1
|
Prehn A, Hobusch C, Härtig W, Michalski D, Krueger M, Flachmeyer B. Increasing reproducibility in preclinical stroke research: the correlation of immunofluorescence intensity measurements and Western blot analyses strongly depends on antibody clonality and tissue pre-treatment in a mouse model of focal cerebral ischemia. Front Cell Neurosci 2023; 17:1183232. [PMID: 37342767 PMCID: PMC10277931 DOI: 10.3389/fncel.2023.1183232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
In the setting of stroke, ischemia not only impairs neuronal function, but also detrimentally affects the different components of the neurovascular unit, which are shown to be involved in the transition from reversible to long-lasting tissue damage. In this context, the glial proteins myelin basic protein (MBP) and the 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNP) as well as the vasculature-associated basement membrane proteins laminin and collagen IV have been identified as ischemia-sensitive elements. However, available data from immunofluorescence and Western blot analyses are often found to be contradictory, which renders interpretation of the respective data rather difficult. Therefore, the present study investigates the impact of tissue pre-treatment and antibody clonality on immunofluorescence measurements of the mentioned proteins in a highly reproducible model of permanent middle cerebral artery occlusion. Here, immunofluorescence labeling using polyclonal antibodies revealed an increased immunofluorescence intensity of MBP, CNP, laminin and collagen IV in ischemic areas, although Western blot analyses did not reveal increased protein levels. Importantly, contrary to polyclonal antibodies, monoclonal ones did not provide increased fluorescence intensities in ischemic areas. Further, we were able to demonstrate that different ways of tissue pre-treatment including paraformaldehyde fixation and antigen retrieval may not only impact on fluorescence intensity measurements in general, but rather one-sidedly affect either ischemic or unaffected tissue. Therefore, immunofluorescence intensity measurements do not necessarily correlate with the actual protein levels, especially in ischemia-affected tissue and should always be complemented by different techniques to enhance reproducibility and to hopefully overcome the translational roadblock from bench to bedside.
Collapse
Affiliation(s)
- Anna Prehn
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | | | - Martin Krueger
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
2
|
Mages B, Fuhs T, Aleithe S, Blietz A, Hobusch C, Härtig W, Schob S, Krueger M, Michalski D. The Cytoskeletal Elements MAP2 and NF-L Show Substantial Alterations in Different Stroke Models While Elevated Serum Levels Highlight Especially MAP2 as a Sensitive Biomarker in Stroke Patients. Mol Neurobiol 2021; 58:4051-4069. [PMID: 33931805 PMCID: PMC8280005 DOI: 10.1007/s12035-021-02372-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023]
Abstract
In the setting of ischemic stroke, the neurofilament subunit NF-L and the microtubule-associated protein MAP2 have proven to be exceptionally ischemia-sensitive elements of the neuronal cytoskeleton. Since alterations of the cytoskeleton have been linked to the transition from reversible to irreversible tissue damage, the present study investigates underlying time- and region-specific alterations of NF-L and MAP2 in different animal models of focal cerebral ischemia. Although NF-L is increasingly established as a clinical stroke biomarker, MAP2 serum measurements after stroke are still lacking. Therefore, the present study further compares serum levels of MAP2 with NF-L in stroke patients. In the applied animal models, MAP2-related immunofluorescence intensities were decreased in ischemic areas, whereas the abundance of NF-L degradation products accounted for an increase of NF-L-related immunofluorescence intensity. Accordingly, Western blot analyses of ischemic areas revealed decreased protein levels of both MAP2 and NF-L. The cytoskeletal alterations are further reflected at an ultrastructural level as indicated by a significant reduction of detectable neurofilaments in cortical axons of ischemia-affected areas. Moreover, atomic force microscopy measurements confirmed altered mechanical properties as indicated by a decreased elastic strength in ischemia-affected tissue. In addition to the results from the animal models, stroke patients exhibited significantly elevated serum levels of MAP2, which increased with infarct size, whereas serum levels of NF-L did not differ significantly. Thus, MAP2 appears to be a more sensitive stroke biomarker than NF-L, especially for early neuronal damage. This perspective is strengthened by the results from the animal models, showing MAP2-related alterations at earlier time points compared to NF-L. The profound ischemia-induced alterations further qualify both cytoskeletal elements as promising targets for neuroprotective therapies.
