1
|
Gramespacher H, Schmieschek MHT, Warnke C, Adler C, Bittner S, Dronse J, Richter N, Zaeske C, Gietzen C, Schlamann M, Baldus S, Fink GR, Onur OA. Analysis of Cerebral CT Based on Supervised Machine Learning as a Predictor of Outcome After Out-of-Hospital Cardiac Arrest. Neurology 2024; 103:e209583. [PMID: 38857458 DOI: 10.1212/wnl.0000000000209583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES In light of limited intensive care capacities and a lack of accurate prognostic tools to advise caregivers and family members responsibly, this study aims to determine whether automated cerebral CT (CCT) analysis allows prognostication after out-of-hospital cardiac arrest. METHODS In this monocentric, retrospective cohort study, a supervised machine learning classifier based on an elastic net regularized logistic regression model for gray matter alterations on nonenhanced CCT obtained after cardiac arrest was trained using 10-fold cross-validation and tested on a hold-out sample (random split 75%/25%) for outcome prediction. Following the literature, a favorable outcome was defined as a cerebral performance category of 1-2 and a poor outcome of 3-5. The diagnostic accuracy was compared with established and guideline-recommended prognostic measures within the sample, that is, gray matter-white matter ratio (GWR), neuron-specific enolase (NSE), and neurofilament light chain (NfL) in serum. RESULTS Of 279 adult patients, 132 who underwent CCT within 14 days of cardiac arrest with good imaging quality were identified. Our approach discriminated between favorable and poor outcomes with an area under the curve (AUC) of 0.73 (95% CI 0.59-0.82). Thus, the prognostic power outperformed the GWR (AUC 0.66, 95% CI 0.56-0.76). The biomarkers NfL, measured at days 1 and 2, and NSE, measured at day 2, exceeded the reliability of the imaging markers derived from CT (AUC NfL day 1: 0.87, 95% CI 0.75-0.99; AUC NfL day 2: 0.90, 95% CI 0.79-1.00; AUC NSE day: 2 0.78, 95% CI 0.62-0.94). DISCUSSION Our data show that machine learning-assisted gray matter analysis of CCT images offers prognostic information after out-of-hospital cardiac arrest. Thus, CCT gray matter analysis could become a reliable and time-independent addition to the standard workup with serum biomarkers sampled at predefined time points. Prospective studies are warranted to replicate these findings.
Collapse
Affiliation(s)
- Hannes Gramespacher
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Maximilian H T Schmieschek
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Clemens Warnke
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Christoph Adler
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Stefan Bittner
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Julian Dronse
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Nils Richter
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Charlotte Zaeske
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Carsten Gietzen
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Marc Schlamann
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Stephan Baldus
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Gereon R Fink
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Oezguer A Onur
- From the Department of Neurology (H.G., M.H.T.S., C.W., J.D., N.R., G.R.F., O.A.O.), Faculty of Medicine and University Hospital Cologne; Division of Cardiology, Pneumology, Angiology and Intensive Care (C.A., S. Baldus), Department of Internal Medicine III, University of Cologne; Department of Neurology (S. Bittner), University Medical Center Mainz; Cognitive Neuroscience (N.R., O.A.O.), Institute of Neuroscience and Medicine (INM-3), Research Center Jülich; and Institute for Diagnostic and Interventional Radiology (C.Z., C.G., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| |
Collapse
|
2
|
Yilmazer-Hanke D, Ouali Alami N, Fang L, Klotz S, Kovacs GG, Pankratz H, Weis J, Katona I, Scheuerle A, Streit WJ, Del Tredici K. Differential Glial Chitotriosidase 1 and Chitinase 3-like Protein 1 Expression in the Human Primary Visual Cortex and Cerebellum after Global Hypoxia-Ischemia. Neuroscience 2022; 506:91-113. [PMID: 36332693 DOI: 10.1016/j.neuroscience.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/15/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Here, we studied the neuroinflammation- and ischemia-related glial markers chitotriosidase 1 (CHIT1) and chitinase-3-like protein 1 (CHI3L1, alias YKL-40) in the human striate cortex and cerebellum at different time points after global hypoxic-ischemic brain injury (HIBI). Both regions differ considerably in their glial cell population but are supplied by the posterior circulation. CHIT1 and CHI3L1 expression was compared to changes in microglial (IBA1, CD68), astrocytic (GFAP, S100β), and neuronal markers (H&E, neurofilament heavy chain, NfH; calretinin, CALR) using immunohistochemistry and multiple-label immunofluorescence. Initial striatal cortical and cerebellar Purkinje cell damage, detectable already 1/2 d after HIBI, led to delayed neuronal death, whereas loss of cerebellar NfH-positive stellate and CALR-positive granule cells was variable. During the first week post-HIBI, a transient reduction of IBA1-positive microglia was observed in both regions, and fragmented/clasmatodendritic cerebellar Bergmann glia appeared. In long-term survivors, both brain regions displayed high densities of activated IBA1-positive cells and CD68-positive macrophages, which showed CHIT1 co-localization in the striate cortex. Furthermore, enlarged GFAP- and S100β-positive astroglia emerged in both regions around 9-10 d post-HIBI, i.e., along with clearance of dead neurons from the neuropil, although GFAP-/S100β-positive gemistocytic astrocytes that co-expressed CHI3L1 were found only in the striate cortex. Thus, only GFAP-/S100β-positive astrocytes in the striate cortex, but not cerebellar Bergmann glia, differentiated into CHI3L1-positive gemistocytes. CHIT1 was co-expressed almost entirely in macrophages in the striate cortex and not cerebellum of long-term survivors, thereby indicating that CHIT1 and CHI3L1 could be valuable biomarkers for monitoring the outcome of global HIBI.
Collapse
Affiliation(s)
- Deniz Yilmazer-Hanke
- Clinical Neuroanatomy, Neurology, School of Medicine, Ulm University, Ulm, Germany.
| | - Najwa Ouali Alami
- Clinical Neuroanatomy, Neurology, School of Medicine, Ulm University, Ulm, Germany
| | - Lubin Fang
- Clinical Neuroanatomy, Neurology, School of Medicine, Ulm University, Ulm, Germany
| | - Sigried Klotz
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Gabor G Kovacs
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Helmut Pankratz
- Institute of Forensic Medicine, Medical Faculty, Ludwig-Maximilian University Munich, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Istvan Katona
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Angelika Scheuerle
- Department of Pathology, Section Neuropathology, University Hospital, Ulm, Germany
| | - Wolfgang J Streit
- Department of Neuroscience, College of Medicine, University of Florida, FL, USA
| | - Kelly Del Tredici
- Clinical Neuroanatomy, Neurology, School of Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
3
|
Griffiths B, Xu L, Sun X, Greer M, Murray I, Stary C. Inhibition of microRNA-200c preserves astrocyte sirtuin-1 and mitofusin-2, and protects against hippocampal neurodegeneration following global cerebral ischemia in mice. Front Mol Neurosci 2022; 15:1014751. [PMID: 36466801 PMCID: PMC9710226 DOI: 10.3389/fnmol.2022.1014751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Memory impairment remains a leading disability in survivors of global cerebral ischemia, occurring secondary to delayed neurodegeneration of hippocampal cornu ammonis-1 (CA1) neurons. MicroRNA-200c (miR-200c) is induced following ischemic stress and we have previously demonstrated that pre-treatment with anti-miR-200c is protective against embolic stroke in mice. In the present study we assessed the role of miR-200c on CA1 neurodegeneration, sirtuin-1 (SIRT1), and mitochondrial dynamic protein expression in a mouse model of transient global cerebral ischemia and in vitro in primary mouse astrocyte cultures after simulated ischemia. Mice were subjected to 10 min bilateral common carotid artery occlusion plus hypotension with 5% isoflurane. After 2 h recovery mice were treated with intravenous injection of either anti-miR-200c or mismatch control. Memory function was assessed by Barnes maze at post-injury days 3 and 7. Mice were sacrificed at post-injury day 7 for assessment of brain cell-type specific expression of miR-200c, SIRT1, and the mitochondrial fusion proteins mitofusin-2 (MFN2) and OPA1 via complexed fluorescent in situ hybridization and fluorescent immunohistochemistry. Global cerebral ischemia induced significant loss of CA1 neurons, impaired memory performance and decreased expression of CA1 SIRT1, MFN2, and OPA1. Post-injury treatment with anti-miR-200c significantly improved survival, prevented CA1 neuronal loss, improved post-injury performance in Barnes maze, and was associated with increased post-injury expression of CA1 SIRT1 and MFN2 in astrocytes. In vitro, primary mouse astrocyte cultures pre-treated with miR-200c inhibitor prior to oxygen/glucose deprivation preserved expression of SIRT1 and MFN2, and decreased reactive oxygen species generation, whereas pre-treatment with miR-200c mimic had opposite effects that could be reversed by co-treatment with SIRT1 activator. These results suggest that miR-200c regulates astrocyte mitochondrial homeostasis via targeting SIRT1, and that CA1 astrocyte mitochondria and SIRT1 represent potential post-injury therapeutic targets to preserve cognitive function in survivors of global cerebral ischemia.
Collapse
Affiliation(s)
- Brian Griffiths
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Lijun Xu
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Xiaoyun Sun
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Majesty Greer
- Howard University College of Medicine, Washington, DC, United States
| | - Isabella Murray
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Creed Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States,*Correspondence: Creed Stary,
| |
Collapse
|
4
|
Zhou Z, Li X, Domel AG, Dennis EL, Georgiadis M, Liu Y, Raymond SJ, Grant G, Kleiven S, Camarillo D, Zeineh M. The Presence of the Temporal Horn Exacerbates the Vulnerability of Hippocampus During Head Impacts. Front Bioeng Biotechnol 2022; 10:754344. [PMID: 35392406 PMCID: PMC8980591 DOI: 10.3389/fbioe.2022.754344] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Hippocampal injury is common in traumatic brain injury (TBI) patients, but the underlying pathogenesis remains elusive. In this study, we hypothesize that the presence of the adjacent fluid-containing temporal horn exacerbates the biomechanical vulnerability of the hippocampus. Two finite element models of the human head were used to investigate this hypothesis, one with and one without the temporal horn, and both including a detailed hippocampal subfield delineation. A fluid-structure interaction coupling approach was used to simulate the brain-ventricle interface, in which the intraventricular cerebrospinal fluid was represented by an arbitrary Lagrangian-Eulerian multi-material formation to account for its fluid behavior. By comparing the response of these two models under identical loadings, the model that included the temporal horn predicted increased magnitudes of strain and strain rate in the hippocampus with respect to its counterpart without the temporal horn. This specifically affected cornu ammonis (CA) 1 (CA1), CA2/3, hippocampal tail, subiculum, and the adjacent amygdala and ventral diencephalon. These computational results suggest that the presence of the temporal horn exacerbate the vulnerability of the hippocampus, highlighting the mechanobiological dependency of the hippocampus on the temporal horn.
