroprotection after HI injury with Allene Oxide Synthase (AOS), an anti-oxidative enzyme of the member of cytochrome P450 family. METHODS Adult male rats received intra-cerebro-ventricular infusions of either saline (vehicle) or AOS (1 μg or 10 μg or 100 μg per rat, intracerebroventricular n = 16 all groups) either 45 min or 3 h after unilateral HI brain injury. Brains were collected 5 days later. The extent of brain damage, neuronal survival, apoptosis, and glial reactions were assessed in the striatum, hippocampus, and cortex. RESULTS Allene Oxide Synthase was associated with reduced neuronal damage scores when given 45 min, but not 3 h, after HI injury (P < 0.0001) in all brain regions. AOS treatment (10 μg) improved neuronal survival in the striatum, cortex, and hippocampus (P < 0.05, P < 0.001) and reduced the microglia reaction (P < 0.05) and numbers of caspase-3-positive cells in the hippocampus (P < 0.01). CONCLUSIONS Early blockade of oxidative stress after HI injury reduces inflammatory response, neuronal necrosis, and apoptosis. The neuroprotective effects of AOS were time of administration-dependent suggesting a relatively restricted window of opportunity for acute brain injury.
Collapse
Affiliation(s)
- Sam Mathai
- The Liggins InstituteThe University of AucklandAucklandNew Zealand
- Department of PhysiologyFaculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | - Alistair J. Gunn
- Department of PhysiologyFaculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | | | - Jian Guan
- The Liggins InstituteThe University of AucklandAucklandNew Zealand
| |
Collapse
|
11
|
Samson AL, Knaupp AS, Sashindranath M, Borg RJ, Au AEL, Cops EJ, Saunders HM, Cody SH, McLean CA, Nowell CJ, Hughes VA, Bottomley SP, Medcalf RL. Nucleocytoplasmic coagulation: an injury-induced aggregation event that disulfide crosslinks proteins and facilitates their removal by plasmin. Cell Rep 2012; 2:889-901. [PMID: 23041318 DOI: 10.1016/j.celrep.2012.08.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/27/2012] [Accepted: 08/24/2012] [Indexed: 11/18/2022] Open
Abstract
Cellular injury causes a myriad of processes that affect proteostasis. We describe nucleocytoplasmic coagulation (NCC), an intracellular disulfide-dependent protein crosslinking event occurring upon late-stage cell death that orchestrates the proteolytic removal of misfolded proteins. In vitro and in vivo models of neuronal injury show that NCC involves conversion of soluble intracellular proteins, including tubulin, into insoluble oligomers. These oligomers, also seen in human brain tissue following neurotrauma, act as a cofactor and substrate for the plasminogen-activating system. In plasminogen(-/-) mice, levels of misfolded β-tubulin were elevated and its clearance delayed following neurotrauma, demonstrating a requirement for plasminogen in the removal of NCC constituents. While additional in vivo studies will further dissect this phenomenon, our study clearly shows that NCC, a process analogous to the formation of thrombi, generates an aggregated protein scaffold that limits release of cellular components and recruits clearance mechanisms to the site of injury.
Collapse
Affiliation(s)
- Andre L Samson
- Australian Centre for Blood Diseases, AMREP, Monash University, Melbourne, Victoria 3004, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Li J, Ma X, Yu W, Lou Z, Mu D, Wang Y, Shen B, Qi S. Reperfusion promotes mitochondrial dysfunction following focal cerebral ischemia in rats. PLoS One 2012; 7:e46498. [PMID: 23029539 PMCID: PMC3460895 DOI: 10.1371/journal.pone.0046498] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2012] [Accepted: 08/14/2012] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Mitochondrial dysfunction has been implicated in the cell death observed after cerebral ischemia, and several mechanisms for this dysfunction have been proposed. Reperfusion after transient cerebral ischemia may cause continued and even more severe damage to the brain. Many lines of evidence have shown that mitochondria suffer severe damage in response to ischemic injury. The purpose of this study was to observe the features of mitochondrial dysfunction in isolated mitochondria during the reperfusion period following focal cerebral ischemia. METHODS Male Wistar rats were subjected to focal cerebral ischemia. Mitochondria were isolated using Percoll density gradient centrifugation. The isolated mitochondria were fixed for electron microscopic examination; calcium-induced mitochondrial swelling was quantified using spectrophotometry. Cyclophilin D was detected by Western blotting. Fluorescent probes were used to selectively stain mitochondria to measure their membrane potential and to measure reactive oxidative species production using flow cytometric analysis. RESULTS Signs of damage were observed in the mitochondrial morphology after exposure to reperfusion. The mitochondrial swelling induced by Ca(2+) increased gradually with the increasing calcium concentration, and this tendency was exacerbated as the reperfusion time was extended. Cyclophilin D protein expression peaked after 24 hours of reperfusion. The mitochondrial membrane potential was decreased significantly during the reperfusion period, with the greatest decrease observed after 24 hours of reperfusion. The surge in mitochondrial reactive oxidative species occurred after 2 hours of reperfusion and was maintained at a high level during the reperfusion period. CONCLUSIONS Reperfusion following focal cerebral ischemia induced significant mitochondrial morphological damage and Ca(2+)-induced mitochondrial swelling. The mechanism of this swelling may be mediated by the upregulation of the Cyclophilin D protein, the destruction of the mitochondrial membrane potential and the generation of excessive reactive oxidative species.
Collapse
Affiliation(s)
- Jun Li
- Department of Anesthesiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xuesong Ma
- Department of Anesthesiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wei Yu
- Department of Anesthesiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhangqun Lou
- Department of Anesthesiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Dunlan Mu
- Department of Anesthesiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Ying Wang
- Department of Anesthesiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Baozhong Shen
- Molecular Imaging Key Laboratory of General Universities and Colleges of Heilongjiang Province, Harbin, China
| | - Sihua Qi
- Department of Anesthesiology, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Müller MM, Middelanis J, Meier C, Surbek D, Berger R. 17β-estradiol protects 7-day old rats from acute brain injury and reduces the number of apoptotic cells. Reprod Sci 2012; 20:253-61. [PMID: 22875845 DOI: 10.1177/1933719112452471] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To test a possible neuroprotective activity of 17β-estradiol in the neonatal rat brain exposed to hypoxic-ischemia (controlled hypoxia after unilateral carotid artery ligation). METHODS Seven-day-old Wistar rats underwent ligation of the left common carotid artery followed by 80 minutes hypoxia in 8% oxygen inducing an ipsilateral brain damage. Seven days later (d14), brains were analyzed quantitatively using a macroscopic and microscopic score for structural damage, hemisphere volumes were calculated, and immunohistochemistry for cleaved-caspase-3 (marker for apoptotic cells) was performed. Animals from the study group (n = 19) received 17β-estradiol (0.05 µg/g body weight intraperitoneally) before (-64, -40, and -16 hours) and after (+3 hours) the hypoxia (hour 0: start of the hypoxia) and the control group (n = 21) received mock treatment. RESULTS Of the 21 pups, 13 in the NaCl group had macroscopically a severe brain damage and 7 of 19 animals in the study group encountered only discrete to mild lesions. Microscopic brain damage in the study group was significantly lower (score 1.5 ± 0.7 vs 2.8 ± 0.8, P < .05). The determined volumes of the affected hemisphere were significantly lower in the NaCl group than in the treatment group. The numbers of apoptotic cells in both hemispheres was equal in the estradiol group, but in the control group, there were significantly more apoptotic cells in the affected hemisphere (control group: ipsilateral: 1435 ± 653 vs contralateral: 143 ± 57 cells, P < .05). DISCUSSION 17β-Estradiol protects newborn rat brains from hypoxic-ischemic injury, in terms of both microscopic cell injury and apoptosis.
Collapse
Affiliation(s)
- Martin M Müller
- Department of Obstetrics and Gynecology, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland.
| | | | | | | | | |
Collapse
|
14
|
Keogh MJ, Drury PP, Bennet L, Davidson JO, Mathai S, Gunn ER, Booth LC, Gunn AJ. Limited predictive value of early changes in EEG spectral power for neural injury after asphyxia in preterm fetal sheep. Pediatr Res 2012; 71:345-53. [PMID: 22391634 DOI: 10.1038/pr.2011.80] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION This study examined whether spectral analysis of the electroencephalogram (EEG) can discriminate between mild and severe hypoxic-ischemic injury in the immature brain. RESULTS Total EEG power was profoundly suppressed after umbilical cord occlusion and recovered to baseline by 5 h after 15-min of occlusion, in contrast with transient recovery in the 25-min (P < 0.05). Power spectra were not different between groups in the first 3 h; α and β power were significantly higher in the 15-min group from 4 h, and Δ and θ power from 5 h (P < 0.05). The 25-min group showed severe neuronal loss in hippocampal regions and basal ganglia at 3 days, in contrast with no/minimal injury in the 15-min group. DISCUSSION EEG power after asphyxia did not discriminate between mild and severe injury in the first 3 h in preterm fetal sheep. Severe subcortical neural injury was associated with persistent loss of high-frequency activity. METHODS Chronically instrumented fetal sheep at 0.7 gestation (101-104 days; term is 147 days) received either 15-min (n = 13) or 25-min (n = 13) of complete umbilical cord occlusion. The Δ (0-3.9 Hz), θ (4-7.9 Hz), α (8-12.9 Hz), and β (13-22 Hz) components of the EEG were determined by power spectral analysis. Brains were taken at 3 days for histopathology.