Collapse
Affiliation(s)
- Bianca Mages
- Institute of Anatomy, Leipzig University, Leipzig, Germany.
| | - Thomas Fuhs
- Section of Soft Matter Physics, Faculty of Physics and Geosciences, Leipzig University, Leipzig, Germany
| | - Susanne Aleithe
- Department of Neurology, Leipzig University, Leipzig, Germany
| | | | | | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | - Stefan Schob
- Department of Neuroradiology, Leipzig University, Leipzig, Germany
| | - Martin Krueger
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | | |
Collapse
|
3
|
Aleithe S, Blietz A, Mages B, Hobusch C, Härtig W, Michalski D. Transcriptional Response and Morphological Features of the Neurovascular Unit and Associated Extracellular Matrix After Experimental Stroke in Mice. Mol Neurobiol 2019; 56:7631-7650. [PMID: 31089963 PMCID: PMC6815284 DOI: 10.1007/s12035-019-1604-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/10/2019] [Indexed: 12/18/2022]
Abstract
Experimental stroke studies yielded insights into single reactions of the neurovascular unit (NVU) and associated extracellular matrix (ECM). However, the extent of simultaneous processes caused by ischemia and their underlying transcriptional changes are still poorly understood. Strictly following the NVU and ECM concept, this study explored transcriptional responses of cellular and non-cellular components as well as their morphological characteristics following ischemia. Mice were subjected to 4 or 24 h of unilateral middle cerebral artery occlusion. In the neocortex and the striatum, cytoskeletal and glial elements as well as blood-brain barrier and ECM components were analyzed using real-time PCR. Western blot analyses allowed characterization of protein levels and multiple immunofluorescence labeling enabled morphological assessment. Out of 37 genes analyzed, the majority exhibited decreased mRNA levels in ischemic areas, while changes occurred as early as 4 h after ischemia. Down-regulated mRNA levels were predominantly localized in the neocortex, such as the structural elements α-catenin 2, N-cadherin, β-catenin 1, and βIII-tubulin, consistently decreasing 4 and 24 h after ischemia. However, a few genes, e.g., claudin-5 and Pcam1, exhibited increased mRNA levels after ischemia. For several components such as βIII-tubulin, N-cadherin, and β-catenin 1, matching transcriptional and immunofluorescence signals were obtained, whereas a few markers including neurofilaments exhibited opposite directions. In conclusion, the variety in gene regulation emphasizes the complexity of interactions within the ischemia-affected NVU and ECM. These data might help to focus future research on a set of highly sensitive elements, which might prospectively facilitate neuroprotective strategies beyond the traditional single target perspective.
Collapse
Affiliation(s)
- Susanne Aleithe
- Department of Neurology, University of Leipzig, Liebigstr. 20, 04103, Leipzig, Germany.
- University of Leipzig, Liebigstr. 19, 04103, Leipzig, Germany.
| | - Alexandra Blietz
- Department of Neurology, University of Leipzig, Liebigstr. 20, 04103, Leipzig, Germany
- University of Leipzig, Liebigstr. 19, 04103, Leipzig, Germany
| | - Bianca Mages
- Department of Neurology, University of Leipzig, Liebigstr. 20, 04103, Leipzig, Germany
- Institute of Anatomy, University of Leipzig, Liebigstr. 13, 04103, Leipzig, Germany
| | - Constance Hobusch
- Institute of Anatomy, University of Leipzig, Liebigstr. 13, 04103, Leipzig, Germany
| | - Wolfgang Härtig
- University of Leipzig, Liebigstr. 19, 04103, Leipzig, Germany
| | - Dominik Michalski
- Department of Neurology, University of Leipzig, Liebigstr. 20, 04103, Leipzig, Germany.
| |
Collapse
|
4
|
Mages B, Aleithe S, Altmann S, Blietz A, Nitzsche B, Barthel H, Horn AKE, Hobusch C, Härtig W, Krueger M, Michalski D. Impaired Neurofilament Integrity and Neuronal Morphology in Different Models of Focal Cerebral Ischemia and Human Stroke Tissue. Front Cell Neurosci 2018; 12:161. [PMID: 29967576 PMCID: PMC6015914 DOI: 10.3389/fncel.2018.00161] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/25/2018] [Indexed: 12/12/2022] Open
Abstract
As part of the neuronal cytoskeleton, neurofilaments are involved in maintaining cellular integrity. In the setting of ischemic stroke, the affection of the neurofilament network is considered to mediate the transition towards long-lasting tissue damage. Although peripheral levels of distinct neurofilament subunits are shown to correlate with the clinically observed severity of cerebral ischemia, neurofilaments have so far not been considered for neuroprotective approaches. Therefore, the present study systematically addresses ischemia-induced alterations of the neurofilament light (NF-L), medium (NF-M), and heavy (NF-H) subunits as well as of α-internexin (INA). For this purpose, we applied a multi-parametric approach including immunofluorescence labeling, western blotting, qRT-PCR and electron microscopy. Analyses comprised ischemia-affected tissue from three stroke models of middle cerebral artery occlusion (MCAO), including approaches of filament-based MCAO in mice, thromboembolic MCAO in rats, and electrosurgical MCAO in sheep, as well as human autoptic stroke tissue. As indicated by altered immunosignals, impairment of neurofilament subunits was consistently observed throughout the applied stroke models and in human tissue. Thereby, altered NF-L immunoreactivity was also found to reach penumbral areas, while protein analysis revealed consistent reductions for NF-L and INA in the ischemia-affected neocortex in mice. At the mRNA level, the ischemic neocortex and striatum exhibited reduced expressions of NF-L- and NF-H-associated genes, whereas an upregulation for Ina appeared in the striatum. Further, multiple fluorescence labeling of neurofilament proteins revealed spheroid and bead-like structural alterations in human and rodent tissue, correlating with a cellular edema and lost cytoskeletal order at the ultrastructural level. Thus, the consistent ischemia-induced affection of neurofilament subunits in animals and human tissue, as well as the involvement of potentially salvageable tissue qualify neurofilaments as promising targets for neuroprotective strategies. During ischemia formation, such approaches may focus on the maintenance of neurofilament integrity, and appear applicable as co-treatment to modern recanalizing strategies.