Collapse
Affiliation(s)
- Zhou Zhou
- Department of Bioengineering, Stanford University, Stanford, CA, United States
- Neuronic Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Xiaogai Li
- Neuronic Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - August G. Domel
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Emily L. Dennis
- TBI and Concussion Center, Department of Neurology, University of Utah, Salt Lake City, UT, United States
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Marios Georgiadis
- Department of Radiology, Stanford University, Stanford, CA, United States
| | - Yuzhe Liu
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Samuel J. Raymond
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Gerald Grant
- Department of Neurosurgery, Stanford University, Stanford, CA, United States
- Department of Neurology, Stanford University, Stanford, CA, United States
| | - Svein Kleiven
- Neuronic Engineering, KTH Royal Institute of Technology, Stockholm, Sweden
| | - David Camarillo
- Department of Bioengineering, Stanford University, Stanford, CA, United States
- Department of Neurosurgery, Stanford University, Stanford, CA, United States
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
| | - Michael Zeineh
- Department of Radiology, Stanford University, Stanford, CA, United States
| |
Collapse
|
5
|
Meyer E, Bonato JM, Mori MA, Mattos BA, Guimarães FS, Milani H, de Campos AC, de Oliveira RMW. Cannabidiol Confers Neuroprotection in Rats in a Model of Transient Global Cerebral Ischemia: Impact of Hippocampal Synaptic Neuroplasticity. Mol Neurobiol 2021; 58:5338-5355. [PMID: 34302281 DOI: 10.1007/s12035-021-02479-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/28/2021] [Indexed: 02/01/2023]
Abstract
Evidence for the clinical use of neuroprotective drugs for the treatment of cerebral ischemia (CI) is still greatly limited. Spatial/temporal disorientation and cognitive dysfunction are among the most prominent long-term sequelae of CI. Cannabidiol (CBD) is a non-psychotomimetic constituent of Cannabis sativa that exerts neuroprotective effects against experimental CI. The present study investigated possible neuroprotective mechanisms of action of CBD on spatial memory impairments that are caused by transient global cerebral ischemia (TGCI) in rats. Hippocampal synaptic plasticity is a fundamental mechanism of learning and memory. Thus, we also evaluated the impact of CBD on neuroplastic changes in the hippocampus after TGCI. Wistar rats were trained to learn an eight-arm aversive radial maze (AvRM) task and underwent either sham or TGCI surgery. The animals received vehicle or 10 mg/kg CBD (i.p.) 30 min before surgery, 3 h after surgery, and then once daily for 14 days. On days 7 and 14, we performed a retention memory test. Another group of rats that received the same pharmacological treatment was tested in the object location test (OLT). Brains were removed and processed to assess neuronal degeneration, synaptic protein levels, and dendritic remodeling in the hippocampus. Cannabidiol treatment attenuated ischemia-induced memory deficits. In rats that were subjected to TGCI, CBD attenuated hippocampal CA1 neurodegeneration and increased brain-derived neurotrophic factor levels. Additionally, CBD protected neurons against the deleterious effects of TGCI on dendritic spine number and the length of dendritic arborization. These results suggest that the neuroprotective effects of CBD against TGCI-induced memory impairments involve changes in synaptic plasticity in the hippocampus.
Collapse
Affiliation(s)
- Erika Meyer
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, Maringá, Paraná, 5790, 87020-900, Brazil
| | - Jéssica Mendes Bonato
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, Maringá, Paraná, 5790, 87020-900, Brazil
| | - Marco Aurélio Mori
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, Maringá, Paraná, 5790, 87020-900, Brazil
| | - Bianca Andretto Mattos
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, Maringá, Paraná, 5790, 87020-900, Brazil
| | - Francisco Silveira Guimarães
- Department of Pharmacology, School of Medicine, USP, Av. Bandeirantes, Ribeirão Preto, São Paulo, 14015-000, Brazil
| | - Humberto Milani
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, Maringá, Paraná, 5790, 87020-900, Brazil
| | - Alline Cristina de Campos
- Department of Pharmacology, School of Medicine, USP, Av. Bandeirantes, Ribeirão Preto, São Paulo, 14015-000, Brazil
| | - Rúbia Maria Weffort de Oliveira
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, Maringá, Paraná, 5790, 87020-900, Brazil.
| |
Collapse
|
6
|
Buonarati OR, Cook SG, Goodell DJ, Chalmers NE, Rumian NL, Tullis JE, Restrepo S, Coultrap SJ, Quillinan N, Herson PS, Bayer KU. CaMKII versus DAPK1 Binding to GluN2B in Ischemic Neuronal Cell Death after Resuscitation from Cardiac Arrest. Cell Rep 2021; 30:1-8.e4. [PMID: 31914378 PMCID: PMC6959131 DOI: 10.1016/j.celrep.2019.11.076] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/25/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
DAPK1 binding to GluN2B was prominently reported to mediate ischemic cell death in vivo. DAPK1 and CaMKII bind to the same GluN2B region, and their binding is mutually exclusive. Here, we show that mutating the binding region on GluN2B (L1298A/ R1300Q) protected against neuronal cell death induced by cardiac arrest followed by resuscitation. Importantly, the GluN2B mutation selectively abolished only CaMKII, but not DAPK1, binding. During ischemic or excitotoxic insults, CaMKII further accumulated at excitatory synapses, and this accumulation was mediated by GluN2B binding. Interestingly, extra-synaptic GluN2B decreased after ischemia, but its relative association with DAPK1 increased. Thus, ischemic neuronal death requires CaMKII binding to synaptic GluN2B, whereas any potential role for DAPK1 binding is restricted to a different, likely extra-synaptic population of GluN2B. Ischemic insults cause excitotoxic neuronal cell death via NMDA receptor overstimulation. Buonarati et al. find that excitotoxic insults cause DAPK1 movement to extra-synaptic NMDA receptors and CaMKII movement to synaptic NMDA receptors; importantly, preventing this CaMKII movement protects neurons from ischemic death.
Collapse
Affiliation(s)
- Olivia R Buonarati
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah G Cook
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dayton J Goodell
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nicholas E Chalmers
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nicole L Rumian
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jonathan E Tullis
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Susana Restrepo
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Steven J Coultrap
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Nidia Quillinan
- Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paco S Herson
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; Program in Neuroscience, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
7
|
Ahn JH, Lee TK, Tae HJ, Kim B, Sim H, Lee JC, Kim DW, Kim YS, Shin MC, Park Y, Cho JH, Park JH, Lee CH, Choi SY, Won MH. Neuronal Death in the CNS Autonomic Control Center Comes Very Early after Cardiac Arrest and Is Not Significantly Attenuated by Prompt Hypothermic Treatment in Rats. Cells 2021; 10:E60. [PMID: 33401719 PMCID: PMC7824613 DOI: 10.3390/cells10010060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
Autonomic dysfunction in the central nervous system (CNS) can cause death after recovery from a cardiac arrest (CA). However, few studies on histopathological changes in animal models of CA have been reported. In this study, we investigated the prevalence of neuronal death and damage in various brain regions and the spinal cord at early times after asphyxial CA and we studied the relationship between the mortality rate and neuronal damage following hypothermic treatment after CA. Rats were subjected to 7-8 min of asphyxial CA, followed by resuscitation and prompt hypothermic treatment. Eight regions related to autonomic control (the cingulate cortex, hippocampus, thalamus, hypothalamus, myelencephalon, and spinal cord) were examined using cresyl violet (a marker for Nissl substance) and Fluoro-Jade B (a marker for neuronal death). The survival rate was 44.5% 1 day post-CA, 18.2% 2 days post-CA and 0% 5 days post-CA. Neuronal death started 12 h post-CA in the gigantocellular reticular nucleus and caudoventrolateral reticular nucleus in the myelencephalon and lamina VII in the cervical, thoracic, lumbar, and sacral spinal cord, of which neurons are related to autonomic lower motor neurons. In these regions, Iba-1 immunoreactivity indicating microglial activation (microgliosis) was gradually increased with time after CA. Prompt hypothermic treatment increased the survival rate at 5 days after CA with an attenuation of neuronal damages and death in the damaged regions. However, the survival rate was 0% at 12 days after CA. Taken together, our study suggests that the early damage and death of neurons related to autonomic lower motor neurons was significantly related to the high mortality rate after CA and that prompt hypothermic therapy could increase the survival rate temporarily after CA, but could not ultimately save the animal.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan 50510, Korea;
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea;
| | - Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Korea;
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Hyejin Sim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung 25457, Korea;
| | - Yoon Sung Kim
- Department of Emergency Medicine, Samcheok Medical Center, Samcheok 25920, Korea;
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (M.C.S.); (Y.P.); (J.H.C.)
| | - Yoonsoo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (M.C.S.); (Y.P.); (J.H.C.)
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (M.C.S.); (Y.P.); (J.H.C.)
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju 38066, Korea;
| | - Choong-Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Korea;
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea;
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea; (B.K.); (H.S.); (J.-C.L.)
| |
Collapse
|
8
|
Butt UJ, Steixner-Kumar AA, Depp C, Sun T, Hassouna I, Wüstefeld L, Arinrad S, Zillmann MR, Schopf N, Fernandez Garcia-Agudo L, Mohrmann L, Bode U, Ronnenberg A, Hindermann M, Goebbels S, Bonn S, Katschinski DM, Miskowiak KW, Nave KA, Ehrenreich H. Hippocampal neurons respond to brain activity with functional hypoxia. Mol Psychiatry 2021; 26:1790-1807. [PMID: 33564132 PMCID: PMC8440186 DOI: 10.1038/s41380-020-00988-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
Physical activity and cognitive challenge are established non-invasive methods to induce comprehensive brain activation and thereby improve global brain function including mood and emotional well-being in healthy subjects and in patients. However, the mechanisms underlying this experimental and clinical observation and broadly exploited therapeutic tool are still widely obscure. Here we show in the behaving brain that physiological (endogenous) hypoxia is likely a respective lead mechanism, regulating hippocampal plasticity via adaptive gene expression. A refined transgenic approach in mice, utilizing the oxygen-dependent degradation (ODD) domain of HIF-1α fused to CreERT2 recombinase, allows us to demonstrate hypoxic cells in the performing brain under normoxia and motor-cognitive challenge, and spatially map them by light-sheet microscopy, all in comparison to inspiratory hypoxia as strong positive control. We report that a complex motor-cognitive challenge causes hypoxia across essentially all brain areas, with hypoxic neurons particularly abundant in the hippocampus. These data suggest an intriguing model of neuroplasticity, in which a specific task-associated neuronal activity triggers mild hypoxia as a local neuron-specific as well as a brain-wide response, comprising indirectly activated neurons and non-neuronal cells.