Collapse
Affiliation(s)
- Michael J Keogh
- Fetal Physiology and Neuroscience Group, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Fujikawa DG, Zhao S, Ke X, Shinmei SS, Allen SG. Mild as well as severe insults produce necrotic, not apoptotic, cells: evidence from 60-min seizures. Neurosci Lett 2009; 469:333-7. [PMID: 20026247 DOI: 10.1016/j.neulet.2009.12.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 12/11/2009] [Accepted: 12/14/2009] [Indexed: 10/20/2022]
Abstract
We tested the hypothesis that mild insults produce apoptotic, and severe insults necrotic, cells by subjecting adult Wistar rats to 60-min instead of 3-h generalized seizures. Rats' brains were evaluated 6 and 24h later for evidence of neuronal necrosis by light and electron microscopy, the presence of TUNEL staining and active caspase-3 immunoreactivity, and for evidence of DNA laddering 24h after seizures. Apoptotic neurons from the retrosplenial cortex of postnatal day 8 rat pups served as positive controls. Six and 24h after seizures, 16 and 15 brain regions respectively out of 24 showed significant numbers of acidophilic neurons by hematoxylin and eosin stain. Three brain regions had significant numbers of TUNEL-positive neurons 24h after seizures. No neurons showed active caspase-3 immunoreactivity. Acidophilic neurons were necrotic by electron-microscopic examination. Ultrastructurally, they were shrunken and electron-dense, with shrunken, pyknotic nuclei and swollen mitochondria with disrupted cristae. Nuclei did not contain the irregular chromatin clumps found after 3-h seizures. None of the six brain regions studied ultrastructurally that show DNA laddering 24h after 3-h seizures showed DNA laddering 24h after 60-min seizures, probably because there were too few damaged neurons, although the lack of chromatin clumping might have been a contributing factor. Following seizures, a mild as well as a severe insult produces caspase-3-negative necrotic neurons. These results do not support the hypothesis that mild insults produce apoptotic, and severe insults, necrotic, cells.
Collapse
Affiliation(s)
- Denson G Fujikawa
- Experimental Neurology Laboratory, VA Greater Los Angeles Healthcare System, North Hills, CA 91343-2036, USA.
| | | | | | | | | |
Collapse
|
16
|
Tashlykov V, Katz Y, Volkov A, Gazit V, Schreiber S, Zohar O, Pick CG. Minimal traumatic brain injury induce apoptotic cell death in mice. J Mol Neurosci 2008; 37:16-24. [PMID: 18651249 DOI: 10.1007/s12031-008-9094-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2008] [Accepted: 04/25/2008] [Indexed: 11/29/2022]
Abstract
In the United States, 1.4 million people suffer from traumatic brain injury (TBI) each year because of traffic, sports, or war-related injuries. The majority of TBI victims suffer mild to minimal TBI (mTBI), but most are released undiagnosed. Detailed pathologies are poorly understood. We characterized the microscopic changes of neurons of closed-head mTBI mice after increased unilateral trauma using hematoxylin and eosin (H&E) stain, and correlated it with the expression of the apoptotic proteins c-jun, p53, and BCL-2. Minimal damage to the brain increases the number of pyknotic appearing neurons and activates the apoptotic proteins in both hemispheres. Although minimal, increased impact was positively correlated with the increased number of damaged neurons. These results may explain the wide variety of behavioral and cognitive deficits closed-head mTBI causes in mice. Our cumulative results point to the pathological origin of post-concussion syndrome and may aid in the development of future neuroprotective strategies for the disease.
Collapse
Affiliation(s)
- V Tashlykov
- Laboratory of Anesthesia, Pain and Neural Research, Bruce Rapaport Medical Faculty, Technion Israel Institute of Technology, Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
17
|
Svedin P, Guan J, Mathai S, Zhang R, Wang X, Gustavsson M, Hagberg H, Mallard C. Delayed peripheral administration of a GPE analogue induces astrogliosis and angiogenesis and reduces inflammation and brain injury following hypoxia-ischemia in the neonatal rat. Dev Neurosci 2007; 29:393-402. [PMID: 17762207 DOI: 10.1159/000105480] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Accepted: 01/05/2007] [Indexed: 11/19/2022] Open
Abstract
Glycine 2-methyl proline glutamate (G-2mPE) is a proline-modified analogue to the naturally existing N-terminal tripeptide glycine-proline-glutamate that is a cleaved product from insulin-like growth factor-1. G-2mPE is designed to be more enzymatically resistant than glycine-proline-glutamate and to increase its bioavailability. The current study has investigated the protective effects of G-2mPE following hypoxic-ischemic brain injury in the neonatal brain. On postnatal day 7, Wistar rats were exposed to hypoxia-ischemia (HI). HI was induced by unilateral ligation of the left carotid artery followed by hypoxia (7.7% O2, 36 degrees C) for 60 min. The drug treatment started 2 h after the insult, and the pups were given either 1.2 mg/kg (bolus), 1.2 mg/ml once a day for 7 days, or vehicle. The degree of brain damage was determined histochemically by thionin/acid fuchsin staining. G-2mPE's anti-inflammatory properties were investigated by IL-1beta, IL-6, and IL-18 ELISA, and effects on apoptosis by caspase 3 activity. Vascularization was determined immunohistochemically by the total length of isolectin-positive blood vessels. Effect on astrocytosis was also determined in the hippocampus. Animals treated with multiple doses of G-2mPE demonstrated reduced overall brain injury 7 days after HI, particularly in the hippocampus and thalamus compared to vehicle-treated rats. The expression of IL-6 was decreased in G-2mPE-treated animals compared to vehicle-treated pups, and both the capillary length and astrogliosis were increased in the drug-treated animals. There was no effect on caspase 3 activity. This study indicates that peripheral administration of G-2mPE, starting 2 h after a hypoxic-ischemic insult, reduces the degree of brain injury in the immature rat brain. The normalization of IL-6 levels and the promotion of both neovascularization and reactive astrocytosis may be potential mechanisms that underlie its protective effects.
Collapse
Affiliation(s)
- Pernilla Svedin
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Graham D, Brown A, Cavanagh J, Meldrum B. Obituary. Neuropathol Appl Neurobiol 2005. [DOI: 10.1111/j.1365-2990.2004.00631.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Guan J, Thomas GB, Lin H, Mathai S, Bachelor DC, George S, Gluckman PD. Neuroprotective effects of the N-terminal tripeptide of insulin-like growth factor-1, glycine-proline-glutamate (GPE) following intravenous infusion in hypoxic-ischemic adult rats. Neuropharmacology 2005; 47:892-903. [PMID: 15527823 DOI: 10.1016/j.neuropharm.2004.07.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2004] [Revised: 06/11/2004] [Accepted: 06/30/2004] [Indexed: 11/24/2022]
Abstract
The N-terminal tripeptide of insulin-like growth factor-1, GPE is neuroprotective when given intracerebroventricularly 2 h after hypoxic-ischemic (HI) brain injury in rats. We have now examined whether GPE can cross the blood-brain barrier and exert neuroprotective actions following intravenous administration. Following a single bolus intravenous injection, GPE was rapidly metabolized and cleared from the circulation. The short half-life (<2 min) in blood was subsequently associated with modest and inconsistent neuroprotection. In contrast, potent neuroprotection of GPE was consistently observed in all brain regions examined following 4 h intravenous infusion (12 mg/kg). The neuroprotective effects of GPE after infusion showed a broad effective dose range (1.2-120 mg/kg) and an extended window of treatment to 7-11 h after injury. The central penetration of GPE after intravenous infusion was injury-dependent. GPE also improved long-term somatofunction with a comparable neuronal outcome. GPE reduced both caspase-3-dependent and -independent apoptosis in the hippocampus. Treatment with GPE also inhibited microglial proliferation and prevented the injury-induced loss of astrocytes. In conclusion, the neuroprotective actions of GPE infusion were global, robust and displayed a broad effective dose range and treatment window. GPE's activity included the prevention of neuronal apoptosis, promotion of astrocyte survival and inhibition of microglial proliferation. With injury specific central penetration, GPE has considerable promise as a systemic neuroprotective treatment after acute encephalopathies.
Collapse
Affiliation(s)
- J Guan
- The Liggins Institute, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, 2-6 Park Avenue, Grafton, Auckland, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
20
|
Petersson KH, Pinar H, Stopa EG, Sadowska GB, Hanumara RC, Stonestreet BS. Effects of exogenous glucose on brain ischemia in ovine fetuses. Pediatr Res 2004; 56:621-9. [PMID: 15319457 DOI: 10.1203/01.pdr.0000139415.96985.bf] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We examined the effects of prolonged moderate hyperglycemia with and without an additional rapid glucose injection on ischemic brain injury in the fetus. Twenty-five ewes (117-124 d of gestation) were assigned to one of four groups: 1) glucose-infused fetuses exposed to 30 min of carotid artery occlusion followed by 48 h of reperfusion (I/R-Glu, n = 8); 2) glucose-infused plus rapid glucose injection given 100 min before 30 min of occlusion followed by 48 h of reperfusion (I/R-GluR, n = 4); 3) placebo-infused exposed to 30 min of occlusion and 48 h of reperfusion (I/R-PL, n = 8); and 4) glucose-infused sham occlusion and 48 h of sham reperfusion (control, n = 5). After baseline measurements, fetuses were infused with glucose (9-16 mg/kg/min) for 48 h before and after carotid occlusion or sham treatment. The I/R-PL group received 0.9% NaCl. Brain pathologic outcome was determined. Serial sections stained with Luxol fast blue-hematoxylin and eosin were scored for white matter, cerebral cortical, and hippocampal lesions. These areas received graded pathologic scores of 0 to 5, reflecting the amount of injury, where 0 = 0%, 1 = 1-25%, 2 = 26-50%, 3 = 51-75%, 4 = 76-95%, and 5 = 96-100% of the area damaged. Comparisons of the pathologic scores for cerebral cortex (CC), white matter (WM), and hippocampus (H) demonstrated that the I/R-GluR (CC: 4.56 +/- 0.11, WM: 4.50 +/- 0.11, H: 3.44 +/- 0.48, mean +/- SEM) had more (p < 0.05) damage than the I/R-Glu (CC: 2.46 +/- 0.47, WM: 1.97 +/- 0.37, H: 1.81 +/- 0.36) and control (CC: 1.12 +/- 0.13, WM: 0.82 +/- 0.34, H: 0.80 +/- 0.34) groups. The pathologic scores in the I/R-Glu were (p < 0.05) greater than the control, but not the I/R-PL (CC: 2.12 +/- 0.35, WM: 2.20 +/- 0.44, H: 1.59 +/- 0.41) group. We conclude that exposure to prolonged moderate hyperglycemia before ischemia and during reperfusion does not affect the extent of brain injury, but exposure to an additional acute increase in plasma glucose concentration before ischemia is extremely detrimental to the fetal brain.