Collapse
Affiliation(s)
- Bianca Mages
- Department of Neurology, University of Leipzig, Leipzig, Germany.,Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany.,Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Susanne Aleithe
- Department of Neurology, University of Leipzig, Leipzig, Germany.,Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Stephan Altmann
- Department of Neurology, University of Leipzig, Leipzig, Germany.,Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Alexandra Blietz
- Department of Neurology, University of Leipzig, Leipzig, Germany.,Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Björn Nitzsche
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany.,Institute of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Anja K E Horn
- Institute of Anatomy and Cell Biology I and German Center for Vertigo and Balance Disorders, Ludwig Maximilian University of Munich, Munich, Germany
| | | | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Martin Krueger
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | | |
Collapse
|
5
|
Tetramethylpyrazine nitrone, a multifunctional neuroprotective agent for ischemic stroke therapy. Sci Rep 2016; 6:37148. [PMID: 27841332 PMCID: PMC5107909 DOI: 10.1038/srep37148] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 10/25/2016] [Indexed: 11/08/2022] Open
Abstract
TBN, a novel tetramethylpyrazine derivative armed with a powerful free radical-scavenging nitrone moiety, has been reported to reduce cerebral infarction in rats through multi-functional mechanisms of action. Here we study the therapeutic effects of TBN on non-human primate model of stroke. Thirty male Cynomolgus macaques were subjected to stroke with 4 hours ischemia and then reperfusion. TBN were injected intravenously at 3 or 6 hours after the onset of ischemia. Cerebral infarction was examined by magnetic resonance imaging at 1 and 4 weeks post ischemia. Neurological severity scores were evaluated during 4 weeks observation. At the end of experiment, protein markers associated with the stroke injury and TBN treatment were screened by quantitative proteomics. We found that TBN readily penetrated the blood brain barrier and reached effective therapeutic concentration after intravenous administration. It significantly reduced brain infarction and modestly preserved the neurological function of stroke-affected arm. TBN suppressed over-expression of neuroinflammatory marker vimentin and decreased the numbers of GFAP-positive cells, while reversed down-regulation of myelination-associated protein 2', 3'-cyclic-nucleotide 3'-phosphodiesterase and increased the numbers of NeuN-positive cells in the ipsilateral peri-infarct area. TBN may serve as a promising new clinical candidate for the treatment of ischemic stroke.
Collapse
|
6
|
Badawi Y, Pal R, Hui D, Michaelis EK, Shi H. Ischemic tolerance in an in vivo model of glutamate preconditioning. J Neurosci Res 2014; 93:623-32. [PMID: 25421886 DOI: 10.1002/jnr.23517] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 09/29/2014] [Accepted: 10/17/2014] [Indexed: 11/10/2022]
Abstract
Ischemia initiates a complicated biochemical cascade of events that triggers neuronal death. This study focuses on glutamate-mediated neuronal tolerance to ischemia-reperfusion. We employed an animal model of lifelong excess release of glutamate, the glutamate dehydrogenase 1 transgenic (Tg) mouse, as a model of in vivo glutamate preconditioning. Nine- and twenty-two-month-old Tg and wild-type (wt) mice were subjected to 90 min of middle cerebral artery occlusion, followed by 24 hr of reperfusion. The Tg mice suffered significantly reduced infarction and edema volume compared with their wt counterparts. We further analyzed proteasomal activity, level of ubiquitin immunostaining, and microtubule-associated protein-2A (MAP2A) expression to understand the mechanism of neuroprotection observed in the Tg mice. We found that, in the absence of ischemia, the Tg mice exhibited higher activity of the 20S and 26S proteasomes, whereas there was no significant difference in the level of hippocampal ubiquitin immunostaining between wt and Tg mice. A surprising, significant increase was observed in MAP2A expression in neurons of the Tg hippocampus following ischemia-reperfusion compared with that in wt hippocampus. The results suggest that increased proteasome activity and MAP2A synthesis and transport might account for the effectiveness of glutamate preconditioning against ischemia-reperfusion.