Collapse
Affiliation(s)
- Umer Javed Butt
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Agnes A. Steixner-Kumar
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Constanze Depp
- grid.419522.90000 0001 0668 6902Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Ting Sun
- grid.419522.90000 0001 0668 6902Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany ,grid.13648.380000 0001 2180 3484Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Imam Hassouna
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Liane Wüstefeld
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Sahab Arinrad
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Matthias R. Zillmann
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Nadine Schopf
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Laura Fernandez Garcia-Agudo
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Leonie Mohrmann
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Ulli Bode
- grid.419522.90000 0001 0668 6902Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Anja Ronnenberg
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Martin Hindermann
- grid.419522.90000 0001 0668 6902Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Sandra Goebbels
- grid.419522.90000 0001 0668 6902Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Stefan Bonn
- grid.13648.380000 0001 2180 3484Institute of Medical Systems Biology, Center for Molecular Neurobiology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Dörthe M. Katschinski
- grid.7450.60000 0001 2364 4210Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Göttingen, Germany
| | - Kamilla W. Miskowiak
- grid.475435.4Psychiatric Centre Copenhagen, University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| |
Collapse
|
9
|
Chalmers NE, Yonchek J, Steklac KE, Ramsey M, Bayer KU, Herson PS, Quillinan N. Calcium/Calmodulin-Dependent Kinase (CaMKII) Inhibition Protects Against Purkinje Cell Damage Following CA/CPR in Mice. Mol Neurobiol 2020; 57:150-158. [PMID: 31520314 PMCID: PMC6980452 DOI: 10.1007/s12035-019-01765-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 01/14/2023]
Abstract
Ischemic brain damage is triggered by glutamate excitotoxicity resulting in neuronal cell death. Previous research has demonstrated that N-methly-D-aspartate (NMDA) receptor activation triggers downstream calcium-dependent signaling pathways, specifically Ca2+/calmodulin-dependent protein kinase II (CaMKII). Inhibiting CaMKII is protective against hippocampal ischemic injury, but there is little known about its role in the cerebellum. To examine the neuroprotective potential of CaMKII inhibition in Purkinje cells, we subjected C57BL/6 or CaMKIIα KO male mice (8-12 weeks old) to cardiac arrest followed by cardiopulmonary resuscitation (CA/CPR). We performed a dose-response study for tat-CN19o and cerebellar injury was analyzed at 7 days after CA/CPR. Acute signaling was assessed at 6 h after CA/CPR using Western blot analysis. We observed increased phosphorylation of the T286 residue of CaMKII, suggesting increased autonomous activation. Analysis of Purkinje cell density revealed a decrease in cell density at 7 days after CA/CPR that was prevented with tat-CN19o at doses of 0.1 and 1 mg/kg. However, neuroprotection in the cerebellum required doses that were 10-fold higher than what was needed in the hippocampus. CaMKIIα KO mice subjected to sham surgery or CA/CPR had similar Purkinje cell densities, suggesting CaMKIIα is required for CA/CPR-induced injury in the cerebellum. We also observed a CA/CPR-induced activation of death-associated protein kinase (DAPK1) that tat-CN19o did not block. In summary, our findings indicate that inhibition of autonomous CaMKII activity is a promising therapeutic approach that is effective across multiple brain regions.
Collapse
Affiliation(s)
- Nicholas E Chalmers
- Neuronal Injury Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Joan Yonchek
- Neuronal Injury Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Kathryn E Steklac
- Neuronal Injury Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew Ramsey
- Neuronal Injury Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Paco S Herson
- Neuronal Injury Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Nidia Quillinan
- Neuronal Injury Program, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
- Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
10
|
Cardiac Arrest Induces Ischemic Long-Term Potentiation of Hippocampal CA1 Neurons That Occludes Physiological Long-Term Potentiation. Neural Plast 2018; 2018:9275239. [PMID: 29853851 PMCID: PMC5944194 DOI: 10.1155/2018/9275239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/15/2018] [Accepted: 04/05/2018] [Indexed: 01/31/2023] Open
Abstract
Ischemic long-term potentiation (iLTP) is a form of synaptic plasticity that occurs in acute brain slices following oxygen-glucose deprivation. In vitro, iLTP can occlude physiological LTP (pLTP) through saturation of plasticity mechanisms. We used our murine cardiac arrest and cardiopulmonary resuscitation (CA/CPR) model to produce global brain ischemia and assess whether iLTP is induced in vivo, contributing to the functionally relevant impairment of pLTP. Adult male mice were subjected to CA/CPR, and slice electrophysiology was performed in the hippocampal CA1 region 7 or 30 days later. We observed increased miniature excitatory postsynaptic current amplitudes, suggesting a potentiation of postsynaptic AMPA receptor function after CA/CPR. We also observed increased phosphorylated GluR1 in the postsynaptic density of hippocampi after CA/CPR. These data support the in vivo induction of ischemia-induced plasticity. Application of a low-frequency stimulus (LFS) to CA1 inputs reduced excitatory postsynaptic potentials in slices from mice subjected to CA/CPR, while having no effects in sham controls. These results are consistent with a reversal, or depotentiation, of iLTP. Further, depotentiation with LFS partially restored induction of pLTP with theta burst stimulation. These data provide evidence for iLTP following in vivo ischemia, which occludes pLTP and likely contributes to network disruptions that underlie memory impairments.
Collapse
|
11
|
Quillinan N, Deng G, Shimizu K, Cruz-Torres I, Schroeder C, Traystman RJ, Herson PS. Long-term depression in Purkinje neurons is persistently impaired following cardiac arrest and cardiopulmonary resuscitation in mice. J Cereb Blood Flow Metab 2017; 37:3053-3064. [PMID: 28168893 PMCID: PMC5536809 DOI: 10.1177/0271678x16683691] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cardiac arrest and cardiopulmonary resuscitation (CA/CPR) produce brain ischemia that results in cognitive and motor coordination impairments subsequent to injury of vulnerable populations of neurons, including cerebellar Purkinje neurons. To determine the effects of CA/CPR on plasticity in the cerebellum, we used whole cell recordings from Purkinje neurons to examine long-term depression (LTD) at parallel fiber (PF) synapses. Acute slices were prepared from adult male mice subjected to 8 min cardiac arrest at 1, 7, and 30 days after resuscitation. Concurrent stimulation of PF and climbing fibers (CFs) resulted in robust LTD of PF-evoked excitatory postsynaptic currents (EPSCs) in controls. LTD was absent in recordings obtained from mice subjected to CA/CPR, with no change in EPSC amplitude from baseline at any time point tested. AMPA and mGluR-mediated responses at the PF were not altered by CA/CPR. In contrast, CF-evoked NMDA currents were reduced following CA/CPR, which could account for the loss of LTD observed. A loss of GluN1 protein was observed following CA/CPR that was surprisingly not associated with changes in mRNA expression. These data demonstrate sustained impairments in synaptic plasticity in Purkinje neurons that survive the initial injury and which likely contribute to motor coordination impairments observed after CA/CPR.
Collapse
Affiliation(s)
- Nidia Quillinan
- 1 Neuronal Injury Program, Department of Anesthesiology, University of Colorado, Aurora, CO, USA
| | - Guiying Deng
- 2 Department of Pharmacology, University of Colorado, Aurora, CO, USA
| | - Kaori Shimizu
- 1 Neuronal Injury Program, Department of Anesthesiology, University of Colorado, Aurora, CO, USA
| | | | - Christian Schroeder
- 1 Neuronal Injury Program, Department of Anesthesiology, University of Colorado, Aurora, CO, USA
| | - Richard J Traystman
- 1 Neuronal Injury Program, Department of Anesthesiology, University of Colorado, Aurora, CO, USA.,2 Department of Pharmacology, University of Colorado, Aurora, CO, USA
| | - Paco S Herson
- 1 Neuronal Injury Program, Department of Anesthesiology, University of Colorado, Aurora, CO, USA.,2 Department of Pharmacology, University of Colorado, Aurora, CO, USA
| |
Collapse
|
12
|
Lu Q, Tucker D, Dong Y, Zhao N, Zhang Q. Neuroprotective and Functional Improvement Effects of Methylene Blue in Global Cerebral Ischemia. Mol Neurobiol 2015; 53:5344-55. [PMID: 26433378 DOI: 10.1007/s12035-015-9455-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/25/2015] [Indexed: 11/25/2022]
Abstract
Transient global cerebral ischemia (GCI) causes delayed neuronal cell death in the vulnerable hippocampus CA1 subfield, as well as behavioral deficits. Ischemia reperfusion (I/R) produces excessive reactive oxygen species and plays a key role in brain injury. The mitochondrial electron respiratory chain is the main cellular source of free radical generation, and dysfunction of mitochondria has a significant impact on the neuronal cell death in ischemic brain. The aim of the present study is to investigate the potential beneficial effects of methylene blue (MB) in a four-vessel occlusion (4VO) GCI model on adult male rats. MB was delivered at a dose of 0.5 mg/kg/day for 7 days, through a mini-pump implanted subcutaneously after GCI. We first found that MB significantly improved ischemic neuronal survival in the hippocampal CA1 region as measured by cresyl violet staining as well as NeuN staining. We also found that MB has the ability to rescue ischemia-induced decreases of cytochrome c oxidase activity and ATP generation in the CA1 region following I/R. Further analysis with labeling of MitoTracker® Red revealed that the depolarization of mitochondrial membrane potential (MMP) was markedly attenuated following MB treatment. In addition, the induction of caspase-3, caspase-8, and caspase-9 activities and the increased numbers of TUNEL-positive cells of the CA1 region were significantly reduced by MB application. Correspondingly, Barnes maze tests showed that the deterioration of spatial learning and memory performance following GCI was significantly improved in the MB-treatment group compared to the ischemic control group. In summary, our study suggests that MB may be a promising therapeutic agent targeting neuronal cell death and cognitive deficits following transient global cerebral ischemia.
Collapse
Affiliation(s)
- Qing Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA
| | - Donovan Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA
| | - Ningjun Zhao
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regent University, 1120 15th Street, CA3050, Augusta, GA, 30912, USA.
| |
Collapse
|
13
|
Region-specific role for GluN2B-containing NMDA receptors in injury to Purkinje cells and CA1 neurons following global cerebral ischemia. Neuroscience 2014; 284:555-565. [PMID: 25450957 DOI: 10.1016/j.neuroscience.2014.10.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 01/04/2023]
Abstract
Motor deficits are present in cardiac arrest survivors and injury to cerebellar Purkinje cells (PCs) likely contribute to impairments in motor coordination and post-hypoxic myoclonus. N-Methyl-D-aspartic acid (NMDA) receptor-mediated excitotoxicity is a well-established mechanism of cell death in several brain regions, but the role of NMDA receptors in PC injury remains understudied. Emerging data in cortical and hippocampal neurons indicate that the GluN2A-containing NMDA receptors signal to improve cell survival and GluN2B-containing receptors contribute to neuronal injury. This study compared neuronal injury in the hippocampal CA1 region to that in PCs and investigated the role of NMDA receptors in PC injury in our mouse model of cardiac arrest and cardiopulmonary resuscitation (CA/CPR). Analysis of cell density demonstrated a 24% loss of PCs within 24 h after 8 min CA/CPR and injury stabilized to 33% by 7 days. The subunit promiscuous NMDA receptor antagonist MK-801 protected both CA1 neurons and PCs from ischemic injury following CA/CPR, demonstrating a role for NMDA receptor activation in injury to both brain regions. In contrast, the GluN2B antagonist, Co 101244, had no effect on PC loss while protecting against injury in the CA1 region. These data indicate that ischemic injury to cerebellar PCs progresses via different cell death mechanisms compared to hippocampal CA1 neurons.
Collapse
|
14
|
Maldonado JR. Neuropathogenesis of delirium: review of current etiologic theories and common pathways. Am J Geriatr Psychiatry 2013; 21:1190-222. [PMID: 24206937 DOI: 10.1016/j.jagp.2013.09.005] [Citation(s) in RCA: 443] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 09/10/2013] [Accepted: 09/13/2013] [Indexed: 12/20/2022]
Abstract
Delirium is a neurobehavioral syndrome caused by dysregulation of neuronal activity secondary to systemic disturbances. Over time, a number of theories have been proposed in an attempt to explain the processes leading to the development of delirium. Each proposed theory has focused on a specific mechanism or pathologic process (e.g., dopamine excess or acetylcholine deficiency theories), observational and experiential evidence (e.g., sleep deprivation, aging), or empirical data (e.g., specific pharmacologic agents' association with postoperative delirium, intraoperative hypoxia). This article represents a review of published literature and summarizes the top seven proposed theories and their interrelation. This review includes the "neuroinflammatory," "neuronal aging," "oxidative stress," "neurotransmitter deficiency," "neuroendocrine," "diurnal dysregulation," and "network disconnectivity" hypotheses. Most of these theories are complementary, rather than competing, with many areas of intersection and reciprocal influence. The literature suggests that many factors or mechanisms included in these theories lead to a final common outcome associated with an alteration in neurotransmitter synthesis, function, and/or availability that mediates the complex behavioral and cognitive changes observed in delirium. In general, the most commonly described neurotransmitter changes associated with delirium include deficiencies in acetylcholine and/or melatonin availability; excess in dopamine, norepinephrine, and/or glutamate release; and variable alterations (e.g., either a decreased or increased activity, depending on delirium presentation and cause) in serotonin, histamine, and/or γ-aminobutyric acid. In the end, it is unlikely that any one of these theories is fully capable of explaining the etiology or phenomenologic manifestations of delirium but rather that two or more of these, if not all, act together to lead to the biochemical derangement and, ultimately, to the complex cognitive and behavioral changes characteristic of delirium.