Collapse
Affiliation(s)
- Katherine H Petersson
- Department of Pediatrics, Brown University Medical School, Providence, Rhode Island 02905, USA
| | | | | | | | | | | |
Collapse
|
21
|
Andersson B, Wu X, Bjelke B, Syková E. Temporal profile of ultrastructural changes in cortical neurons after a photochemical lesion. J Neurosci Res 2004; 77:901-12. [PMID: 15334608 DOI: 10.1002/jnr.20217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A photochemical lesion was induced in the right sensory motor cortex of rat brains. We examined at various time points the occurrence of different types of neuronal death with respect to a potential therapeutic window. The lesion appearance was documented by magnetic resonance imaging, and functional recovery was evaluated by behavioral tests showing recovery at 48 hr after lesioning. At 0.5, 1, 3, 6, 12, 24, 48, and 72 hr postlesion, cortical layers IV and V were examined by light and electron microscopy. Ultrastructural changes, which corresponded well to light microscopy findings, were found in both hemispheres. In the lesioned area, the neuropil appeared disorganized at 0.5 hr, and apoptotic and necrotic cell death was found at 0.5-3 hr. After 3 hr, the tissue was disintegrated. On the contralateral side, chromatin clumping appeared at 0.5-3 hr. At 3 hr, ruptured membranes were found, a sign of irreversible cell death. At 6-72 hr, the membranes were intact, and the chromatin was not clumped but heterogeneously distributed. The nuclei contained dispersed nucleoli at 48-72 hr. The morphology correlated well with magnetic resonance images and functional behavior. Our study demonstrates that a photochemical lesion is a useful model for studying morphological changes in injured cells. It results in a permanent infarction within 3 hr. In that the morphology on the contralateral side drastically changed between 3 and 6 hr, the cellular alterations at these time points might represent a break point at which cells either progress toward cell death or recover.
Collapse
Affiliation(s)
- Benita Andersson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| | | | | | | |
Collapse
|
22
|
Berger R, Middelanis J, Vaihinger HM, Mies G, Wilken B, Jensen A. Creatine protects the immature brain from hypoxic-ischemic injury. ACTA ACUST UNITED AC 2004; 11:9-15. [PMID: 14706677 DOI: 10.1016/j.jsgi.2003.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE We tested the neuroprotective effects of creatine against hypoxic-ischemic injury in the immature brain. METHODS Hippocampal slices were prepared from fetal guinea pigs at 0.9 gestation and incubated in artificial cerebrospinal fluid (aCSF) equilibrated with carbogen. Slices were subjected to oxygen-glucose deprivation (OGD) for 30 or 40 minutes. Two hours after OGD, adenosine triphosphate (ATP) and protein synthesis were analyzed. Creatine (3 mM) was applied to tissue slices of the study groups 2 hours before the insult. In a second set of experiments 7-day-old Wistar rats were anesthetized, and the left carotid artery was ligated. After 1 hour of recovery the pups were subjected to a hypoxic gas mixture (8% oxygen and 92% nitrogen) for 80 minutes. Seven days later the brains of the neonates were removed and analyzed for hypoxic-ischemic injury. The rat pups of the test group were treated with creatine (3 g/kg subcutaneously) before (-64 hours, -40 hours, and -16 hours) and after (+3 hours) the hypoxic-ischemic insult, with zero time corresponding to the start of hypoxia, whereas the animals of the control group received the solvent. RESULTS Creatine significantly improved the recovery of protein synthesis 2 hours after OGD in hippocampal slices but had no effect on ATP levels. Whereas seven animals of the control group developed severe cystic cerebral infarction, only mild to moderate damage was observed in the rat pups of the study group. In contrast to creatine-treated pups, the volume of the ipsilateral hemisphere was considerably smaller than that of the contralateral one in control animals (104 +/- 22 versus 138 +/- 14 mL, P<.001). Except at the frontal level (A 6.0 mm), neuronal cell injury was significantly lower in the cortex of the animals that had received creatine. This was also true for the evaluated subfields in the hippocampus. CONCLUSION We conclude that creatine protects the immature brain from hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Richard Berger
- Department of Obstetrics and Gynecology, University of Bochum, Bochum, Germany.
| | | | | | | | | | | |
Collapse
|
23
|
Zukin RS, Jover T, Yokota H, Calderone A, Simionescu M, Lau CG. Molecular and Cellular Mechanisms of Ischemia-Induced Neuronal Death. Stroke 2004. [DOI: 10.1016/b0-44-306600-0/50049-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
Elitt CM, Sadowska GB, Stopa EG, Pinar H, Petersson KH, Stonestreet BS. Effects of antenatal steroids on ischemic brain injury in near-term ovine fetuses. Early Hum Dev 2003; 73:1-15. [PMID: 12932889 DOI: 10.1016/s0378-3782(03)00030-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Hypoxia/ischemia in utero can result in brain damage to the fetus and newborn. Antenatal steroids are a routine part of the management of women who develop premature labor. Pretreatment of young postnatal rats with dexamethasone before hypoxic/ischemic insults has been reported to attenuate brain injury. However, the effects of antenatal steroids on ischemic brain injury in fetuses have not been investigated. OBJECTIVE We examined the effects of maternally administered antenatal corticosteroids on ischemic brain injury in near-term ovine fetuses. METHODS Chronically instrumented fetuses at 122 days of gestation were studied 12 h after the last of four 4 mg dexamethasone, or placebo injections were given over 48 h to the ewes. Groups were dexamethasone/ischemic, placebo/ischemic and sham-treated control. Fetuses were exposed to 30 min of carotid occlusion (ischemia) or no occlusion (control) and 72 h of reperfusion. Whole brain coronal sections stained with Luxol fast blue-hematoxylin-eosin were scored for white matter and cerebral cortical lesions. Both areas received pathological scores of 0 to 5 reflecting the degree of injury (0=0%, 1=1-10%, 2=11-50%, 3=51-90%, 4=91-99% and 5=100%). Bilateral carotid blood flow also was measured before, during and after brain ischemia in the dexamethasone/ischemic and placebo/ischemic groups. RESULTS White matter (WM) and cerebral cortical scores did not differ between the dexamethasone/ischemic and placebo/ischemic (WM: 3.0+/-1.9 and 2.9+/-1.7; cortex: 3.1+/-1.7 and 2.6+/-1.8, mean+/-S.D.) groups. White matter and cerebral cortical scores were higher in the dexamethasone/ischemic (WM: 3.0+/-1.9, P<0.02; cortex: 3.1+/-1.7, P<0.005) and placebo/ischemic (WM: 2.9+/-1.7, P<0.006; cortex: 2.6+/-1.8, P<0.007) than control (WM: 0.2+/-0.4; cortex: 0.2+/-0.4) group. Carotid blood flow was relatively higher (P<0.05) after 24, 48 and 72 h of reperfusion in the dexamethasone/ischemic than placebo/ischemic group. CONCLUSIONS We conclude that maternal pretreatment with antenatal dexamethasone did not attenuate ischemic brain injury in the fetus, and that carotid blood flow was higher during reperfusion in fetuses of dexamethasone than placebo-treated ewes, most likely secondary to decreases in arterial oxygen tension.
Collapse
Affiliation(s)
- Christopher M Elitt
- Department of Pediatrics, Brown University School of Medicine, Women and Infants' Hospital of Rhode Island, 101 Dudley Street, Providence, RI 02905-240, USA
| | | | | | | | | | | |
Collapse
|
25
|
Coumans ABC, Middelanis JS, Garnier Y, Vaihinger HM, Leib SL, Von Duering MU, Hasaart THM, Jensen A, Berger R. Intracisternal application of endotoxin enhances the susceptibility to subsequent hypoxic-ischemic brain damage in neonatal rats. Pediatr Res 2003; 53:770-5. [PMID: 12621122 DOI: 10.1203/01.pdr.0000059221.40073.82] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Perinatal brain damage is associated not only with hypoxic-ischemic insults but also with intrauterine inflammation. A combination of antenatal inflammation and asphyxia increases the risk of cerebral palsy >70 times. The aim of the present study was to determine the effect of intracisternal (i.c.) administration of endotoxin [lipopolysaccharides (LPS)] on subsequent hypoxic-ischemic brain damage in neonatal rats. Seven-day-old Wistar rats were subjected to i.c. application of NaCl or LPS (5 microg/pup). One hour later, the left common carotid artery was exposed through a midline neck incision and ligated with 6-0 surgical silk. After another hour of recovery, the pups were subjected to a hypoxic gas mixture (8% oxygen/92% nitrogen) for 60 min. The animals were randomized to four experimental groups: 1) sham control group, left common carotid artery exposed but not ligated (n = 5); 2) LPS group, subjected to i.c. application of LPS (n = 7); 3) hypoxic-ischemic study group, i.c. injection of NaCl and exposure to hypoxia after ligation of the left carotid artery (n = 17); or 4) hypoxic-ischemic/LPS study group, i.c. injection of LPS and exposure to hypoxia after ligation of the left carotid artery (n = 19). Seven days later, neonatal brains were assessed for neuronal cell damage. In a second set of experiments, rat pups received an i.c. injection of LPS (5 microg/pup) and were evaluated for tumor necrosis factor-alpha expression by immunohistochemistry. Neuronal cell damage could not be observed in the sham control or in the LPS group. In the hypoxic-ischemic/LPS group, neuronal injury in the cerebral cortex was significantly higher than in animals that were subjected to hypoxia/ischemia after i.c. application of NaCl. Injecting LPS intracisternally caused a marked expression of tumor necrosis factor-alpha in the leptomeninges. Applying LPS intracisternally sensitizes the immature rat brain to a subsequent hypoxic-ischemic insult.