Collapse
Affiliation(s)
- Yomna Badawi
- Neuroscience Program, University of Kansas, Lawrence, Kansas; Department of Pharmacology and Toxicology, University of Kansas, Lawrence, Kansas
| | | | | | | | | |
Collapse
|
7
|
Kim B, Haque A, Arnaud FG, Teranishi K, Steinbach T, Auker CR, McCarron RM, Freilich D, Scultetus AH. Use of recombinant factor VIIa (rFVIIa) as pre-hospital treatment in a swine model of fluid percussion traumatic brain injury. J Emerg Trauma Shock 2014; 7:102-11. [PMID: 24812455 PMCID: PMC4013725 DOI: 10.4103/0974-2700.130880] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 11/19/2013] [Indexed: 11/04/2022] Open
Abstract
CONTEXT Recombinant factor VIIa (rFVIIa) has been used as an adjunctive therapy for acute post-traumatic hemorrhage and reversal of iatrogenic coagulopathy in trauma patients in the hospital setting. However, investigations regarding its potential use in pre-hospital management of traumatic brain injury (TBI) have not been conducted extensively. AIMS In the present study, we investigated the physiology, hematology and histology effects of a single pre-hospital bolus injection of rFVIIa compared to current clinical practice of no pre-hospital intervention in a swine model of moderate fluid percussion TBI. MATERIALS AND METHODS Animals were randomized to receive either a bolus of rFVIIa (90 μg/kg) or nothing 15 minutes (T15) post-injury. Hospital arrival was simulated at T60, and animals were euthanized at experimental endpoint (T360). RESULTS Survival was 100% in both groups; baseline physiology parameters were similar, vital signs were comparable. Animals that received rFVIIa demonstrated less hemorrhage in subarachnoid space (P = 0.0037) and less neuronal degeneration in left hippocampus, pons, and cerebellum (P = 0.00009, P = 0.00008, and P = 0.251, respectively). Immunohistochemical staining of brain sections showed less overall loss of microtubule-associated protein 2 (MAP2) and less Flouro-Jade B positive cells in rFVIIa-treated animals. CONCLUSIONS Early pre-hospital administration of rFVIIa in this swine TBI model reduced neuronal necrosis and intracranial hemorrhage (ICH). These results merit further investigation of this approach in pre-hospital trauma care.
Collapse
Affiliation(s)
- Bobby Kim
- Department of Neuro Trauma, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Ashraful Haque
- Department of Neuro Trauma, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Françoise G Arnaud
- Department of Neuro Trauma, Naval Medical Research Center, Silver Spring, Maryland, USA ; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Kohsuke Teranishi
- Department of Neuro Trauma, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Thomas Steinbach
- Department of Veterinary Pathology, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Charles R Auker
- Department of Neuro Trauma, Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Richard M McCarron
- Department of Neuro Trauma, Naval Medical Research Center, Silver Spring, Maryland, USA ; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Daniel Freilich
- Department of Neuro Trauma, Naval Medical Research Center, Silver Spring, Maryland, USA ; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Anke H Scultetus
- Department of Neuro Trauma, Naval Medical Research Center, Silver Spring, Maryland, USA ; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
8
|
Abstract
OBJECTIVE This study was designed to investigate the effects of hypoxia on neural process proliferation by studying its effects on growth cone tubulin and insulin-like growth factor (IGF)-I receptor content. METHODS Six fetal lambs were catheterized in the brachial artery and vein. Maternal oxygenation was reduced in steps from a fractional inspired oxygen concentration (FiO2) of 20% to 6% by addition of nitrogen to the inhaled gas mixture for a period of 4 h of reduced oxygen intake. Fetal arterial blood was sampled after the maternal FiO2 and oxygen were stable for >5 min at maternal FiO2 of 20% to 6%. Controls were obtained from normoxic fetuses whose ewes had similar surgery and were kept at an FiO2 of 20% throughout the experiment. Growth cones were isolated from the fetal cerebrum and cerebellum. alpha-tubulin and IGF-I receptors were quantified by immunoblotting. Tubulin and IGF-I receptor mRNA expressions were quantified by real-time polymerase chain reaction. RESULTS Maternal nitrogen breathing reduced fetal arterial pH from 7.32+/-0.06 to 6.99+/-0.02 (p<0.001). Hypoxia increased IGF-I receptors from 143+/-10 to 327+/-14 (p<0.001) and from 272+/-26 to 396+/-34 (p<0.001) fluorescence units/microg protein in the cerebrum and cerebellum, respectively. It also increased alpha-tubulin from 713+/-30 to 1873+/-126 (p<0.001) and from 780+/-34 to 2362+/-79 (p<0.001) fluorescence units/microg protein in the cerebrum and cerebellum, respectively. Expression of IGF-I receptor mRNA increased significantly in the hypoxic animals both in the cerebrum and the cerebellum, but there was no change in expression of alpha-tubulin mRNA. CONCLUSIONS This increase in IGF-I receptor expression and growth cone content may be an adaptive response to hypoxia to maintain neurite growth by facilitating binding of IGF-I. Hypoxia also increased the growth cone level of alpha-tubulin but did not increase its mRNA expression, which may indicate an inability to polymerize tubulin and build microtubules.