Collapse
Affiliation(s)
- José R Maldonado
- Departments of Psychiatry, Internal Medicine & Surgery and the Psychosomatic Medicine Service, Stanford University School of Medicine, and Board of Directors, American Delirium Society, Stanford, CA.
| |
Collapse
|
15
|
Paine MG, Che D, Li L, Neumar RW. Cerebellar Purkinje cell neurodegeneration after cardiac arrest: Effect of therapeutic hypothermia. Resuscitation 2012; 83:1511-6. [DOI: 10.1016/j.resuscitation.2012.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 05/06/2012] [Accepted: 05/21/2012] [Indexed: 12/31/2022]
|
16
|
Thalamocortical dysfunction and thalamic injury after asphyxial cardiac arrest in developing rats. J Neurosci 2012; 32:4972-81. [PMID: 22492052 DOI: 10.1523/jneurosci.5597-11.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Global hypoxia-ischemia interrupts oxygen delivery and blood flow to the entire brain. Previous studies of global brain hypoxia-ischemia have primarily focused on injury to the cerebral cortex and to the hippocampus. Susceptible neuronal populations also include inhibitory neurons in the thalamic reticular nucleus. We therefore investigated the impact of global brain hypoxia-ischemia on the thalamic circuit function in the somatosensory system of young rats. We used single neuron recordings and controlled whisker deflections to examine responses of thalamocortical neurons to sensory stimulation in rat survivors of 9 min of asphyxial cardiac arrest incurred on postnatal day 17. We found that 48-72 h after cardiac arrest, thalamocortical neurons demonstrate significantly elevated firing rates both during spontaneous activity and in response to whisker deflections. The elevated evoked firing rates persist for at least 6-8 weeks after injury. Despite the overall increase in firing, by 6 weeks, thalamocortical neurons display degraded receptive fields, with decreased responses to adjacent whiskers. Nine minutes of asphyxial cardiac arrest was associated with extensive degeneration of neurites in the somatosensory nucleus as well as activation of microglia in the reticular nucleus. Global brain hypoxia-ischemia during cardiac arrest has a long-term impact on processing and transfer of sensory information by thalamic circuitry. Thalamic circuitry and normalization of its function may represent a distinct therapeutic target after cardiac arrest.
Collapse
|
17
|
Warren DE, Duff MC, Magnotta V, Capizzano AA, Cassell MD, Tranel D. Long-term neuropsychological, neuroanatomical, and life outcome in hippocampal amnesia. Clin Neuropsychol 2012; 26:335-69. [PMID: 22401298 DOI: 10.1080/13854046.2012.655781] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Focal bilateral hippocampal damage typically causes severe and selective amnesia for new declarative information (facts and events), a cognitive deficit that greatly impacts the ability to live a normal, fully independent life. We describe the case of 1846, a 48-year-old woman with profound hippocampal amnesia following status epilepticus and an associated anoxic episode at age 30. Patient 1846 has undergone extensive neuropsychological testing on many occasions over the 18 years since her injury, and we present data indicating that her memory impairment has remained severe and stable during that time. New, high-resolution, structural MRI studies of 1846's brain reveal substantial bilateral hippocampal atrophy resembling that of other well-known amnesic patients. In spite of severe amnesia 1846 lives a full and mostly independent adult life, facilitated by an extensive social support network of family and friends. Her case provides an example of a rare and unlikely positive outcome in the face of severe memory problems.
Collapse
Affiliation(s)
- David E Warren
- Department of Neurology, University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Bolkvadze T, Pitkänen A. Development of post-traumatic epilepsy after controlled cortical impact and lateral fluid-percussion-induced brain injury in the mouse. J Neurotrauma 2012; 29:789-812. [PMID: 22023672 DOI: 10.1089/neu.2011.1954] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The present study investigated the development of hyperexcitability and epilepsy in mice with traumatic brain injury (TBI) induced by controlled cortical impact (CCI) or lateral fluid-percussion injury (FPI), which are the two most commonly used experimental models of human TBI in rodents. TBI was induced with CCI to 50 (14 controls) and with lateral FPI to 45 (15 controls) C57BL/6S adult male mice. The animals were followed-up for 9 months, including three 2-week periods of continuous video-electroencephalographic (EEG) monitoring, and a seizure susceptibility test with pentylenetetrazol (PTZ). In the end, the animals were perfusion-fixed for histology. The experiment included two independent cohorts of animals. Late post-traumatic spontaneous electrographic seizures were detected in 9% of mice after CCI and 3% after lateral FPI. Eighty-two percent of mice after CCI and 71% after lateral FPI had spontaneous epileptiform spiking on EEG. In addition, 58% of mice with lateral FPI showed spontaneous epileptiform discharges. A PTZ test demonstrated increased seizure susceptibility in the majority of mice in both models, compared to control mice. There was no further progression in the occurrence of epilepsy or epileptiform spiking when follow-up was extended from 6 to 9 months. The severity of cortical or hippocampal damage did not differentiate mice with or without epileptiform activity in either model. Finally, two independent series of experiments in both injury models provided comparable data demonstrating reproducibility of the modeling. These data show that different types of impact can trigger epileptogenesis in mice. Even though the frequency of spontaneous seizures in C57BL/6S mice is low, a large majority of animals develop hyperexcitability.
Collapse
Affiliation(s)
- Tamuna Bolkvadze
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | |
Collapse
|
19
|
Hempel R, Onopa R, Convit A. Type 2 diabetes affects hippocampus volume differentially in men and women. Diabetes Metab Res Rev 2012; 28:76-83. [PMID: 21695767 PMCID: PMC3273865 DOI: 10.1002/dmrr.1230] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) has been shown to result in medical complications on several organ systems including the kidneys, eyes, cardiovascular system, and most recently described the brain, including the hippocampus. There is also evidence that females are disproportionately affected by these medical complications. Brain volume reductions have also been associated with chronic low-grade inflammation and dyslipidaemia. This study investigated the relationships among T2DM, gender, inflammation, dyslipidaemia, and hippocampal volumes. METHOD Participant groups consisted of 40 obese adults with T2DM and 47 lean adults, group-matched on age, gender, race, and education. Each participant underwent medical examination including a standard panel of blood tests, a magnetic resonance imaging, and cognitive evaluation. RESULTS We show that there is a gender difference in the association of T2DM and hippocampal volumes: diabetic women are most affected despite having better glucose control than their male counterparts. Although females with T2DM had disproportionately lower high density lipoprotein as well as better haemoglobin A1c, neither of these results explained why females with T2DM had the smallest hippocampal volumes. CONCLUSIONS These important findings indicate that in addition to the higher rate of traditional medical complication, females with T2DM are likely to suffer more brain complications than males. These observations, if supported by larger studies, suggest that in the future gender could be considered when customizing diabetes treatment.
Collapse
Affiliation(s)
- R Hempel
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
20
|
Nelson PT, Schmitt FA, Lin Y, Abner EL, Jicha GA, Patel E, Thomason PC, Neltner JH, Smith CD, Santacruz KS, Sonnen JA, Poon LW, Gearing M, Green RC, Woodard JL, Van Eldik LJ, Kryscio RJ. Hippocampal sclerosis in advanced age: clinical and pathological features. ACTA ACUST UNITED AC 2011; 134:1506-18. [PMID: 21596774 DOI: 10.1093/brain/awr053] [Citation(s) in RCA: 212] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hippocampal sclerosis is a relatively common neuropathological finding (∼10% of individuals over the age of 85 years) characterized by cell loss and gliosis in the hippocampus that is not explained by Alzheimer's disease. Hippocampal sclerosis pathology can be associated with different underlying causes, and we refer to hippocampal sclerosis in the aged brain as hippocampal sclerosis associated with ageing. Much remains unknown about hippocampal sclerosis associated with ageing. We combined three different large autopsy cohorts: University of Kentucky Alzheimer's Disease Centre, the Nun Study and the Georgia Centenarian Study to obtain a pool of 1110 patients, all of whom were evaluated neuropathologically at the University of Kentucky. We focused on the subset of cases with neuropathology-confirmed hippocampal sclerosis (n=106). For individuals aged≥95 years at death (n=179 in our sample), each year of life beyond the age of 95 years correlated with increased prevalence of hippocampal sclerosis pathology and decreased prevalence of 'definite' Alzheimer's disease pathology. Aberrant TAR DNA protein 43 immunohistochemistry was seen in 89.9% of hippocampal sclerosis positive patients compared with 9.7% of hippocampal sclerosis negative patients. TAR DNA protein 43 immunohistochemistry can be used to demonstrate that the disease is usually bilateral even when hippocampal sclerosis pathology is not obvious by haematoxylin and eosin stains. TAR DNA protein 43 immunohistochemistry was negative on brain sections from younger individuals (n=10) after hippocampectomy due to seizures, who had pathologically confirmed hippocampal sclerosis. There was no association between cases with hippocampal sclerosis associated with ageing and apolipoprotein E genotype. Age of death and clinical features of hippocampal sclerosis associated with ageing (with or without aberrant TAR DNA protein 43) were distinct from previously published cases of frontotemporal lobar degeneration TAR DNA protein 43. To help sharpen our ability to discriminate patients with hippocampal sclerosis associated with ageing clinically, the longitudinal cognitive profile of 43 patients with hippocampal sclerosis associated with ageing was compared with the profiles of 75 controls matched for age, gender, education level and apolipoprotein E genotype. These individuals were followed from intake assessment, with 8.2 (average) longitudinal cognitive assessments. A neuropsychological profile with relatively high-verbal fluency but low word list recall distinguished the hippocampal sclerosis associated with ageing group at intake (P<0.015) and also 5.5-6.5 years before death (P<0.005). This may provide a first step in clinical differentiation of hippocampal sclerosis associated with ageing versus pure Alzheimer's disease in their earliest stages. In summary, in the largest series of autopsy-verified patients with hippocampal sclerosis to date, we characterized the clinical and pathological features associated with hippocampal sclerosis associated with ageing.