Collapse
Affiliation(s)
- Audrey B C Coumans
- Department of Obstetrics and Gynecology, University Hospital Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Hoffman JR, Greenberg JH, Furuya D, Craik RL, Fanelli P, Breslow S, Sheehan S, Ketschek A, Damkaoutis C, Reivich M, Hand P. Rats recovering from unilateral barrel-cortex ischemia are capable of completing a whisker-dependent task using only their affected whiskers. Brain Res 2003; 965:91-9. [PMID: 12591124 DOI: 10.1016/s0006-8993(02)04141-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Rats use their vibrissae for a variety of exploratory tasks including location of objects and discrimination of texture. This study examines recovery in vibrissal function following a unilateral ischemic injury to the somatosensory cortex. Vibrissal function was examined in adult food-restricted rats performing on a two-texture discrimination device. Animals were trained and tested until the criteria of >80% correct choices was demonstrated on three consecutive days. Ischemic rats were constrained to use the affected whiskers by clipping the ipsilateral vibrissae. One group was tested after ischemia, a second group was trained before ischemia and then tested, and a third group was pre-trained and received whisker stimulation and tested post-ischemia. Nai;ve animals recovering from ischemia took longer to reach criteria than intact or unilateral trimmed control animals. Pre-trained animals with compression ischemia receiving whisker stimulation with sucrose water completed the task to criteria in the fewest number of trials. The results indicate that recovery of vibrissal function occurs following a unilateral ischemic injury. Histological analysis in animals without whisker stimulation indicates that the number of normal appearing cortical barrels following ischemia was inversely correlated to the number of trials needed to complete the behavioral task. This suggests that the natural recovery of the ability to discriminate textures is related to the degree of damage to the barrel cortex. The relationship between cortical barrels and behavioral recovery did not hold for the ischemic animals receiving whisker stimulation. This latter group demonstrated recovery despite marked anatomical lesions suggesting that the intervention influenced reorganization.
Collapse
Affiliation(s)
- John R Hoffman
- Department of Biology, Arcadia University, 450 S. Easton Road, Glenside, PA 19038, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Structural and Functional Damage Sustained by Mitochondria After Traumatic Brain Injury in the Rat: Evidence for Differentially Sensitive Populations in the Cortex and Hippocampus. J Cereb Blood Flow Metab 2003. [DOI: 10.1097/00004647-200302000-00009] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Lifshitz J, Friberg H, Neumar RW, Raghupathi R, Welsh FA, Janmey P, Saatman KE, Wieloch T, Grady MS, McIntosh TK. Structural and functional damage sustained by mitochondria after traumatic brain injury in the rat: evidence for differentially sensitive populations in the cortex and hippocampus. J Cereb Blood Flow Metab 2003; 23:219-31. [PMID: 12571453 DOI: 10.1097/01.wcb.0000040581.43808.03] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The cellular and molecular pathways initiated by traumatic brain injury (TBI) may compromise the function and structural integrity of mitochondria, thereby contributing to cerebral metabolic dysfunction and cell death. The extent to which TBI affects regional mitochondrial populations with respect to structure, function, and swelling was assessed 3 hours and 24 hours after lateral fluid-percussion brain injury in the rat. Significantly less mitochondrial protein was isolated from the injured compared with uninjured parietotemporal cortex, whereas comparable yields were obtained from the hippocampus. After injury, cortical and hippocampal tissue ATP concentrations declined significantly to 60% and 40% of control, respectively, in the absence of respiratory deficits in isolated mitochondria. Mitochondria with ultrastructural morphologic damage comprised a significantly greater percent of the population isolated from injured than uninjured brain. As determined by photon correlation spectroscopy, the mean mitochondrial radius decreased significantly in injured cortical populations (361 +/- 40 nm at 24 hours) and increased significantly in injured hippocampal populations (442 +/- 36 at 3 hours) compared with uninjured populations (Ctx: 418 +/- 44; Hipp: 393 +/- 24). Calcium-induced deenergized swelling rates of isolated mitochondrial populations were significantly slower in injured compared with uninjured samples, suggesting that injury alters the kinetics of mitochondrial permeability transition (MPT) pore activation. Cyclosporin A (CsA)-insensitive swelling was reduced in the cortex, and CsA-sensitive and CsA-insensitive swelling both were reduced in the hippocampus, demonstrating that regulated MPT pores remain in mitochondria isolated from injured brain. A proposed mitochondrial population model synthesizes these data and suggests that cortical mitochondria may be depleted after TBI, with a physically smaller, MPT-regulated population remaining. Hippocampal mitochondria may sustain damage associated with ballooned membranes and reduced MPT pore calcium sensitivity. The heterogeneous mitochondrial response to TBI may underlie posttraumatic metabolic dysfunction and contribute to the pathophysiology of TBI.
Collapse
Affiliation(s)
- Jonathan Lifshitz
- Head Injury Center, Department of Neurosurgery, University of Pensylvania, Philadelphia, Pensylvania 19104-6316, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Hotta H, Uchida S, Kagitani F. Effects of stimulating the nucleus basalis of Meynert on blood flow and delayed neuronal death following transient ischemia in the rat cerebral cortex. THE JAPANESE JOURNAL OF PHYSIOLOGY 2002; 52:383-93. [PMID: 12519473 DOI: 10.2170/jjphysiol.52.383] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
An increase in cortical cerebral blood flow (CBF), independent of metabolic vasodilation, via the activation of cholinergic neurons originating in the nucleus basalis of Meynert (NBM) in the basal forebrain and projecting to the widespread cortices was recently demonstrated. In the present study, we aimed to clarify whether the increase in CBF following a stimulation of the NBM can improve delayed death of the cortical neurons following transient ischemia in rats. CBF was measured with a laser Doppler flowmeter, and the delayed neuronal death of the cerebral cortex produced by intermittent (every 5 s) occlusions of the unilateral common carotid artery for 60 min was measured histologically in the cortical hemisphere at 3 different coronal levels (6 microm thickness). In control rats without occlusion there were 6000-8000 intact neurons and 9-19 damaged neurons in the cortical hemisphere at each coronal level. During the occlusions, CBF ipsilateral to the occluded artery decreased by 13-32% of the preocclusion level. Five days after the occlusions, the numbers of damaged neurons were increased to 75-181. Repetitive electrical stimulation was delivered to the NBM, ipsilateral to the occluded artery, starting 5 min before the occlusions and finishing around the end of them. The increase in CBF induced by NBM stimulation prevented the occlusion-induced decrease in CBF in all 3 of the cortices. The delayed death of the cortical neurons previously observed after the occlusions was scarcely observable in all the cortices when NBM was stimulated. The present results suggest that NBM-originating vasodilative activation can protect the ischemia-induced delayed death of cortical neurons by preventing a blood flow decrease in widespread cortices.
Collapse
Affiliation(s)
- Harumi Hotta
- Motor and Autonomic Nervous System Integration Research Group, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, 173-0015 Japan.
| | | | | |
Collapse
|
30
|
Petersson KH, Pinar H, Stopa EG, Faris RA, Sadowska GB, Hanumara RC, Stonestreet BS. White matter injury after cerebral ischemia in ovine fetuses. Pediatr Res 2002; 51:768-76. [PMID: 12032276 DOI: 10.1203/00006450-200206000-00019] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The effects of cerebral ischemia on white matter changes in ovine fetuses were examined after exposure to bilateral carotid artery occlusion. Fetal sheep were exposed to 30 min of ischemia followed by 48 (I/R-48, n = 8) or 72 (I/R-72, n = 10) h of reperfusion or control sham treatment (control, n = 4). Serial coronal sections stained with Luxol fast blue/hematoxylin and eosin were scored for white matter, cerebral cortical, and hippocampal lesions. All areas received graded pathologic scores of 0 to 5, reflecting the degree of injury where 0 = 0%, 1 = 1% to 25%, 2 = 26% to 50%, 3 = 51% to 75%, 4 = 76% to 95%, and 5 = 96% to 100% of the area damaged. Dual-label immunofluorescence using antibodies against glial fibrillary acidic protein (GFAP) and myelin basic protein (MBP) were used to characterize white matter lesions. Basic fibroblast growth factor (FGF-2) was measured in the frontal cortex by ELISA. Results of the pathologic scores showed that the white matter of the I/R-72 (2.74 +/- 0.53, mean +/- SEM) was more (p < 0.05) damaged when compared with the control (0.80 +/- 0.33) group. Cortical lesions were greater (p < 0.05) in the I/R-48 (2.12 +/- 0.35) than the control (0.93 +/- 0.09) group. White matter lesions were characterized by reactive GFAP-positive astrocytes and a loss of MBP in oligodendrocytes. The ratio of MBP to GFAP decreased (p < 0.05) as a function of ischemia, indicative of a proportionally greater loss of MBP than GFAP. FGF-2 concentrations were higher (p < 0.05) in the I/R-72 than the control group and there was a direct correlation between the pathologic scores (PS) and FGF-2 concentrations (FGF-2 = e((1.6 PS-0.90)) + 743, n = 17, r = 0.73, p < 0.001). We conclude that carotid artery occlusion results in quantifiable white matter lesions that are associated with a loss of MBP from myelin, and that FGF-2, a purported mediator of recovery from brain injury in adult subjects, increases in concentration in proportion to the severity of brain damage in the fetus.