Collapse
Affiliation(s)
- B L G Morgan
- Division of Maternal-Fetal, Department of Obstetrics and Gynecology, UCLA Medical Center, Torrance, CA 90509, USA
| | | |
Collapse
|
9
|
Abstract
Recently reported contrast in phase images of human and animal brains obtained with gradient-recalled echo MRI holds great promise for the in vivo study of biological tissue structure with substantially improved resolution. Herein we investigate the origins of this contrast and demonstrate that it depends on the tissue "magnetic architecture" at the subcellular and cellular levels. This architecture is mostly determined by the structural arrangements of proteins, lipids, non-heme tissue iron, deoxyhemoglobin, and their magnetic susceptibilities. Such magnetic environment affects/shifts magnetic resonance (MR) frequencies of the water molecules moving/diffusing in the tissue. A theoretical framework allowing quantitative evaluation of the corresponding frequency shifts is developed based on the introduced concept of a generalized Lorentzian approximation. It takes into account both tissue architecture and its orientation with respect to the external magnetic field. Theoretical results quantitatively explain frequency contrast between GM, WM, and CSF previously reported in motor cortex area, including the absence of the contrast between WM and CSF. Comparison of theory and experiment also suggests that in a normal human brain, proteins, lipids, and non-heme iron provide comparable contributions to tissue phase contrast; however, the sign of iron and lipid contributions is opposite to the sign of contribution from proteins. These effects of cellular composition and architecture are important for quantification of tissue microstructure based on MRI phase measurements. Also theory predicts the dependence of the signal phase on the orientation of WM fibers, holding promise as additional information for fiber tracking applications.
Collapse
|
10
|
Calpain-mediated signaling mechanisms in neuronal injury and neurodegeneration. Mol Neurobiol 2008; 38:78-100. [PMID: 18686046 DOI: 10.1007/s12035-008-8036-x] [Citation(s) in RCA: 281] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 07/17/2008] [Indexed: 12/19/2022]
Abstract
Calpain is a ubiquitous calcium-sensitive protease that is essential for normal physiologic neuronal function. However, alterations in calcium homeostasis lead to persistent, pathologic activation of calpain in a number of neurodegenerative diseases. Pathologic activation of calpain results in the cleavage of a number of neuronal substrates that negatively affect neuronal structure and function, leading to inhibition of essential neuronal survival mechanisms. In this review, we examine the mechanistic underpinnings of calcium dysregulation resulting in calpain activation in the acute neurodegenerative diseases such as cerebral ischemia and in the chronic neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, prion-related encephalopathy, and amylotrophic lateral sclerosis. The premise of this paper is that analysis of the signaling and transcriptional consequences of calpain-mediated cleavage of its various substrates for any neurodegenerative disease can be extrapolated to all of the neurodegenerative diseases vulnerable to calcium dysregulation.
Collapse
|
11
|
Windelborn JA, Lipton P. Lysosomal release of cathepsins causes ischemic damage in the rat hippocampal slice and depends on NMDA-mediated calcium influx, arachidonic acid metabolism, and free radical production. J Neurochem 2008; 106:56-69. [PMID: 18363826 DOI: 10.1111/j.1471-4159.2008.05349.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
NMDA-mediated calcium entry and reactive oxygen species (ROS) production are well-recognized perpetrators of ischemic neuronal damage. The current studies show that these events lead to the release of the protein hydrolase, cathepsin B, from lysosomes 2 h following 5-min oxygen-glucose deprivation in the rat hippocampal slice. This release reflects a lysosomal membrane permeabilization (LMP) and was measured as the appearance of diffuse immunolabeled cathepsin B in the cytosol of CA1 pyramidal neurons. Necrotic neuronal damage begins after the release of cathepsins and is prevented by inhibitors of either cathepsin B or D indicating that the release of cathepsins is an important mediator of severe damage. There was an increase in superoxide levels, measured by dihydroethidium fluorescence, at the same time as LMP and reducing ROS levels with antioxidants, Trolox or N-tert-butyl-alpha-phenyl nitrone, blocked LMP. Both LMP and ROS production were blocked by an NMDA channel blocker (MK-801) and by inhibitors of mitogen-activated protein kinase kinase (U0126), calcium-dependent/independent phospholipases A2 (methyl arachidonyl fluorophosphonate) but not calcium-independent phospholipases A2 (bromoenol lactone) and cyclooxygenase-2 (NS398). A cell-permeant specific inhibitor of calpain (PD150606) prevented LMP, but not ROS production. It is concluded that LMP results in part from calcium-initiated and extracellular signal-regulated kinase-initiated arachidonic acid metabolism, which produces free radicals; it also requires the action of calpain.