Collapse
Affiliation(s)
- Peter T Nelson
- Department of Pathology, Division of Neuropathology and the Sanders-Brown Centre on Ageing, University of Kentucky, 800 S. Limestone, Lexington, KY 40536-0230, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Normoxic versus hyperoxic resuscitation in pediatric asphyxial cardiac arrest: effects on oxidative stress. Crit Care Med 2011; 39:335-43. [PMID: 21057313 DOI: 10.1097/ccm.0b013e3181ffda0e] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To determine the effects of normoxic vs. hyperoxic resuscitation on oxidative stress in a model of pediatric asphyxial cardiac arrest. DESIGN Prospective, interventional study. SETTING University research laboratory. SUBJECTS Postnatal day 16-18 rats (n = 5 per group). INTERVENTIONS Rats underwent asphyxial cardiac arrest for 9 min. Rats were randomized to receive 100% oxygen, room air, or 100% oxygen with polynitroxyl albumin (10 mL·kg⁻¹ intravenously, 0 and 30 min after resuscitation) for 1 hr from the start of cardiopulmonary resuscitation. Shams recovered in 100% oxygen or room air after surgery. MEASUREMENTS AND MAIN RESULTS Physiological variables were recorded at baseline to 1 hr after resuscitation. At 6 hrs after asphyxial cardiac arrest, levels of reduced glutathione and protein-thiols (fluorescent assay), activities of total superoxide dismutase and mitochondrial manganese superoxide dismutase (cytochrome c reduction method), manganese superoxide dismutase expression (Western blot), and lipid peroxidation (4-hydroxynonenal Michael adducts) were evaluated in brain tissue homogenates. Hippocampal 3-nitrotyrosine levels were determined by immunohistochemistry 72 hrs after asphyxial cardiac arrest. Survival did not differ among groups. At 1 hr after resuscitation, Pao2, pH, and mean arterial pressure were decreased in room air vs. 100% oxygen rats (59 ± 3 vs. 465 ± 46 mm Hg, 7.36 ± 0.05 vs. 7.42 ± 0.03, 35 ± 4 vs. 45 ± 5 mm Hg; p < .05). Rats resuscitated with 100% oxygen had decreased hippocampal reduced glutathione levels vs. sham (15.3 ± 0.4 vs. 20.9 ± 4.1 nmol·mg protein⁻¹; p < .01). Hippocampal manganese superoxide dismutase activity was significantly increased in 100% oxygen rats vs. sham (14 ± 2.4 vs. 9.5 ± 1.6 units·mg protein⁻¹, p < .01), with no difference in protein expression of manganese superoxide dismutase. Room air and 100% oxygen plus polynitroxyl albumin groups had hippocampal reduced glutathione and manganese superoxide dismutase activity levels comparable with sham. Protein thiol levels were unchanged across groups. Compared with all other groups, rats receiving 100% oxygen had increased immunopositivity for 3-nitrotyrosine in the hippocampus and increased lipid peroxidation in the cortex. CONCLUSIONS Resuscitation with 100% oxygen leads to increased oxidative stress in a model that mimics pediatric cardiac arrest. This may be prevented by using room air or giving an antioxidant with 100% oxygen resuscitation.
Collapse
|
22
|
Distribution of neuropathological lesions in pig brains after different durations of cardiac arrest. Resuscitation 2010; 81:1577-83. [PMID: 20727661 DOI: 10.1016/j.resuscitation.2010.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 07/14/2010] [Accepted: 07/14/2010] [Indexed: 11/21/2022]
Abstract
AIM OF THE STUDY To evaluate all brain regions reported to be selectively vulnerable to global ischaemia in a pig cardiac arrest model with different durations of no-flow by establishing a semi-quantitative brain histopathologic scoring system and to compare histological damage with neurological deficits. METHODS In a prospective randomised laboratory investigation, 35 female Large White pigs weighing 35-45 kg underwent ventricular fibrillation cardiac arrest for 0, 7, 10 or 13 min. In the brains of all animals that survived until the final endpoint (72 h post-arrest), 22 distinct regions were evaluated on paraffin-embedded sections in terms of type and extent of lesions. The results of the histological examination were compared to the results of a neurological outcome evaluation after 72 h. RESULTS Significant differences were found in all cortex regions, the caudate nucleus and putamen, the hippocampal formation, the cerebellar cortex, and the thalamus between the ischaemic groups (7- and 10-min groups) and the control group (0-min group). No 13-min group animal survived. The main findings were neuronal necrosis and oedema. In animals from the 10-min group, many neurons were reabsorbed in the cerebral cortex, caudate nucleus and cerebellar granule cell layer. There was a highly significant correlation between histological damage and neurological deficits. CONCLUSIONS The pattern of neuronal lesions in this pig model bear good resemblance to the pattern known in humans and other animal models. The amount of histological lesions in selectively vulnerable brain regions correlates to neurological outcome.
Collapse
|
23
|
Peskine A, Rosso C, Picq C, Caron E, Pradat-Diehl P. Neurological sequelae after cerebral anoxia. Brain Inj 2010; 24:755-61. [DOI: 10.3109/02699051003709581] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
Abstract
OBJECTIVES To describe the sequelae of cerebral anoxia following out-of-hospital cardiac arrest, to study the functional outcome and to seek a link between the acute stage and the disability. METHOD A retrospective study was performed. The initial findings and the neurological and neuropsychological status are recorded of 12 patients admitted to the rehabilitation unit for after-effects of cerebral anoxia following out-of-hospital cardiac arrest. RESULTS After clinical and neuropsychological assessment, all patients displayed cognitive impairment. Two groups of patients appeared: seven patients were severely disabled with a dysexecutive and behavioural frontal lobe syndrome and memory deficit; five out of the seven also presented an extra-pyramidal syndrome; the other five patients presented behavioural dysfunction related to frontal lobe disorder but were independent in daily life activities. No correlation was found between the acute stage data and the outcome. CONCLUSION Neurological and neuropsychological impairment after cerebral anoxia may be severe but seems difficult to predict. A dysexecutive syndrome was noted in all 12 patients.
Collapse
Affiliation(s)
- Anne Peskine
- Department of Physical Medicine and Rehabilitation, Hospital Pitié Salpêtrière, Paris, France
| | | | | |
Collapse
|
25
|
Pathoetiological model of delirium: a comprehensive understanding of the neurobiology of delirium and an evidence-based approach to prevention and treatment. Crit Care Clin 2008; 24:789-856, ix. [PMID: 18929943 DOI: 10.1016/j.ccc.2008.06.004] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Delirium is the most common complication found in the general hospital setting. Yet, we know relatively little about its actual pathophysiology. This article contains a summary of what we know to date and how different proposed intrinsic and external factors may work together or by themselves to elicit the cascade of neurochemical events that leads to the development delirium. Given how devastating delirium can be, it is imperative that we better understand the causes and underlying pathophysiology. Elaborating a pathoetiology-based cohesive model to better grasp the basic mechanisms that mediate this syndrome will serve clinicians well in aspiring to find ways to correct these cascades, instituting rational treatment modalities, and developing effective preventive techniques.
Collapse
|
26
|
Schmidt S, Gawlik V, Hölter SM, Augustin R, Scheepers A, Behrens M, Wurst W, Gailus-Durner V, Fuchs H, Hrabé de Angelis M, Kluge R, Joost HG, Schürmann A. Deletion of glucose transporter GLUT8 in mice increases locomotor activity. Behav Genet 2008; 38:396-406. [PMID: 18461434 PMCID: PMC2480596 DOI: 10.1007/s10519-008-9208-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Accepted: 04/14/2008] [Indexed: 02/07/2023]
Abstract
Transport of glucose into neuronal cells is predominantly mediated by the glucose transporters GLUT1 and GLUT3. In addition, GLUT8 is expressed in some regions of the brain. By in situ hybridization we detected GLUT8-mRNA in hippocampus, thalamus, and cortex. However, its cellular and physiological function is still unknown. Thus, GLUT8 knockout (Slc2a8−/−) mice were used for a screening approach in the modified hole board (mHB) behavioral test to analyze the role of GLUT8 in the central nervous system. Slc2a8−/− mice showed increased mean velocity, total distance traveled and performed more turns in the mHB test. This hyperactivity of Slc2a8−/− mice was confirmed by monitoring locomotor activity in the home cage and voluntary activity in a running wheel. In addition, Slc2a8−/− mice showed increased arousal as indicated by elevated defecation, reduced latency to the first defecation and a tendency to altered grooming. Furthermore, the mHB test gave evidence that Slc2a8−/− mice exhibit a reduced risk assessment because they performed less rearings in an unprotected area and showed significantly reduced latency to stretched body posture. Our data suggest that behavioral alterations of Slc2a8−/− mice are due to dysfunctions in neuronal processes presumably as a consequence of defects in the glucose metabolism.
Collapse
Affiliation(s)
- S Schmidt
- Department of Pharmacology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, Nuthetal, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Fung SJ, Xi MC, Zhang JH, Sampogna S, Yamuy J, Morales FR, Chase MH. Apnea promotes glutamate-induced excitotoxicity in hippocampal neurons. Brain Res 2007; 1179:42-50. [PMID: 17888415 PMCID: PMC3169175 DOI: 10.1016/j.brainres.2007.08.044] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2007] [Revised: 08/13/2007] [Accepted: 08/18/2007] [Indexed: 10/22/2022]
Abstract
Patients with obstructive sleep apnea (OSA) exhibit hippocampal damage and cognitive deficits. To determine the effect of apnea on the synaptic transmission in the hippocampus, we performed electrophysiological studies in an in vivo guinea pig model of OSA. Specifically, we determined the cornu ammonis region 1 (CA1) field excitatory postsynaptic potential (fEPSP) response to cornu ammonis region 3 (CA3) stimulation and examined the presynaptic mechanisms underlying the changes in the fEPSP. Single episodes of apnea resulted in a maximal potentiation of the fEPSPs at 1 to 3 min after the termination of each episode of apnea. The mean amplitude and slope of the post-apneic fEPSP was significantly larger compared with the pre-apneic control. These changes were accompanied by a significant decrease in the paired-pulse facilitation ratio during the post-apneic period compared with the pre-apneic control. The N-methyl-D-aspartate (NMDA) glutamate receptor antagonist MK-801, when applied locally to the CA1 recording site by pressure ejection, blocked the apnea-induced potentiation of the fEPSP. In the experimental animals that were subjected to extended periods of recurrent apnea, CA1 neurons exhibited positive immunoreactivity for fragmented DNA strands, which indicates apoptotic cell death. The present results demonstrate that apnea-induced potentiation of the hippocampal CA1 fEPSP is mediated by an NMDA receptor mechanism. We therefore conclude that recurrent apnea produces abnormally high levels of glutamate that results in the apoptosis of CA1 neurons. We hypothesize that this damage is reflected by the cognitive deficits that are commonly observed in patients with breathing disorders such as OSA.
Collapse
Affiliation(s)
- Simon J Fung
- WebSciences International, 1251 Westwood Blvd., Los Angeles, CA 90024, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Tasker RC. Changes in white matter late after severe traumatic brain injury in childhood. Dev Neurosci 2006; 28:302-8. [PMID: 16943653 DOI: 10.1159/000094156] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2005] [Accepted: 04/04/2006] [Indexed: 11/19/2022] Open
Abstract
Severe traumatic brain injury in childhood, particularly that complicated by raised intracranial pressure, has significant long-term effects on the brain. Since magnetic resonance imaging provides a means of visualizing neuroanatomic structure in exquisite detail, the scope of this review is to revisit the pathology of traumatic brain injury described in recent clinical imaging studies. Acute imaging provides insight into the acute mechanism of focal and diffuse injury. There is some reduction in threshold for white matter pathology in the hemisphere ipsilateral to injury. After injury, there may be long-term effects on white matter architecture and the potential for brain growth. In this context, the pattern of hippocampal rather than parahippocampal gyrus tissue loss provides insight into the likely cause of white matter injury being cerebral hypoperfusion.
Collapse
Affiliation(s)
- Robert C Tasker
- Department of Paediatrics, School of Clinical Medicine, Cambridge University, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
29
|
Abstract
This article reviews the important differences between children and adults suffering brain injury following cardiac arrest. The differences in etiology, pathophysiology, neuronal vulnerability, and repair in the context of the developing brain are reviewed. The available clinical data are reviewed, and selected treatment priori-ties are declared. The article includes a discussion of knowledge gaps and future directions.