Collapse
Affiliation(s)
- Katherine H Petersson
- Department of Pediatrics, Brown University School of Medicine, Providence, Rhode Island 02906, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Garnier Y, Löbbert T, Jensen A, Berger R. Lubeluzole pretreatment does not provide neuroprotection against transient global cerebral ischemia in fetal sheep near term. Pediatr Res 2002; 51:517-22. [PMID: 11919339 DOI: 10.1203/00006450-200204000-00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The aim of the present study was to test the neuroprotective effect of the novel benzothiazol compound lubeluzole on neuronal cell damage in fetal sheep arising from global cerebral ischemia. Thirteen fetal sheep were prepared at a mean gestational age of 127 +/- 1 d (term is at 147 d). Six fetuses were treated with lubeluzole (0.33 mg/kg estimated body weight) before induction of global cerebral ischemia (-90, -60, and -30 min), while the remainder (n = 7) received solvent. Cerebral ischemia was induced by occluding both carotid arteries for 30 min. Cerebral blood flow was measured by injecting radio-labeled microspheres before (-90 min), during (+3 min and +27 min), and after (+40 min, +3 h, and +72 h) cerebral ischemia. Neuronal cell damage was assessed in the cerebrum and deeper brain structures by light microscopy. Values are given as means +/- SD. In control fetuses, blood flow to the cerebrum was reduced from 100 +/- 25 mL.100 g(-1) min(-1) to less than 20 mL.100 g(-1) min(-1) during ischemia. Shortly after ischemia, hyperperfusion occurred (217 +/- 66 mL.100 g(-1)min(-1)) followed by a tendency toward hypoperfusion (72 +/- 17 mL.100 g(-1) min(-1)) later on (+3 h). Significant differences in blood flow to the various brain structures between the control and study groups could not be observed. Neuronal cell damage was concentrated in the parasagittal regions of the cerebrum. Preischemic application of lubeluzole did not have any effect on the extent of neuronal cell damage. From these results, we conclude that pretreatment with lubeluzole fails to protect the brain of fetal sheep near term from injury after transient global cerebral ischemia. However, because the observation period lasted only 3 d, a possible effect of lubeluzole on pathophysiological mechanisms inducing delayed neuronal cell death cannot be fully excluded.
Collapse
Affiliation(s)
- Yves Garnier
- Department of Obstetrics and Gynecology, Ruhr-Universität Bochum, Bochum, Germany
| | | | | | | |
Collapse
|
32
|
Obernier JA, Bouldin TW, Crews FT. Binge Ethanol Exposure in Adult Rats Causes Necrotic Cell Death. Alcohol Clin Exp Res 2002. [DOI: 10.1111/j.1530-0277.2002.tb02573.x] [Citation(s) in RCA: 188] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
33
|
Solenski NJ, diPierro CG, Trimmer PA, Kwan AL, Helm GA, Helms GA. Ultrastructural changes of neuronal mitochondria after transient and permanent cerebral ischemia. Stroke 2002; 33:816-24. [PMID: 11872909 DOI: 10.1161/hs0302.104541] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Mitochondrial swelling is one of the most striking and initial ultrastructural changes after acute brain ischemia. The purpose of the present study was to examine the role of reperfusion of the cerebral cortex after transient focal cerebral ischemia on neuronal mitochondrial damage. METHODS Male Sprague-Dawley rats (n=16) were subjected to either temporary or permanent occlusion of the middle cerebral artery and bilateral carotid arteries. Three experimental conditions were compared: group I, permanent ischemia (3, 5, and 24 hours); group II, transient ischemia (2, 24 hours of reperfusion); and sham surgery. Anesthetized rats were killed by cardiac perfusion, and brain tissue was removed ipsilaterally and contralaterally from the ischemic core section of the frontoparietal cortex. Fixed tissue was prepared for electron microscopic examination, and electron microscopic thin sections of random neurons were photographed. Perinuclear neuronal mitochondria were analyzed in a blinded manner for qualitative ultrastructural changes (compared with sham control) by 2 independent investigators using an objective grading system. RESULTS Cortical neuronal mitochondria exposed to severe ischemic/reperfusion conditions demonstrated dramatic signs of injury in the form of condensation, increased matrix density, and deposits of electron-dense material followed by disintegration by 24 hours. In contrast, mitochondria exposed to an equivalent time of permanent ischemia demonstrated increasing loss of matrix density with pronounced swelling followed by retention of their shape by 24 hours. CONCLUSIONS Neuronal mitochondria undergoing transient versus permanent ischemia exhibit significantly different patterns of injury. Structural damage to neuronal mitochondria of the neocortex occurs more acutely and to a greater extent during the reperfusion phase in comparison to ischemic conditions alone. Further research is in progress to delineate the role of oxygen free radical production in the observed mitochondrial damage during postischemic reoxygenation.
Collapse
Affiliation(s)
- Nina J Solenski
- Department of Neurology, University of Virginia, Health Sciences System, Charlottesville 22908, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Guan J, Miller OT, Waugh KM, McCarthy DC, Gluckman PD. Insulin-like growth factor-1 improves somatosensory function and reduces the extent of cortical infarction and ongoing neuronal loss after hypoxia-ischemia in rats. Neuroscience 2002; 105:299-306. [PMID: 11672597 DOI: 10.1016/s0306-4522(01)00145-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Treatment with insulin-like growth factor-1 has been demonstrated to reduce the extent of cortical infarction 5 days after hypoxic-ischemic brain injury. As neuronal death can be progressive and long lasting after initial injury, the present study examined the long-term effects of insulin-like growth factor-1 on late neuronal loss 20 days after hypoxic-ischemic injury, together with evaluating neurobehavioral outcome as assumed by somatosensory function. Unilateral brain injury was induced in adult rats by carotid artery ligation followed by 10 min of hypoxia (6% O2). A single dose of insulin-like growth factor-1 (50 microg) was administered intracerebroventricularly via a stereotaxically pre-fixed cannula 2 h after injury. A bilateral tactile stimulation test was used to examine the degree of somatosensory function at 3, 5, 10 and 20 days after the hypoxia in both insulin-like growth factor-1- (n=12) and its vehicle- (n=12) treated rats, along with sham-operated rats (n=9). Cortical infarction and percentage of selective neuronal loss in the cerebral cortex were examined 20 days after the hypoxic-ischemic injury in both treatment groups. Hypoxic-ischemic injury resulted in a significant delay in the time taken to contact the patch over the period examined (left/right ratio 5.1+/-0.79), particularly at 3 days (7.0+/-2.8) after the hypoxia, compared to sham-operated rats (1.1+/-0.9, P<0.05). The overall effect of insulin-like growth factor-1 in reducing the time taken to contact the patch was significant (P=0.03, 2.6+/-0.79) compared to the vehicle group. There was a trend towards a reduction of cortical infarction after insulin-like growth factor-1 treatment (P=0.058), however insulin-like growth factor-1 significantly reduced the percentage of selective neuronal loss (P=0.027) 20 days following the hypoxia. From these data we suggest that insulin-like growth factor-1 improves somatosensory function by reducing both the extent of cortical infarction and ongoing progressive neuronal death during brain recovery from hypoxic-ischemic injury.
Collapse
Affiliation(s)
- J Guan
- Liggins Institute, Faculty of Medicine and Health Sciences, The University of Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
35
|
Wang MJ, Huang HM, Hsieh SJ, Jeng KC, Kuo JS. Resveratrol inhibits interleukin-6 production in cortical mixed glial cells under hypoxia/hypoglycemia followed by reoxygenation. J Neuroimmunol 2001; 112:28-34. [PMID: 11108930 DOI: 10.1016/s0165-5728(00)00374-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Reactive oxygen intermediates (ROIs) are important mediators of a variety of pathological processes, including inflammation and ischemia/reperfusion injury. Cytokines and chemokines are detected at mRNA level in human and animal ischemic brains. This suggests that hypoxia/reoxygenation may induce cytokine production through generation of ROIs. In this study, we investigated the cytokine induction and inhibition by antioxidants in rat cortical mixed glial cells exposed to in vitro ischemia-like insults (hypoxia plus glucose deprivation). The results showed that interleukin-6 (IL-6) mRNA and protein, but not tumor necrosis factor-alpha (TNF-alpha) or interleukin-1beta (IL-1beta), were induced during hypoxia/hypoglycemia followed by reoxygenation in the mixed glial cells. The accumulation of IL-6 mRNA was induced as early as 15 min after hypoxia/hypoglycemia and its level was further increased after subsequent reoxygenation. Among the antioxidants studied, only resveratrol suppressed IL-6 gene expression and protein secretion in mixed glial cultures under hypoxia/hypoglycemia followed by reoxygenation. These findings suggest that resveratrol might be useful in treating ischemic-induced inflammatory processes in stroke.