Collapse
Affiliation(s)
- James A Windelborn
- Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | |
Collapse
|
12
|
Maeda M, Ampo KI, Kiryu-Seo S, Konishi H, Ohba N, Kadono C, Kiyama H. The p53-independent nuclear translocation of Cyclin G1 in degenerating neurons by ischemic and traumatic insults. Exp Neurol 2005; 193:350-60. [PMID: 15869937 DOI: 10.1016/j.expneurol.2005.01.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2004] [Revised: 12/24/2004] [Accepted: 01/04/2005] [Indexed: 11/21/2022]
Abstract
Cyclin G1 (CG1) was identified as a p53-transactivated target gene, and yet its physiological and pathological roles have been unclear. Here, we demonstrate that CG1 is translocated from cytoplasm to the nuclei of neurons in response to variety of injuries. In the normal matured rodent brain, CG1 immunoreactivity was hardly observed; however, some brain injuries exhibited intense CG1 immunoreactivity in the nuclei of the damaged neurons. Transient common carotid artery occlusion (CCAO) in the gerbil showed strong CG1-like immunoreactivity in the hippocampal CA1 neurons, and permanent middle cerebral artery occlusion (MCAO) in the mouse showed strong CG1-like immunoreactivity in the nuclei of neurons located in the ischemic brain regions. TUNEL staining did not exactly overlap with the CG1-positive cells, but overlapped highly with Fluoro-Jade B staining, a degeneration marker. Brain trauma caused by knife cut, cold injury, and kinate injection also showed CG1 accumulation in the neuronal nuclei located near the injury site. These observations were obtained in p53-deficient mice as well, suggesting that the accumulation of CG1 in the injured neurons is p53-independent. A similar nuclear translocation of endogenous CG1 was confirmed in a primary culture of cortical neurons when a toxic level of N-methyl-D-aspartate (NMDA) was applied. These results demonstrate that nuclear translocation of CG1 from cytoplasmic region occurs in damaged and degenerating neurons in a p53-independent manner, and the CG1 nuclear staining could be a good marker for the neurons received fatal damages.
Collapse
Affiliation(s)
- Mitsuyo Maeda
- Department of Anatomy, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | | | | | | | | | | | | |
Collapse
|
13
|
Neurotrophic effects of vascular endothelial growth factor on organotypic cortical explants and primary cortical neurons. J Neurosci 2003. [PMID: 14657160 DOI: 10.1523/jneurosci.23-35-11036.2003] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is well known to play an important regulatory role in vascular growth and development. Because gene knock-outs of VEGF and its receptors flk-1 and flt-1 result in early embryonic lethality, determining roles for VEGF in CNS development has been particularly difficult. Recent studies have shown that VEGF is upregulated after various injuries to the adult brain and that the cytokine affords protection to cultured neurons affected by oxidative or excitotoxic stress. The present study demonstrates, for the first time, that VEGF is directly neurotrophic to CNS neurons in culture. We applied VEGF to normoxic fetal organotypic cortical explants as a model of CNS neuropil, in addition to primary cortical neurons, to assess direct growth effects absent vascular or astroglial activity. We found that VEGF provided a significant dose-responsive increase in the neuronal microtubule markers TUJ1 and MAP-2, as well as mRNA for MAP-2 and flk-1. Antisense oligodeoxynucleotides to flk-1, but not flt-1, inhibited neuritic outgrowth, whereas inhibitors of the signaling pathways MEK1 and P13-AKT both abrogated VEGF-induced growth. VEGF applied to primary cortical neurons produced significant increases in neuronal cell body diameter and the number of emerging neurites mediated by flk-1. Possibly, VEGF achieves its effects by acting on the neuronal microtubular content, which is involved with growth, stability and maturation. Several studies have now shown that VEGF is neurotrophic and neuroprotective independent of a vascular component; we suggest that VEGF plays seminal pleiotrophic roles in CNS development and repair.
Collapse
|
14
|
Akulinin VA, Dahlstrom A. Quantitative analysis of MAP2 immunoreactivity in human neocortex of three patients surviving after brain ischemia. Neurochem Res 2003; 28:373-8. [PMID: 12608711 DOI: 10.1023/a:1022401922669] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Transient global ischemia caused by cardiac arrest results in lesions that involve all brain structures. The aim of this study was to investigate the distribution of MAP2 immunoreactivity in neurons in the brain of patients surviving for various times after an ischemic incident, using confocal laser scanning microscopy. We performed a quantitative analysis of the distribution and density of MAP2-positive structures in human neocortical areas after survival times of 1 week, 3 months, and 1 year after the cardiac arrest. Three important observations were made in the present study: (i) in all human brain areas investigated (motor, temporal, frontal, and visual cortex) a decrease of MAP2 immunoreactivity was found; (ii) in all studied areas the most significant decrease in MAP2 was found in layers II-III, compared with layers V-VII; (iii) the decrease of MAP2 immunoreactivity in layers II-III was related to the duration of the postischemic period. The maximal decrease, by 66.3% (P < .05), in MAP2-positive pyramidal neurons, was observed in layers II-III in the motor cortex after 1 year of survival after cardiac arrest.