Collapse
Affiliation(s)
- Robert W Hickey
- Division of Pediatric Emergency Medicine, Department of Pediatrics, University of Pittsburgh, Children's Hospital of Pittsburgh, 3705 Fifth Avenue, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
30
|
Strudwick SK, Carne C, Gardiner J, Foster JK, Davis EA, Jones TW. Cognitive functioning in children with early onset type 1 diabetes and severe hypoglycemia. J Pediatr 2005; 147:680-5. [PMID: 16291363 DOI: 10.1016/j.jpeds.2005.06.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Revised: 03/25/2005] [Accepted: 06/03/2005] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To investigate whether severe hypoglycemia in young children with early-onset type 1 diabetes (T1DM) is associated with subsequent abnormalities in cognitive status. STUDY DESIGN Recruitment was from a large population-based database of children and adolescents with T1DM. Children and adolescents with early-onset T1DM (<6 years) were eligible for the study. Diabetic individuals (n = 41) with a prospectively documented history of seizure or coma were compared with peers with no history of severe hypoglycemic events (n = 43). A comprehensive test battery of learning and memory was used together with intellectual and behavioral measures. RESULTS No significant group differences were revealed on the intellectual, memory, or behavioral measures. Similarly, those participants with a history of early first seizure did not differ from their peers with no seizures. There were no significant group differences on the memory subtests that were examined given their potential sensitivity to compromised hippocampal function. CONCLUSIONS There was no clear evidence from this cohort that episodes of seizure or coma, even those occurring in very early childhood, resulted in broad cognitive dysfunction, nor was there evidence of specific memory difficulties at the time of testing in children and adolescents with early-onset T1DM.
Collapse
Affiliation(s)
- Susan K Strudwick
- Department of Psychological Medicine, Princess Margaret Hospital, Perth, Australia
| | | | | | | | | | | |
Collapse
|
31
|
Kim M, Yu ZX, Fredholm BB, Rivkees SA. Susceptibility of the developing brain to acute hypoglycemia involving A1 adenosine receptor activation. Am J Physiol Endocrinol Metab 2005; 289:E562-9. [PMID: 16150954 DOI: 10.1152/ajpendo.00112.2005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been suggested that the developing brain is less vulnerable to the adverse effects of hypoglycemia than the mature brain; however, this issue remains controversial. We also do not know the magnitude or duration of hypoglycemia needed to trigger hypoglycemic brain injury during development. To address this issue a series of in vivo and in vitro studies were performed. First, we established an acute model of insulin-induced hypoglycemia in mice by administering 3 U/kg of neutral-protamine Hagadorn insulin subcutaneously. When we examined degenerating neurons in hippocampus and striatum by TUNEL labeling, injury was observed after 4 h of hypoglycemia in postnatal day (P)7 mice, and we observed more cell injury in animals rendered hypoglycemic at P7 than at P21. Studies of hippocampal slice cultures revealed that reduction in glucose concentration induced more neuronal injury in slices prepared from P3 and P7 than from P14 and P21 mice. Treatment of slices with an adenosine A(1) receptor (A(1)AR) antagonist reduced the hypoglycemic damage, whereas agonists increased damage, particularly in slices prepared from very young pups. This suggests a critically important role for A(1)ARs, which was further demonstrated by the reduction of hypoglycemic damage in hippocampal slices prepared from A(1)AR(-/-) mice. Furthermore, insulin-induced hypoglycemia in P7 A(1)AR(-/-) mice did not increase TUNEL-positive cells, but a major increase was seen in A(1)AR(+/-) mice. These observations show that the developing nervous system is indeed sensitive to acute hypoglycemic injury and that A(1)AR activation contributes to damage induced by hypoglycemia, particularly in immature mouse brain.
Collapse
Affiliation(s)
- Mina Kim
- Section of Developmental Endocrinology and Biology, Yale Child Health Research Center, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
32
|
Tasker RC, Salmond CH, Westland AG, Pena A, Gillard JH, Sahakian BJ, Pickard JD. Head circumference and brain and hippocampal volume after severe traumatic brain injury in childhood. Pediatr Res 2005; 58:302-8. [PMID: 16006434 DOI: 10.1203/01.pdr.0000169965.08854.25] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Vulnerability of the hippocampus to traumatic brain injury (TBI) in adults is related to severity of injury and white matter atrophy. The objectives of this study were to determine features of anthropometry and cerebral morphometry late after TBI in childhood and to assess whether hippocampal volume is related to severity of initial ictus and changes in white matter at follow-up. Thirty-three patients underwent magnetic resonance imaging 4.9 y after severe TBI that necessitated intensive care; 23 had mechanical ventilation and intracranial pressure monitoring longer than 3 d. Magnetic resonance imaging analyses included volume of brain, hemisphere, ventricles, and hippocampal and perihippocampal regions; spatial distribution of voxel-based morphometry differences in white matter; and eigenvalues of diffusion tensor imaging diffusivity. Patients with longer intensive care ictus had smaller-than-expected occipitofrontal head circumference. Eight of these, identified by voxel-based morphometry, had periventricular white matter loss and smaller-than-expected brain volume for OFC, suggesting "atrophy"; the remainder had expected volume for a smaller OFC, suggesting "growth disturbance." Ninety-three percent of the variation in right hippocampal volume was accounted for by factors related to severity of injury and white matter atrophy. It is concluded that anthropometry and cerebral morphometric measurements late after severe TBI in childhood provides useful outcome data and indicate that, despite adequate growth in stature, effects of TBI on brain growth and hippocampal volume may extend into adulthood.
Collapse
Affiliation(s)
- Robert C Tasker
- Department of Paediatrics, Addenbrooke's Hospital, Cambridge, CB2 2QQ, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
33
|
Fink EL, Alexander H, Marco CD, Dixon CE, Kochanek PM, Jenkins LW, Lai Y, Donovan HA, Hickey RW, Clark RSB. Experimental model of pediatric asphyxial cardiopulmonary arrest in rats. Pediatr Crit Care Med 2004; 5:139-44. [PMID: 14987343 PMCID: PMC3235952 DOI: 10.1097/01.pcc.0000112376.29903.8f] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Develop a clinically relevant model of pediatric asphyxial cardiopulmonary arrest in rats. DESIGN Prospective interventional study. SETTING University research laboratory. SUBJECTS Postnatal day 16-18 rats (n = 9/group). INTERVENTIONS Anesthetized rats were endotracheally intubated and mechanically ventilated, and vascular catheters were inserted. Vecuronium was administered, and the ventilator was disconnected from the rats for 8 mins, whereupon rats were resuscitated with epinephrine, sodium bicarbonate, and chest compressions until spontaneous circulation returned. Shams underwent all procedures except asphyxia. MEASUREMENTS AND MAIN RESULTS Asphyxial arrest typically occurred by 1 min after the ventilator was disconnected. Return of spontaneous circulation typically occurred <30 secs after resuscitation. An isoelectric electroencephalograph was observed for 30 mins after asphyxia, and rats remained comatose for 12-24 hrs. Overall survival rate was 85%. Motor function measured using beam balance and inclined plane tests was impaired on days 1 and 2, but recovered by day 3, in rats after asphyxia vs. sham injury (p <.05). Spatial memory acquisition measured using the Morris-water maze on days 7-14 and 28-35 was also impaired in rats after asphyxia vs. sham injury (total latency 379 +/- 28 vs. 501 +/- 40 secs, respectively, p <.05). DNA fragmentation was detected in CA1 hippocampal neurons bilaterally 3-7 days after asphyxia. Neurodegeneration detected using Fluorojade B was seen in bilateral CA1 hippocampi and layer V cortical neurons 3-7 days after asphyxia, with persistent neurodegeneration in CA1 hippocampus detected up to 5 wks after asphyxia. CA1 hippocampal neuron survival after asphyxia was 39-43% (p <.001 vs. sham). Evidence of DNA or cellular injury was not detected in sham rats. CONCLUSIONS This model of asphyxial cardiopulmonary arrest in postnatal day 17 rats produces many of the clinical manifestations of pediatric hypoxic-ischemic encephalopathy. This model may be useful for the preclinical testing of novel and currently available interventions aimed at improving neurologic outcome in infants and children after cardiopulmonary arrest.
Collapse
Affiliation(s)
- Ericka L. Fink
- Department of Critical Care Medicine, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Henry Alexander
- Department of Critical Care Medicine, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christina D. Marco
- Department of Critical Care Medicine, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - C. Edward Dixon
- Department of Neurological Surgery, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick M. Kochanek
- Department of Critical Care Medicine, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Larry W. Jenkins
- Department of Neurological Surgery, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yichen Lai
- Department of Critical Care Medicine, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Holly A. Donovan
- Department of Critical Care Medicine, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert W. Hickey
- Department of Pediatrics, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert S. B. Clark
- Department of Critical Care Medicine, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, the Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, and the Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
34
|
Maxwell WL, Dhillon K, Harper L, Espin J, MacIntosh TK, Smith DH, Graham DI. There is differential loss of pyramidal cells from the human hippocampus with survival after blunt head injury. J Neuropathol Exp Neurol 2003; 62:272-9. [PMID: 12638731 DOI: 10.1093/jnen/62.3.272] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The experimental literature has shown that neurons within sub-fields of the hippocampus possess differential sensitivities to cell loss after different types of insult to the brain. In humans, after blunt head injury, differential neuronal responses between sub-fields of the hippocampus up to 72 hours after injury have been documented. But, in only a small part of the literature have data for alterations in real numbers of neurons been provided. In this study the hypothesis was tested that, after severe blunt head injury in humans, the total number of neurons within a defined volume of brain tissue differed between different sub-fields of the hippocampus and between groups of patients with differing post-traumatic survivals. Stereological methods were used to measure total cross-sectional area of sub-fields of the hippocampus taken at the level of the lateral geniculate nucleus and count numbers of neurons within each of the CA1, CA2, CA3, and CA4 sub-fields of the hippocampus in patients. The patients used in this study were categorized as follows: Group 1 (early) had survived for 1 week or less; Group 2 (late) survived 6 months or longer after fatal severe head injury; and Group 3 (controls) consisted of age-matched patients that had no history of head injury or disease prior to death. There was a significant loss in cross-sectional area in sub-fields CA3 and CA4 at 1 week or less after injury and in sub-field CA1 at 6 months and greater survival. There was no change in CA2. There was loss of neurons from within a predefined volume of brain tissue in sub-fields CA1, CA3, and CA4 one week or less after injury. But there was no loss in CA2. There was continued loss of neurons from sub-fields CA1 and CA4 between 1 week and 6 months and greater survival, but there was no loss of neurons in sub-fields CA2 and CA3 within the same period. These novel data show that after human severe head injury there is first an acute loss (1 week or less survival) of pyramidal neurons in all hippocampal sub-fields except CA2. Second, there is an ongoing loss of neurons in sub-field CA1 and, most notably, in sub-field CA4, in patients surviving for more than 6 months. However, in neither group of patients is there loss of neurons from sub-field CA2.