Collapse
Affiliation(s)
- M J Wang
- Department of Education and Research, Taichung Veterans General Hospital, 40705, ROC, Taichung, Taiwan
| | | | | | | | | |
Collapse
|
36
|
|
37
|
Kagitani F, Uchida S, Hotta H, Sato A. Effects of nicotine on blood flow and delayed neuronal death following intermittent transient ischemia in rat hippocampus. THE JAPANESE JOURNAL OF PHYSIOLOGY 2000; 50:585-95. [PMID: 11173554 DOI: 10.2170/jjphysiol.50.585] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A cholinergic neural vasodilative response in the cerebral cortex and hippocampus, independent of metabolic vasodilation, was recently demonstrated by activating the nicotinic acetylcholine receptors (nAChRs) via activation of cholinergic neurons originating in the nucleus basalis of Meynert and septal complex in the basal forebrain and projecting to the cortex and hippocampus (see reviews by Sato A and Sato Y: Neurosci Res 14: 242--274, 1992; Sato A and Sato Y: Alzheimer Dis Assoc Disord 9: 28--38, 1995). In the present study, we aimed to examine whether an increase in regional blood flow in the hippocampus (Hpc-BF) following stimulation of the nAChRs by i.v. injection of nicotine could improve the delayed death of the hippocampal neurons following transient ischemia in rats. Hpc-BF was measured by using a laser Doppler flowmeter. During intermittent (every 2 min) transient occlusion for a total of 6 min of bilateral carotid arteries besides permanent ligation of bilateral vertebral arteries, Hpc-BF decreased to about 16% of the preocclusion level, and 5 or 7 d later, after the occlusion, delayed neuronal death occurred in approximately 70% of the CA1 hippocampal neurons. Hpc-BF was increased dose-dependently by injection of nicotine (30--100 microg/kg, i.v.), independent of mean arterial pressure. Nicotine (30--100 microg/kg) administered 5 min before occlusion slightly but significantly attenuated the occlusion-induced decrease in Hpc-BF. The delayed death of the CA1 hippocampal neurons occurring after transient occlusion was attenuated by pretreatment with nicotine (30--100 microg/kg) to approximately 50% of the total neurons. The results indicate that nAChR stimulation-induced increases in Hpc-BF can protect against ischemia-induced delayed death of hippocampal neurons.
Collapse
Affiliation(s)
- F Kagitani
- Department of the Autonomic Nervous System, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015 Japan
| | | | | | | |
Collapse
|
38
|
Abstract
This review examines the appearance of hallmarks of apoptosis following experimental stroke. The reviewed literature leaves no doubt that ischemic cell death in the brain is active, that is, requires energy; is gene directed, that is, requires new gene expression; and is capase-mediated, that is, uses apoptotic proteolytic machinery. However, sufficient differences to both classical necrosis and apoptosis exist which prevent easy mechanistic classification. It is concluded that ischemic cell death in the brain is neither necrosis nor apoptosis but is a chimera which appears on a continuum that has apoptosis and necrosis at the poles. The position on this continuum could be modulated by the intensity of the ischemic injury, the consequent availability of ATP and new protein synthesis, and both the age and context of the neuron in question. Thus the ischemic neuron may look necrotic but have actively died in an energy dependent manner with new gene expression and destruction via the apoptotic proteolytic machinery.
Collapse
Affiliation(s)
- J P MacManus
- Apoptosis Research Group, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario.
| | | |
Collapse
|
39
|
Fujikawa DG. Confusion between neuronal apoptosis and activation of programmed cell death mechanisms in acute necrotic insults. Trends Neurosci 2000; 23:410-1. [PMID: 10941188 DOI: 10.1016/s0166-2236(00)01601-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
40
|
Fujikawa DG, Shinmei SS, Cai B. Kainic acid-induced seizures produce necrotic, not apoptotic, neurons with internucleosomal DNA cleavage: implications for programmed cell death mechanisms. Neuroscience 2000; 98:41-53. [PMID: 10858610 DOI: 10.1016/s0306-4522(00)00085-3] [Citation(s) in RCA: 161] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Prolonged seizures (status epilepticus) induced by kainic acid activate programmed cell death mechanisms, and it is believed that kainic acid-induced status epilepticus induces neuronal apoptosis. In order to test this hypothesis, adult rats were subjected to 3-h kainic acid-induced seizures, with 24- or 72-h recovery periods. Neuronal death was assessed by light microscopy with the Hematoxylin and Eosin stain and with in situ terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL stain), by electron microscopy, and by agarose gel electrophoresis of DNA extracted from five vulnerable brain regions. Spontaneous and MK-801-induced apoptotic neurons from retrosplenial cortex of neonatal rats, evaluated by light and electron microscopy, were used as positive controls for apoptosis. Surprisingly, the large chromatin clumps of apoptotic neurons were TUNEL negative, whereas the cytoplasm showed light-to-moderate TUNEL staining, consistent with a lack of identifiable nuclear membranes ultrastructurally, and with intermingling of nuclear and cytoplasmic contents. Ultrastructurally, the acidophilic neurons produced by kainic acid-induced status epilepticus, identified with Hematoxylin and Eosin stain, were dark, shrunken and necrotic, with pyknotic nuclei containing small, dispersed chromatin clumps, and with cytoplasmic vacuoles, some of which were swollen, disrupted mitochondria. No apoptotic cells were seen. Acidophilic neurons were found in up to 20 of 23 brain regions examined and comprised 10-25% of the total number of neurons examined. A subset of these neurons (<10% of the total number of neurons in five of 23 regions) had TUNEL-positive nuclei 72h but not 24h after status epilepticus. Internucleosomal DNA cleavage (DNA "laddering") occurred in the four most damaged brain regions examined by electron microscopy 24h after SE and the three most damaged regions 72h after status epilepticus. Our results demonstrate that kainic acid-induced status epilepticus produces neuronal necrosis and not apoptosis in adult rats. The necrotic neurons show nuclear pyknosis, chromatin condensation and DNA laddering. Programmed cell death mechanisms activated by kainic acid-induced status epilepticus occur in neurons which become necrotic and could contribute to necrotic, as well as apoptotic, neuronal death.
Collapse
Affiliation(s)
- D G Fujikawa
- Experimental Neurology Laboratory, VA Greater Los Angeles Healthcare System, Sepulveda Ambulatory Care Center and Nursing Home Care Unit, Sepulveda CA 91343, USA.
| | | | | |
Collapse
|
41
|
Victorov IV, Prass K, Dirnagl U. Improved selective, simple, and contrast staining of acidophilic neurons with vanadium acid fuchsin. BRAIN RESEARCH. BRAIN RESEARCH PROTOCOLS 2000; 5:135-9. [PMID: 10775832 DOI: 10.1016/s1385-299x(00)00004-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Acidophilia is one of the hallmarks of acute neuronal damage and death in brain ischemia, excitotoxic and traumatic lesions and epileptic seizures. We here describe a novel and simple method for visualizing acidophilic neurons on paraffin sections, using vanadium acid fuchsin (VAF) staining and toluidine blue or hematoxylin counterstaining. Paraffin sections of the brain fixed in ethanol-formalin-acetic acid mixture are stained in 0.1% acid fuchsin containing 0.125% of ammonium metavanadate and 1% of glacial acetic acid, differentiated if overstained in 0.01% of borax solution, and counterstained with 0.05-0.025% of toluidine blue in acetate buffer (pH 3.3) or Gill's II hematoxylin. The sections are dehydrated, cleared in xylene and mounted in Canada balsam or any synthetic mounting media for light microscopy. VAF combined with toluidine blue or hematoxylin results in highly selective and reproducible color contrast staining of acidophilic neurons as well as glial nuclei and hyperchromatic neurons. As a progressive method, acid fuchsin staining usually does not require differentiation. The red acidophilic neurons are clearly visible on the background of non-damaged cells, which significantly facilitates the identification, and localization of damaged neurons, even at low magnification under the light microscope.
Collapse
Affiliation(s)
- I V Victorov
- Laboratory of Experimental Neurocytology, Brain Research Institute, Pereulok Obukha 5, 103064, Moscow, Russia.
| | | | | |
Collapse
|
42
|
Guan J, Gunn AJ, Sirimanne ES, Tuffin J, Gunning MI, Clark R, Gluckman PD. The window of opportunity for neuronal rescue with insulin-like growth factor-1 after hypoxia-ischemia in rats is critically modulated by cerebral temperature during recovery. J Cereb Blood Flow Metab 2000; 20:513-9. [PMID: 10724116 DOI: 10.1097/00004647-200003000-00010] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Insulin-like growth factor (IGF-1) is induced in damaged brain tissue after hypoxia-ischemia, and exogenous administration of IGF-1 shortly after injury has been shown to be neuroprotective. However, it is unknown whether treatment with IGF-1 delayed by more than a few hours after injury may be protective. Hypothermia after brain injury has been reported to delay the development of ischemic neuronal death. The authors therefore hypothesize that a reduction in the environmental temperature during recovery from hypoxia-ischemia could prolong the window of opportunity for IGF-1 treatment. Unilateral brain damage was induced in adult rats using a modified Levine model of right carotid artery ligation followed by brief hypoxia (6% O2 for 10 minutes). The rats were maintained in either a warm (31 degrees C) or cool (23 degrees C) environment for the first 2 hours after hypoxia. All rats were subsequently transferred to the 23 degrees C environment until the end of the experiment. A single dose of IGF-1 (50 microg) or its vehicle was given intracerebroventricularly at either 2 or 6 hours after hypoxia. Histologic outcome in the lateral cortex was quantified 5 days after hypoxia. Finally, cortical temperature was recorded from 1 hour before and 2 hours after hypoxia in separate groups of rats exposed to the "warm" and "cool" protocols. In rats exposed to the warm recovery environment, IGF-1 reduced cortical damage (P < 0.05) when given 2 hours but not 6 hours after insult. In contrast, with early recovery in the cool environment, a significant protective effect of IGF-1 in the lateral cortex (P < 0.05) was found with administration 6 hours after insult. In conclusion, a reduction in cerebral temperature during the early recovery phase after severe hypoxia-ischemia did not significantly reduce the severity of injury after 5 days' recovery; however, it markedly shifted and extended the window of opportunity for delayed treatment with IGF-1.