Collapse
Affiliation(s)
- Victor A Akulinin
- Department of Histology, Embryology and Cell Biology, Omsk State Medical Academy, Lenin 1 Omsk 644099, Russia.
| | | |
Collapse
|
15
|
Beaulieu JM, Kriz J, Julien JP. Induction of peripherin expression in subsets of brain neurons after lesion injury or cerebral ischemia. Brain Res 2002; 946:153-61. [PMID: 12137917 DOI: 10.1016/s0006-8993(02)02830-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Peripherin is a type III intermediate filament predominantly expressed in neurons having direct axonal projections toward peripheral structures. Here, we report that brain injuries can trigger expression of peripherin and the formation of peripherin accumulations in neurons that are normally silent for this gene. Stab lesions made with nitrocellulose implants induced within 4 days the formation of peripherin accumulations, devoid of neurofilament proteins, in thalamic neurites at the site of the lesion. The local administration of interleukin-6 or leukemia inhibitory factor at the site of the stab lesion extended the expression pattern of peripherin to other neuronal subsets in areas of the cortex and/or of the hippocampus adjacent to injury. We also show that transient focal ischemia in mice, a model of stroke, can trigger within 72 h the formation of neuronal peripherin accumulations in neurons of the cortex, thalamus and hippocampus. This new type of potentially noxious intermediate filament protein accumulations, composed of peripherin, may be of relevance to many brain degenerative disorders with occurrence of proinflammatory cytokines.
Collapse
Affiliation(s)
- Jean-Martin Beaulieu
- Centre for Research in Neurosciences, McGill University, The Montreal General Hospital Research Institute, 1650 Cedar Avenue, Quebec H3G 1A4, Canada
| | | | | |
Collapse
|
16
|
Abdel-Rahman A, Shetty AK, Abou-Donia MB. Acute exposure to sarin increases blood brain barrier permeability and induces neuropathological changes in the rat brain: dose-response relationships. Neuroscience 2002; 113:721-41. [PMID: 12150792 DOI: 10.1016/s0306-4522(02)00176-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We hypothesize that a single exposure to an LD(50) dose of sarin induces widespread early neuropathological changes in the adult brain. In this study, we evaluated the early changes in the adult brain after a single exposure to different doses of sarin. Adult male rats were exposed to sarin by a single intramuscular injection at doses of 1, 0.5, 0.1 and 0.01 x LD(50). Twenty-four hours after the treatment, both sarin-treated and vehicle-treated (controls) animals were analyzed for: (i) plasma butyrylcholinesterase (BChE) activity; (ii) brain acetylcholinesterase (AChE) activity, (iii) m2 muscarinic acetylcholine receptor (m2 mAChR) ligand binding; (iv) blood brain barrier (BBB) permeability using [H(3)]hexamethonium iodide uptake assay and immunostaining for endothelial barrier antigen (EBA); and (v) histopathological changes in the brain using H&E staining, and microtubule-associated protein (MAP-2) and glial fibrillary acidic protein immunostaining. In animals treated with 1 x LD(50) sarin, the significant changes include a decreased plasma BChE, a decreased AChE in the cerebrum, brainstem, midbrain and the cerebellum, a decreased m2 mAChR ligand binding in the cerebrum, an increased BBB permeability in the cerebrum, brainstem, midbrain and the cerebellum associated with a decreased EBA expression, a diffuse neuronal cell death and a decreased MAP-2 expression in the cerebral cortex and the hippocampus, and degeneration of Purkinje neurons in the cerebellum. Animals treated with 0.5 x LD(50) sarin however exhibited only a few alterations, which include decreased plasma BChE, an increased BBB permeability in the midbrain and the brain stem but without a decrease in EBA expression, and degeneration of Purkinje neurons in the cerebellum. In contrast, animals treated with 0.1 and 0.01 x LD(50) did not exhibit any of the above changes. However, m2 mAChR ligand binding in the brainstem was increased after exposure to all doses of the sarin.Collectively, the above results indicate that, the early brain damage after acute exposure to sarin is clearly dose-dependent, and that exposure to 1 x LD(50) sarin induces detrimental changes in many regions of the adult rat brain as early as 24 hours after the exposure. The early neuropathological changes observed after a single dose of 1 x LD(50) sarin could lead to a profound long-term neurodegenerative changes in many regions of the brain, and resulting behavioral abnormalities.
Collapse
Affiliation(s)
- A Abdel-Rahman
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
17
|
Abstract
Brain ischemia triggers a complex cascade of molecular events that unfolds over hours to days. Identified mechanisms of postischemic neuronal injury include altered Ca(2+) homeostasis, free radical formation, mitochondrial dysfunction, protease activation, altered gene expression, and inflammation. Although many of these events are well characterized, our understanding of how they are integrated into the causal pathways of postischemic neuronal death remains incomplete. The primary goal of this review is to provide an overview of molecular injury mechanisms currently believed to be involved in postischemic neuronal death specifically highlighting their time course and potential interactions.