Collapse
Affiliation(s)
- W L Maxwell
- Department of Anatomy, IBLS, University of Glasgow, Glasgow, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
35
|
Convit A, Wolf OT, Tarshish C, de Leon MJ. Reduced glucose tolerance is associated with poor memory performance and hippocampal atrophy among normal elderly. Proc Natl Acad Sci U S A 2003; 100:2019-22. [PMID: 12571363 PMCID: PMC149951 DOI: 10.1073/pnas.0336073100] [Citation(s) in RCA: 227] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Poor glucose tolerance and memory deficits, short of dementia, often accompanies aging. The purpose of this study was to ascertain whether, among nondiabetic, nondemented middle-aged and elderly individuals, poorer glucose tolerance is associated with reductions in memory performance and smaller hippocampal volumes. We studied 30 subjects who were evaluated consecutively in an outpatient research setting. The composition of the participant group was 57% female and 68.6 +/- 7.5 years of age; the participants had an average education of 16.2 +/- 2.3 years, a score on the Mini Mental State Examination of 28.6 +/- 1.5, a glycosylated hemoglobin (HbA1C) of 5.88 +/- 0.74%, and a body mass index of 24.9 +/- 4.1 kg/m(2). Glucose tolerance was measured by an i.v. glucose tolerance test. Memory was tested by using the Wechsler Paragraphs recall tests at the time of administering the i.v. glucose tolerance test. The hippocampus and other brain volumes were measured by using validated methods on standardized MRIs. Decreased peripheral glucose regulation was associated with decreased general cognitive performance, memory impairments, and atrophy of the hippocampus, a brain area that is key for learning and memory. These associations were independent of age and Mini Mental State Examination scores. Therefore, these data suggest that metabolic substrate delivery may influence hippocampal structure and function. This observation may bring to light a mechanism for aging brain injury that may have substantial medical impact, given the large number of elderly individuals with impaired glucose metabolism.
Collapse
Affiliation(s)
- Antonio Convit
- Center for Brain Health, Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA.
| | | | | | | |
Collapse
|
36
|
Sabbatini M, Catalani A, Consoli C, Marletta N, Tomassoni D, Avola R. The hippocampus in spontaneously hypertensive rats: an animal model of vascular dementia? Mech Ageing Dev 2002; 123:547-59. [PMID: 11796140 DOI: 10.1016/s0047-6374(01)00362-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hypertension is a main risk factor for cerebrovascular disease, including vascular dementia. The present study was designed to evaluate if hypertension-dependent changes of the hippocampus of spontaneously hypertensive rats (SHR) of different ages were related with those occurring in vascular dementia. The hippocampus was chosen as the brain area involved in learning and memory. Systolic pressure was slightly increased in 2-month-old SHR in comparison with age-matched normotensive Wistar-Kyoto (WKY) rats and augmented progressively with age in SHR. No microanatomical changes were observed in the hippocampus of SHR of 2 months in comparison with age-matched WKY rats. A limited decrease of white matter volume was observed in 4-month-old SHR. In SHR of 6 months, a reduction of grey matter volume both in the CA1 subfield and in the dentate gyrus occurred. Evaluation of phosphorylated 200-kDa neurofilament immunoreactivity revealed a decreased immune reaction area in the CA1 subfield of 6-month-old SHR compared to age-matched WKY rats and no changes in the expression and localization of the dendritic marker microtubule associated protein (MAP)-2. In 6-month-old SHR, an increase of glial fibrillary acidic protein (GFAP)-expression was found by Western blot analysis. Immunohistochemistry revealed an increase in number (hyperplasia), but not in size of astrocytes. These findings indicate the occurrence of cytoskeletal breakdown and astroglial changes primarily in the CA1 subfield of the hippocampus of SHR of 6 months. The occurrence in the hippocampus of SHR of regressive changes and astroglial reaction similar to those occurring in neurodegenerative disorders with cognitive impairment suggests that they represent an animal model of vascular dementia.
Collapse
Affiliation(s)
- Maurizio Sabbatini
- Sezione di Anatomia Umana, Dipartimento di Scienze Farmacologiche e Medicina Sperimentale, Università di Camerino, Via M. Scalzino 5, 62032, Camerino, Italy.
| | | | | | | | | | | |
Collapse
|
37
|
Kawamoto Y, Nakamura S, Matsuo A, Akiguchi I, Shibasaki H. Immunohistochemical localization of glial cell line-derived neurotrophic factor in the human central nervous system. Neuroscience 2001; 100:701-12. [PMID: 11036204 DOI: 10.1016/s0306-4522(00)00326-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Glial cell line-derived neurotrophic factor, initially purified from the rat glial cell line B49, has the ability to promote the survival and differentiation of various types of neurons in the central and peripheral nervous systems. In the present study, to evaluate the physiological role of glial cell line-derived neurotrophic factor in the central nervous system, we investigated the cellular and regional distribution of glial cell line-derived neurotrophic factor immunoreactivity in autopsied control human brains and spinal cords using a polyclonal glial cell line-derived neurotrophic factor-specific antibody. On western blot analysis, the antibody reacted with recombinant human glial cell line-derived neurotrophic factor, and recognized a single band at a molecular weight of approximately 34,000 in human brain homogenates. Glial cell line-derived neurotrophic factor immunoreactivity was observed mainly in the neuronal somata, dendrites and axons. In the telencephalon, diencephalon and brainstem, the cell bodies and proximal processes of several neuronal subtypes were immunostained with punctate dots. Furthermore, immunopositive nerve fibers were also observed, and numerous axons were intensely immunolabeled in the internal segment of the globus pallidus and the pars reticulata of the substantia nigra. In the cerebellum, the most conspicuous immunostaining was found in the Purkinje cells, in which the somata and dendrites were strongly immunolabeled. Intense immunoreactivity was also detected in the posterior horn of the spinal cord. In addition to the neuronal elements, immunopositive glial cell bodies and processes were observed in various regions. Our results suggest that glial cell line-derived neurotrophic factor is widely localized, but can be found selectively in certain neuronal subpopulations of the human central nervous system. Glial cell line-derived neurotrophic factor may regulate the maintenance of neuronal functions under normal circumstances.
Collapse
Affiliation(s)
- Y Kawamoto
- Department of Neurology, Faculty of Medicine, Kyoto University, 606-8507, Kyoto, Japan.
| | | | | | | | | |
Collapse
|
38
|
Tang YP, Murata Y, Nagaya T, Noda Y, Seo H, Nabeshima T. NGFI-B, c-fos, and c-jun mRNA expression in mouse brain after acute carbon monoxide intoxication. J Cereb Blood Flow Metab 1997; 17:771-80. [PMID: 9270494 DOI: 10.1097/00004647-199707000-00007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The expression of immediate early genes (IEG) has been documented in the brain after various kinds of insults such as ischemia and hypoxia. To determine whether acute carbon monoxide intoxication (ACOI) might trigger IEG expression, adult ddY mice were subjected to carbon monoxide exposure at a rate of 30 mL/min for 35 seconds. The levels of NGFI-B, c-fos, and c-jun mRNA were determined by Northern blot analysis. A time-course study in the cerebral cortex indicated that the induction of NGFI-B, c-fos, and c-jun mRNA started as early as 15 minutes, reached a peak at 30 minutes, and returned to the basal level at 1 hour after the ACOI. In addition, the temporal feature of the induction of these IEG mRNA in the hippocampus was very similar to that in the cerebral cortex. Examination of brain regions at 30 minutes after the ACOI revealed a significant induction of NGFI-B mRNA in the cerebellum, thalamus-hypothalamus, brainstem. as well as in the cortex and hippocampus, but not in the striatum or olfactory bulb. Furthermore, the neuroanatomical distribution of c-fos mRNA at 30 minutes after the ACOI was very similar to that of the NGFI-B mRNA. The widespread distribution of these IEG in the brain, especially in the cerebellum and brainstem, indicates that the major cause for the triggering of IEG expression in the brain by the ACOI might be a diffuse hypoxia. These findings show for the first time the temporal and spatial expression of IEG in the brain after ACOI.
Collapse
Affiliation(s)
- Y P Tang
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Newcomb JK, Kampfl A, Posmantur RM, Zhao X, Pike BR, Liu SJ, Clifton GL, Hayes RL. Immunohistochemical study of calpain-mediated breakdown products to alpha-spectrin following controlled cortical impact injury in the rat. J Neurotrauma 1997; 14:369-83. [PMID: 9219852 DOI: 10.1089/neu.1997.14.369] [Citation(s) in RCA: 105] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
This study examined the effect of unilateral controlled cortical impact on the appearance of calpain-mediated alpha-spectrin breakdown products (BDPs) in the rat cortex and hippocampus at various times following injury. Coronal sections were taken from animals at 15 min, 1 h, 3 h, 6 h, and 24 h after injury and immunolabeled with an antibody that recognizes calpain-mediated BDPs to alpha-spectrin (Roberts-Lewis et al., 1994). Sections from a separate group of rats were also taken at the same times and stained with hematoxylin and eosin. Analyses of early time points (15 min, 1 h, 3 h, and 6 h following injury) revealed alpha-spectrin BDPs in structurally intact neuronal soma and dendrites in cortex ipsilateral to site of injury that was not present in tissue from sham-injured control rats. By 24 h after injury labeling was not restricted to clearly defined neuronal structures in ipsilateral cortex, although there was an increased extent of diffuse labeling. BDPs to alpha-spectrin in axons were not detected until 24 h after injury, in contrast to the more rapid accumulation of BDPs observed in neuronal soma and dendrites. The presence of BDPs to alpha-spectrin in the cortex at the site of impact, and in the rostral and contralateral cortex, coincided with morphopathology detected by hematoxylin and eosin. alpha-Spectrin BDPs were also observed in the hippocampus ipsilateral to the injury in the absence of overt cell death. This investigation provides further evidence that calpain is activated after controlled cortical impact and could contribute to necrosis at the site of injury. The appearance of calpain-mediated BDPs at sites distal to the contusion site and in the hippocampus also suggests that calpain activation may precede and/or occur in the absence of extensive morphopathological changes.
Collapse
Affiliation(s)
- J K Newcomb
- Vivian L. Smith Center for Neurologic Research, Department of Neurosurgery, University of Texas Houston Health Science Center, Houston 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhang L, Mitani A, Yanase H, Kataoka K. Continuous monitoring and regulating of brain temperature in the conscious and freely moving ischemic gerbil: Effect of MK-801 on delayed neuronal death in hippocampal CA1. J Neurosci Res 1997. [DOI: 10.1002/(sici)1097-4547(19970215)47:4<440::aid-jnr9>3.0.co;2-e] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
41
|
Stafstrom CE, Tien RD, Montine TJ, Boustany RM. Refractory status epilepticus associated with progressive magnetic resonance imaging signal change and hippocampal neuronal loss. ACTA ACUST UNITED AC 1996. [DOI: 10.1016/s0896-6974(96)00023-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
42
|
Rupright J, Woods EA, Singh A. Hypoxic brain injury: evaluation by single photon emission computed tomography. Arch Phys Med Rehabil 1996; 77:1205-8. [PMID: 8931538 DOI: 10.1016/s0003-9993(96)90150-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Single photon emission computed tomography (SPECT) with 99mTc-labeled hexamethylpropylene-amine oxime (HMPAO) was used to evaluate cerebral blood flow in 6 patients with hypoxic brain injury (HBI). The SPECT scans were compared with electroencephalograms (EEGs), magnetic resonance imaging (MRI) scans, or computer tomography (CT) scans. The findings on SPECT scans in all 6 patients supported the clinical impression of HBI. SPECT showed decreased perfusion in regions of the brain including the hippocampus, cerebellum white matter, rostral brain stem, the cortex, and occiput. MRI, CT, and EEG provided information that was often not representative of HBI. It was believed that SPECT provided more useful information in the identification of HBI. SPECT imaging with 99mTc HMPAO appears to be a valuable adjunct for evaluation and clinical diagnosis of HBI patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Brain Damage, Chronic/diagnosis
- Brain Damage, Chronic/diagnostic imaging
- Brain Damage, Chronic/etiology
- Cerebrovascular Circulation
- Electroencephalography
- Female
- Humans
- Hypoxia, Brain/complications
- Hypoxia, Brain/diagnosis
- Hypoxia, Brain/diagnostic imaging
- Magnetic Resonance Angiography
- Male
- Organotechnetium Compounds
- Oximes
- Technetium Tc 99m Exametazime
- Tomography, Emission-Computed, Single-Photon
- Tomography, X-Ray Computed
Collapse
Affiliation(s)
- J Rupright
- Department of Physical Medicine and Rehabilitation, Rusk Rehabilitation Center, Columbia, MO 65212, USA
| | | | | |
Collapse
|
43
|
Abstract
Studies in humans and monkeys have identified structures in the medial temporal lobe essential for memory (the hippocampal region, i.e., the dentate gyrus, the hippocampus, and the subicular complex, and the adjacent perirhinal, entorhinal, and parahippocampal cortices). Additional work has revealed that for both species, damage limited to the hippocampal region produces less severe memory impairment than damage that includes additional structures within the medial temporal lobe. This work has been based on both neurosurgical lesions and on lesions produced by global ischemia or anoxia. An important issue about ischemic damage is whether the damage identifiable in histopathological examination provides an accurate estimate of direct neural damage or whether additional direct damage might be present that is sufficient to disrupt neuronal function in areas important for memory and sufficient to impair behavioral performance, but not sufficient to progress to cell death and to be detectable in conventional histopathology. This commentary explores the issue of ischemic damage and memory impairment. Although few studies have addressed this issue directly, the currently available data from global ischemia in rats, monkeys, and humans are consistent with the hypothesis that the detectable neuronal damage is responsible for the severity of the observed behavioral impairment. Yet it is also true that this hypothesis has not been the target of very much systematic work. We encourage additional experimental work, especially in rats, that could further illuminate how to evaluate the behavioral effects of ischemic lesions.