Collapse
Affiliation(s)
- J Guan
- Research Center for Developmental Medicine and Biology, University of Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
This review is directed at understanding how neuronal death occurs in two distinct insults, global ischemia and focal ischemia. These are the two principal rodent models for human disease. Cell death occurs by a necrotic pathway characterized by either ischemic/homogenizing cell change or edematous cell change. Death also occurs via an apoptotic-like pathway that is characterized, minimally, by DNA laddering and a dependence on caspase activity and, optimally, by those properties, additional characteristic protein and phospholipid changes, and morphological attributes of apoptosis. Death may also occur by autophagocytosis. The cell death process has four major stages. The first, the induction stage, includes several changes initiated by ischemia and reperfusion that are very likely to play major roles in cell death. These include inhibition (and subsequent reactivation) of electron transport, decreased ATP, decreased pH, increased cell Ca(2+), release of glutamate, increased arachidonic acid, and also gene activation leading to cytokine synthesis, synthesis of enzymes involved in free radical production, and accumulation of leukocytes. These changes lead to the activation of five damaging events, termed perpetrators. These are the damaging actions of free radicals and their product peroxynitrite, the actions of the Ca(2+)-dependent protease calpain, the activity of phospholipases, the activity of poly-ADPribose polymerase (PARP), and the activation of the apoptotic pathway. The second stage of cell death involves the long-term changes in macromolecules or key metabolites that are caused by the perpetrators. The third stage of cell death involves long-term damaging effects of these macromolecular and metabolite changes, and of some of the induction processes, on critical cell functions and structures that lead to the defined end stages of cell damage. These targeted functions and structures include the plasmalemma, the mitochondria, the cytoskeleton, protein synthesis, and kinase activities. The fourth stage is the progression to the morphological and biochemical end stages of cell death. Of these four stages, the last two are the least well understood. Quite little is known of how the perpetrators affect the structures and functions and whether and how each of these changes contribute to cell death. According to this description, the key step in ischemic cell death is adequate activation of the perpetrators, and thus a major unifying thread of the review is a consideration of how the changes occurring during and after ischemia, including gene activation and synthesis of new proteins, conspire to produce damaging levels of free radicals and peroxynitrite, to activate calpain and other Ca(2+)-driven processes that are damaging, and to initiate the apoptotic process. Although it is not fully established for all cases, the major driving force for the necrotic cell death process, and very possibly the other processes, appears to be the generation of free radicals and peroxynitrite. Effects of a large number of damaging changes can be explained on the basis of their ability to generate free radicals in early or late stages of damage. Several important issues are defined for future study. These include determining the triggers for apoptosis and autophagocytosis and establishing greater confidence in most of the cellular changes that are hypothesized to be involved in cell death. A very important outstanding issue is identifying the critical functional and structural changes caused by the perpetrators of cell death. These changes are responsible for cell death, and their identity and mechanisms of action are almost completely unknown.
Collapse
Affiliation(s)
- P Lipton
- Department of Physiology, University of Wisconsin School of Medicine, Madison, Wisconsin, USA
| |
Collapse
|
44
|
Laurini RN, Arbeille B, Gemberg C, Akoka S, Locatelli A, Lansac J, Arbeille P. Brain damage and hypoxia in an ovine fetal chronic cocaine model. Eur J Obstet Gynecol Reprod Biol 1999; 86:15-22. [PMID: 10471137 DOI: 10.1016/s0301-2115(99)00036-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To assess the development of brain damage in an ovine fetal chronic cocaine model. To evaluate the effect of isolated hypoxic tests on this model and to correlate hemodynamic findings (brain-sparing effect) following fetal hypoxia and the occurrence of brain damage. STUDY DESIGN Fifteen ewes were divided into a control group (n=7) and a cocaine treated group (n=8). From day 65 to day 134 the cocaine treated animals received a daily (5 days per week) intramuscular injection (2 mg/kg cocaine) and the control animals a placebo injection (2 ml of isotonic solution). Both groups underwent hypoxic tests (cord compression (3 min) and aortic compression (1 min)) at 90 and 134 days. In addition, anesthesia for magnetic resonance imaging (MRI) examination was carried out at 125 days. Fetal blood samples were collected during both series of hypoxic tests and the cerebral and umbilical flows were monitored by Doppler. Samples from 25 brains (control n = 10; cocaine n= 15) were processed for light and electron microscopic examination. Quantification of brain damage was done on semithin sections from six areas of cortex and germinal matrix on each fetus. RESULTS Similar forms of brain damage (selective neuronal loss limited to the parasaggital cortex, striatum, hippocampus and Purkinje cells) was present in both groups but lesions were more frequent in the cocaine treated group as shown by quantitative analysis for the proportion of abnormal capillaries (65% vs. 35%), capillary edema (61% vs. 34%) and abnormal neurons showing delayed neuronal degeneration (DND) (66% vs. 36%) in the cocaine and control group respectively. There was no significant difference in immunoreactivity for glial fibrillary acidic protein (GFAP) but it was more marked in the cerebellum of cocaine treated animals. Fetal blood samples showed a moderate sustained hypoxia and Doppler findings demonstrated the presence of a brain sparing effect associated with increased uterine and umbilical vascular resistance in the cocaine treated group. Nevertheless, the amplitude of the heart rate increase and cerebral dilatation was significantly lower in the cocaine treated animals. CONCLUSION This ovine fetal chronic cocaine model showed the presence of brain damage. Cocaine treatment seems to potentiate the effect of the hypoxic tests. Independent of the cause, the brain damage developed in the presence of brain sparing effect, strongly suggesting that this phenomenon is a sign of a pathological fetal condition and no guarantee that it will prevent tissue damage.
Collapse
Affiliation(s)
- R N Laurini
- Institut Universitaire de Pathologie, Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
45
|
Wu DD, Lai M, Hughes PE, Sirimanne E, Gluckman PD, Williams CE. Expression of the activin axis and neuronal rescue effects of recombinant activin A following hypoxic-ischemic brain injury in the infant rat. Brain Res 1999; 835:369-78. [PMID: 10415398 DOI: 10.1016/s0006-8993(99)01638-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Neurotrophic factors are induced in the brain in response to injury and may restrict the extent of neuronal loss and facilitate recovery. We have previously reported a strong neuronal induction of activin betaA subunit mRNA expression after a hypoxic-ischemic (HI) injury in the rat brain. Here, we further extended our studies to examine a role for the activin inhibitory binding protein, follistatin after injury and also to determine the potential of activin as a neuronal rescue agent. Ribonuclease protection assay (RPA) was used to quantify the time course of the mRNA expression of activin betaA subunit and follistatin, following a 60-min HI brain injury. Activin betaA subunit mRNA level increased in the contralateral hemisphere 5 h after injury and returned to normal at 10 h post injury. In contrast, follistatin mRNA levels decreased in the same hemisphere at 5 and 10 h after injury. The effect of intracerebroventrically (i. c.v.) administered recombinant human activin A or its antagonist, inhibin A, on neuronal death after a 15-min HI brain injury was determined for a number of brain regions. One microgram activin A (n=23) reduced the neuronal loss in the hippocampal CA1/2 region, dorsolateral striatum but not in the parietal cortex. In contrast, 1 microg of inhibin A (n=18) did not have a significant effect on the extent of neuronal loss in any of the affected regions. This pattern of neuroprotection was consistent with the distribution of immunoreactivity for the activin receptor type II subunit. These results demonstrate that activin A, but not its functional antagonist inhibin A, can enhance the survival of injured hippocampal and striatal neurons. Since follistatin is thought to exert a neutralising effect on activin A activity, the down-regulation of follistatin expression post injury may be allowing activin A to become more accessible to neurons after injury. Overall, these results suggest a role of the activin axis in modulating the survival of specific populations of injured neurons.
Collapse
Affiliation(s)
- D D Wu
- Research Centre for Developmental Medicine and Biology, School of Medicine, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | | | | | |
Collapse
|
46
|
Fujikawa DG, Shinmei SS, Cai B. Lithium-pilocarpine-induced status epilepticus produces necrotic neurons with internucleosomal DNA fragmentation in adult rats. Eur J Neurosci 1999; 11:1605-14. [PMID: 10215913 DOI: 10.1046/j.1460-9568.1999.00573.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Prolonged and continuous epileptic seizures [status epilepticus (SE)] produce a widespread pattern of neuronal death, primarily in limbic brain regions. Because it has been suggested that seizure-induced neuronal death may be apoptotic in nature, we tested the hypothesis that lithium-pilocarpine-induced status epilepticus (LPCSE) produces apoptotic neurons. LPCSE lasting 3 h was induced in male Wistar rats which were allowed to recover for 24 or 72 h before perfusion-fixation. Neuronal death was assessed by light microscopy with the haematoxylin-and-eosin stain (H&E), with in situ DNA nick-end labelling (TUNEL stain), by electron microscopy, and by agarose gel electrophoresis of DNA extracted from vulnerable brain regions. Ultrastructurally, acidophilic neurons identified with H&E were dark, shrunken and necrotic in appearance, exhibiting pyknotic nuclei, irregular, dispersed chromatin clumps and cytoplasmic vacuolization. No cells with apoptotic features were seen. Acidophilic neurons were found in 21 out of 23 brain regions examined, and comprised 26-45% of the total number of neurons examined. A subset of these neurons (< 10% of the total number of neurons) were TUNEL-positive at 72 h, but not 24 h, after SE. Internucleosomal DNA cleavage (DNA 'laddering') was found in the six brain regions examined ultrastructurally 24 and 72 h after SE. These results indicate that, in adult rats, LPCSE produces neuronal injury with the appearance of necrosis rather than apoptosis. The necrotic neurons show nuclear pyknosis, chromatin condensation and internucleosomal DNA fragmentation, confirming the nonspecificity of these nuclear changes. Internucleosomal DNA cleavage and other programmed cell death mechanisms can be activated by SE in neurons which become necrotic.