Collapse
Affiliation(s)
- L R Squire
- VA Medical Center, San Diego, CA 92161, USA
| | | |
Collapse
|
44
|
Katz L, Ebmeyer U, Safar P, Radovsky A, Neumar R. Outcome model of asphyxial cardiac arrest in rats. J Cereb Blood Flow Metab 1995; 15:1032-9. [PMID: 7593335 DOI: 10.1038/jcbfm.1995.129] [Citation(s) in RCA: 195] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
An outcome model with asphyxial cardiac arrest in rats has been developed for quantifying brain damage. Twenty-two rats were randomized into three groups. Control group I was normal, was conscious, and had no asphyxia (n = 6). Sham group II had anesthesia and surgery but no asphyxia (n = 6). All 12 rats in groups I and II survived to 72 h and were functionally and histologically normal. Arrest group III (the model; n = 10) had light anesthesia and apneic asphyxia of 8 min, which led to cessation of circulation at 3-4 min of apnea, resulting in cardiac arrest (no flow) of 4-5 min. All 10 rats had spontaneous circulation restored by standard external cardiopulmonary resuscitation. Nine rats survived controlled ventilation for 1 h and observation to 72 h, while one rat died before extubation. All nine survivors were conscious at 72 h, with neurologic deficit scores (0% = best; 100% = worst) of 7 +/- 6% (2-16%). All brain regions at five coronal levels were examined for ischemic neurons. The prevalence of ischemic neurons in five regions was categorically scored. The average total brain histopathologic damage score in group III (n = 9) was 2.1 (p < 0.05 vs. group I or II). A reproducible outcome model of cardiac arrest in rats was documented. It provides a tool for investigating pathophysiological mechanisms of neuronal death caused by a transient global hypoxic-ischemic brain insult.
Collapse
Affiliation(s)
- L Katz
- Safar Center for Resuscitation Research, University of Pittsburgh Medical Center, Pennsylvania 15260, USA
| | | | | | | | | |
Collapse
|
45
|
Tsuru A, Mizuguchi M, Takashima S. Cystic leukomalacia in the cerebellar folia of premature infants. Acta Neuropathol 1995; 90:400-2. [PMID: 8546030 DOI: 10.1007/bf00315013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cystic necrosis in the cerebellar white matter was found in three premature infants. The necrosis was characteristically localized in the center of the white matter of the superficial cerebellar folia, sparing the overlying cortex. The patients were aged between 28 and 34 gestational weeks, and had a clinical history of severe systemic hypotension. Thus, cystic leukomalacia represents a characteristic brain lesion in premature infants which may be caused by cerebellar hypoperfusion.
Collapse
Affiliation(s)
- A Tsuru
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | | | | |
Collapse
|
46
|
Boeve BF, Bell DG, Noseworthy JH. Bilateral temporal lobe MRI changes in uncomplicated hypoglycemic coma. Neurol Sci 1995; 22:56-8. [PMID: 7750077 DOI: 10.1017/s031716710004052x] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We report bilateral temporal lobe MRI findings in a patient following an episode of prolonged hypoglycemia uncomplicated by coexisting anoxia, hypotension, acidosis, drug intoxication, infection, or status epilepticus. The MRI findings are discussed in relation to the experimental and human data on hypoglycemic neuronal injury.
Collapse
Affiliation(s)
- B F Boeve
- Department of Neurology, Mayo Clinic/Mayo Foundation, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
47
|
Kotapka MJ, Graham DI, Adams JH, Gennarelli TA. Hippocampal pathology in fatal human head injury without high intracranial pressure. J Neurotrauma 1994; 11:317-24. [PMID: 7996585 DOI: 10.1089/neu.1994.11.317] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Traumatically induced hippocampal damage is a frequent sequela of fatal human head injury and is traditionally considered to be the result of decreased cerebral perfusion secondary to raised intracranial pressure (ICP). However, in previous studies employing an experimental model of acceleration head injury, hippocampal lesions have been observed in the absence of high ICP. To further elucidate the role of raised ICP in the production of posttraumatic hippocampal neuronal damage, 14 cases of fatal human nonmissile head injury, in which the measured ICP was less than 20 mm Hg, were subjected to light microscopic evaluation for the frequency and anatomic distribution of hippocampal damage. The mean maximal ICP of the 14 patients was 17.6 mm Hg. Detailed light microscopic evaluation revealed hippocampal lesions in 12 of the 14 cases studied (86%). These lesions were typically bilateral foci of selective neuronal loss in the CA1 subfield of the hippocampus. The nature and distribution of hippocampal lesions were similar to those previously reported both in fatal human head injury associated with elevated ICP and in experimental acceleration head injury without raised ICP. These results provide further evidence that the occurrence of hippocampal neuronal loss following head injury is not exclusively dependent on elevated ICP. Other mechanisms, such as pathologic excitation of neurons, may be involved.
Collapse
Affiliation(s)
- M J Kotapka
- Division of Neurosurgery, University of Pennsylvania, Philadelphia
| | | | | | | |
Collapse
|
48
|
Lunetta M, Damanti AR, Fabbri G, Lombardo M, Di Mauro M, Mughini L. Evidence by magnetic resonance imaging of cerebral alterations of atrophy type in young insulin-dependent diabetic patients. J Endocrinol Invest 1994; 17:241-5. [PMID: 7930374 DOI: 10.1007/bf03348967] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Aim of this study was to investigate a) if through Magnetic Resonance Imaging (MRI) it was possible to reveal cerebral alterations in patients with insulin-dependent diabetes mellitus (IDDM); b) if there was any correlation with hypoglycemic episodes, glycometabolic control, microvascular alterations and diabetic peripheral neuropathy. For this purpose ten ID-DM patients under treatment with human insulin, aged 19-30 yr with the disease, the duration being from 1 to 19 yr, were investigated by MRI using a Philips Gyroscan. Spin Echo sequences were used with images in T1 T2 in sagittal and axial planes. To measure the ventricular dilatation the cerebroventricular index (CVI) was evaluated. The MRI has put in evidence in 7/10 patients a dilatation in the lateral ventricles and subarachnoidal spaces of the cerebral vault and the cerebellum clearly due to cerebral atrophy. The CVI mean values (34.78 +/- 2.92) were statistically (p < 0.001) higher in diabetic patients respect to control subjects (CVI mean values 27.5 +/- 1.58). These alterations did not present clear correlations with the degree of glycometabolic control, duration of disease, number of symptomatic hypoglycemic episodes and threshold for hypoglycemic symptoms, retinal microvascular alterations, microalbuminuria, diabetic peripheral neuropathy. The clinical or functional relevance of CVI changes and the exact pathogenic mechanism remains to be clarified.
Collapse
Affiliation(s)
- M Lunetta
- Cattedra di Malattie del Metabolismo, II Istituto di Patologia Medica, Catania, Italy
| | | | | | | | | | | |
Collapse
|
49
|
Kitamura O. Immunohistochemical investigation of hypoxic/ischemic brain damage in forensic autopsy cases. Int J Legal Med 1994; 107:69-76. [PMID: 7819122 DOI: 10.1007/bf01225492] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A neuropathological study of 41 forensic autopsy cases of hypoxic/ischemic brain damage has been undertaken, using immunohistochemical staining to detect the 70-kDa heat shock protein (hsp70) and the status of the glial cells. In cases surviving 2-5 h after hypoxic/ischemic injury, ischemic cell changes were seen whereas glial reactions were not apparent. In cases of longer survival, neuronal necrosis and a loss of neurons were seen, and these changes were accompanied by proliferation of glial fibrillary acidic protein (GFAP), vimentin-positive astrocytes and microglia which transformed into rod cells or lipid-laden macrophages. In cases with a history of hypoxic attacks, GFAP-positive and vimentin-negative astrocytes had proliferated in the CA3 and CA4 regions of hippocampus. The cases of severe hypoxic injury, such as an asthmatic attack and choking, showed no ischemic changes in the hippocampal neurons. On the other hand, the CA1 pyramidal cells showed neuronal necrosis in a patient suffering from tetralogy of Fallot (TOF), who survived for 2 h after a traffic accident. Therefore, it is suggested that even moderate hypoxic injury induces astrocytosis in the CA3 and CA4 regions and may affect the neuronal proteins and the metabolism, and that in cases with a history of hypoxic attacks neuronal damage may be severe even several hours after ischemic injury. The protein hsp70 expression was found in the CA2, CA3 and CA4 regions in cases of long-term survival after severe hypoxic/ischemic injury and in cases of alcoholic intake or toluene abuse just before acute death.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- O Kitamura
- Department of Legal Medicine, Nagasaki University School of Medicine, Japan
| |
Collapse
|
50
|
Yamaoka Y, Shimohama S, Kimura J, Fukunaga R, Taniguchi T. Neuronal damage in the rat hippocampus induced by in vivo hypoxia. EXPERIMENTAL AND TOXICOLOGIC PATHOLOGY : OFFICIAL JOURNAL OF THE GESELLSCHAFT FUR TOXIKOLOGISCHE PATHOLOGIE 1993; 45:205-9. [PMID: 8219709 DOI: 10.1016/s0940-2993(11)80389-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Rats were subjected to hypoxia for 30 min in a chamber containing 5% O2 and 95% N2. The distribution of damaged neurons in the hippocampus was then examined at various predetermined times, ranging from 3 hours to 21 days after hypoxia. Hematoxylin-eosin stained sections showed shrunken and eosinophilic neurons in the CA3 and CA4 regions. Similar, but less severe, changes were also observed in the granule cell layer of the dentate gyrus. In contrast, neurons in the CA1 region were relatively resistant to hypoxia. These results showed the susceptibility of the hippocampus to hypoxia, although the affected neurons are not the same as those vulnerable to ischemia.
Collapse
Affiliation(s)
- Y Yamaoka
- Department of Neurology, Faculty of Medicine, Kyoto University, Japan
| | | | | | | | | |
Collapse
|