Collapse
Affiliation(s)
- D G Fujikawa
- Experimental Neurology Laboratory, VA Greater Los Angeles Healthcare System, Sepulveda Ambulatory Care Center and Nursing Home, Sepulveda, CA 91343, USA.
| | | | | |
Collapse
|
47
|
Guan J, Waldvogel HJ, Faull RL, Gluckman PD, Williams CE. The effects of the N-terminal tripeptide of insulin-like growth factor-1, glycine-proline-glutamate in different regions following hypoxic-ischemic brain injury in adult rats. Neuroscience 1999; 89:649-59. [PMID: 10199602 DOI: 10.1016/s0306-4522(98)00338-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Insulin-like growth factor-1 has pleiotropic effects in the central nervous system and can act both as a survival and a differentiation factor. Insulin-like growth factor-1 can be proteolytically cleaved into des-N-(1-3)-insulin-like growth factor-1 and a N-terminal tripeptide fragment, glycine-proline-glutamate. Both insulin-like growth factor-1 and des-N-(1-3)-insulin-like growth factor-1 can improve neuronal survival after hypoxic-ischemic brain injury in vivo. The present study investigates the effects of glycine-proline-glutamate on different brain regions and neuronal populations after hypoxic-ischemic injury. Unilateral hypoxic-ischemic injury was induced in adult rats. Glycine-proline-glutamate (3 microg) was administered centrally 2 h after the injury and the extent of brain damage determined five days later. In a separate trial immunohistochemical techniques were used to determine the effects of glycine-proline-glutamate on specific populations of neurons in the striatum after the injury. Compared to the vehicle treatment, glycine-proline-glutamate (n=19) treatment reduced the extent of cortical damage and neuronal loss in the CA1-2 subregions of the hippocampus (P<0.05). In the striatum, there was a trend towards a reduction in neuronal loss after glycine-proline-glutamate treatment (P=0.053) compared to the vehicle (n=21)-treated animals. In a separate study, glycine-proline-glutamate (n=8) treatment prevented the loss of choline acetyltransferase (P<0.05), glutamate acid decarboxylase (P<0.05) and somatostatin (P<0.05) containing neurons in the ipsilateral striatum following hypoxic-ischemic brain injury and also increased the numbers of neuronal nitric oxide synthase (P<0.05) containing neurons in the contralateral side. These studies suggest that in addition to neuroprotective effects, glycine-proline-glutamate can influence neuronal activity after hypoxic-ischemic injury.
Collapse
Affiliation(s)
- J Guan
- Research Centre for Developmental Medicine and Biology, University of Auckland, New Zealand
| | | | | | | | | |
Collapse
|
48
|
Davies CA, Loddick SA, Stroemer RP, Hunt J, Rothwell NJ. An integrated analysis of the progression of cell responses induced by permanent focal middle cerebral artery occlusion in the rat. Exp Neurol 1998; 154:199-212. [PMID: 9875281 DOI: 10.1006/exnr.1998.6891] [Citation(s) in RCA: 79] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Defining the chronology and severity of cell damage in an evolving lesion after ischemia is important for understanding the underlying mechanisms in the development of therapeutic intervention. In the present study, we used a combination of histological and immunocytochemical methods to evaluate cell responses from 30 min to 48 h after permanent occlusion of the middle cerebral artery (MCAO) in the rat. Specific immunocytochemical markers clearly revealed acute early responses in neurons (neurofilament protein 200), astrocytes (glial fibrillary acidic protein), and microglia/macrophages (OX-42 and ED-1) such as enlarged, convoluted neuronal processes, and disintegration of glia. Progressive topographic changes in the developing lesion, pinpointed by immunolabeling, indicated the severity and extension of the cell damage. Proliferation and hypertrophy of astrocytes and microglia around the infarct, and contralaterally, occurred 24-48 h after MCAO and coincided with mass necrosis and infiltration of neutrophils and macrophages into the core. These observations corroborate the suggestion that the inflammatory process is involved in the progression of the infarct.
Collapse
Affiliation(s)
- C A Davies
- School of Biological Sciences, University of Manchester, United Kingdom
| | | | | | | | | |
Collapse
|
49
|
Berger R, Lehmann T, Karcher J, Garnier Y, Jensen A. Low dose flunarizine protects the fetal brain from ischemic injury in sheep. Pediatr Res 1998; 44:277-82. [PMID: 9727701 DOI: 10.1203/00006450-199809000-00003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Flunarizine, a calcium channel blocker, reduced cerebral damage caused by hypoxic-ischemic insults in neonatal rats and in fetal sheep near term. However, the high dose regimen used in these studies produced cardiovascular side effects that might have counteracted the neuroprotective properties of flunarizine. Therefore, the neuroprotective effect was tested in a low dose protocol (1 mg/kg estimated body weight). Twelve fetal sheep near term were instrumented chronically. Six fetuses were pretreated with 1 mg of flunarizine per kg of estimated body weight 1 h before ischemia, whereas the remainder (n=6) received solvent. Cerebral ischemia was induced by occluding both carotid arteries for 30 min. To exclude the possibility that the neuroprotective effects of flunarizine were caused by cerebrovascular alterations we measured cerebral blood flow by injecting radiolabeled microspheres before (-1 h), during (3 min and 27 min) and after (40 min, 3 h, and 72 h) cerebral ischemia. At the end of the experiment (72 h) the ewe was given a lethal dose of sodium pentobarbitone and saturated potassium chloride i.v., and the fetal brain was perfused with formalin. Neuronal cell damage was assessed in various brain structures by light microscopy after cresyl violet/fuchsin staining using a scoring system: 1, 0-5% damage; 2, 5-50% damage; 3, 50-95% damage; 4, 95-99% damage; and 5, 100% damage. In 10 other fetal sheep effects of low dose flunarizine on circulatory centralization caused by acute asphyxia could be excluded. In the treated group neuronal cell damage was reduced significantly in many cerebral areas to varying degrees (range for control group, 1.03-2.14 versus range for treated group, 1.00-1.13; p < 0.05 to p < 0.001, respectively). There were only minor differences in blood flow to the various brain structures between groups. We conclude that pretreatment with low dose flunarizine protects the brain of fetal sheep near term from ischemic injury. This neuroprotective effect is not mediated by changes in cerebral blood flow. We further conclude that low dose flunarizine may be clinically useful as a treatment providing fetal neuroprotection, particularly because the fetal cardiovascular side effects are minimal.
Collapse
Affiliation(s)
- R Berger
- Department of Obstetrics and Gynecology, Ruhr-Universität Bochum, Germany
| | | | | | | | | |
Collapse
|
50
|
Jones PA, Smith RA, Stone TW. Protection against hippocampal kainate excitotoxicity by intracerebral administration of an adenosine A2A receptor antagonist. Brain Res 1998; 800:328-35. [PMID: 9685693 DOI: 10.1016/s0006-8993(98)00540-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have previously shown that the peripheral administration of an A2A receptor agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamidoadenosine hydrochloride (CGS 21680) protected the hippocampus against kainate-induced excitotoxicity. The present study utilises the intrahippocampal route to further investigate CGS 21680-mediated protection as well as examining the role of adenosine and both A1 and A2A receptors in kainate-induced excitotoxicity. Injections were made directly into the hippocampus of anaesthetised male Wistar rats. Following surgery and the administration of 0.25 nmol kainate in 1 microl of solution, the animals were left to recover for seven days before perfusion and brain slicing. Haematoxylin and eosin staining revealed substantial damage to the CA3 region. Co-administration of the A2A receptor agonist CGS 21680 over a range of doses did not protect the region to any degree. Similarly neither the A1 receptor agonist R-phenylisopropyladenosine (R-PIA), nor adenosine itself reduced kainate-induced damage. The intrahippocampal injection of the selective A2A receptor antagonist, 4-(2-[7-amino-2-¿2-furyl¿¿1,2, 4¿triazolo¿2,3-a¿¿1,3,5¿triazin-5-yl-amino]ethyl)phenol (ZM241385) however, significantly decreased kainate damage to the CA3 region. These results show that adenosine A2A receptor-induced protection is most likely to be mediated peripherally and is probably not due to activation of A2A receptors within the hippocampus. The lack of protection observed with either R-PIA or adenosine may be due to an inhibitory action of the A2A receptor on the neuroprotective A1 receptor. Importantly, this study also questions the role of endogenously released adenosine in protecting the hippocampus from excitotoxic damage.
Collapse
Affiliation(s)
- P A Jones
- Institute of Biomedical and Life Sciences, Laboratory of Human Anatomy, University of Glasgow, Glasgow G12 8QQ, UK
| | | | | |
Collapse
|