Collapse
Affiliation(s)
- R W Neumar
- Department of Emergency Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19107-4283, USA.
| |
Collapse
|
18
|
Hodgson DM, Taylor AN, Zhang Z, Rosenberg A. Lysosphingomyelin prevents behavioral aberrations and hippocampal neuron loss induced by the metabotropic glutamate receptor agonist quisqualate. Prog Neuropsychopharmacol Biol Psychiatry 1999; 23:877-92. [PMID: 10509381 DOI: 10.1016/s0278-5846(99)00047-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
1. Excessive excitation of brain neurons by the excitatory neurotransmitter, glutamate, induces a cascade of events leading to increased intracellular Ca++, neuronal degeneration and death. 2. Recent in vitro research has demonstrated that a natural cationic amphiphile in the brain, lysosphingomyelin, may be able to prevent neuronal degeneration by repressing phosphosinositidase-C overactivation induced by excessive excitation of the metabotropic glutamate receptor. 3. This research tested the latter finding in vivo in a rat model of glutamate excitotoxicity. Intracerebroventricular (i.c.v.) administration of the Group 1 metabotropic glutamate receptor (mGluR) agonist, quisqualate, produced seizures, akinesia, destruction of hippocampal pyramidal cell dendritic microtubule-associated protein-2, and major loss of hippocampal CA sector neurons. 4. Prophylactic i.c.v. infusion of lysosphingomyelin powerfully attenuates these quisqualate-induced behaviors and prevents neuronal degeneration. 5. Lysosphingomyelin may be of clinical use in allaying progressive Group 1 mGluR-induced hippocampal cognitive and motor disorders including Alzheimer's disease, brain seizure, and stroke.
Collapse
Affiliation(s)
- D M Hodgson
- Dept. of Neurobiology, School of Medicine, University of California, Los Angeles, USA
| | | | | | | |
Collapse
|
19
|
Morioka M, Hamada J, Ushio Y, Miyamoto E. Potential role of calcineurin for brain ischemia and traumatic injury. Prog Neurobiol 1999; 58:1-30. [PMID: 10321795 DOI: 10.1016/s0301-0082(98)00073-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Calcineurin belongs to the family of Ca2+/calmodulin-dependent protein phosphatase, protein phosphatase 2B. Calcineurin is the only protein phosphatase which is regulated by a second messenger, Ca2+. Furthermore, calcineurin is highly localized in the central nervous system, especially in those neurons vulnerable to ischemic and traumatic insults. For these reasons, calcineurin is considered to play important roles in neuron-specific functions. Recently, on the basis of the finding that FK506 and cyclosporin A serve as calcineurin-specific inhibitors, this enzyme has become the subject of much study. It is clear that calcineurin is involved in many neuronal (or non-neuronal) functions such as neurotransmitter release, regulation of receptor functions, signal transduction systems, neurite outgrowth, gene expression and neuronal cell death. In this review, we describe the calcineurin functions, functions of the substrates, and the pathogenesis of traumatic and ischemic insults, and we discuss the potential role of calcineurin. There are many similarities in traumatic and ischemic pathogenesis of the brain in which the release of excessive glutamate is followed by an intracellular Ca2+ increase. However, the intracellular cascade which leads to neuronal cell death after the release of excess Ca2+ is unclear. Although calcineurin is thought to be a key toxic enzyme on the basis of studies using immunosuppressants (FK506 or cyclosporin A), many of the functions of the substrates for calcineurin protect against neuronal cell death. We concluded that calcineurin is a bi-directional enzyme for neuronal cell death, having protective and toxic actions, and the balance of the bi-directional effects may be important in ischemic and traumatic pathogenesis.
Collapse
Affiliation(s)
- M Morioka
- Department of Neurosurgery, Kumamoto University School of Medicine, Japan.
| | | | | | | |
Collapse
|
20
|
Popa-Wagner A, Schröder E, Schmoll H, Walker LC, Kessler C. Upregulation of MAP1B and MAP2 in the rat brain after middle cerebral artery occlusion: effect of age. J Cereb Blood Flow Metab 1999; 19:425-34. [PMID: 10197512 DOI: 10.1097/00004647-199904000-00008] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Although stroke in humans usually afflicts the elderly, most experimental studies on the nature of cerebral ischemia have used young animals. This is especially important when studying restorative processes that are age dependent. To explore the potential of older animals to initiate regenerative processes after cerebral ischemia, the authors studied the expression of the juvenile-specific cytoskeletal protein, microtubule-associated protein (MAP) 1B, and the adult-specific protein, MAP2, in male Sprague-Dawley rats at 3 months and 20 months of age. The levels of MAP1B and MAP2 transcripts and the corresponding proteins declined with increasing age in the hippocampus. In the cortex, the levels of the transcripts did not change significantly with age, but the morphologic features of immunostained fibers were clearly affected by age; that is, cortical MAP1B fibers became thicker, and MAP2 fibers, more diffuse, in aged rats. Focal cerebral ischemia, produced by reversible occlusion of the right middle cerebral artery, resulted in a large decrease in the expression of both MAP1B and MAP2 in the infarct core at the messenger ribonucleic acid and protein levels. However, at 1 week after the stroke, there was vigorous expression of MAP1B and its messenger ribonucleic acid, as well as MAP2 protein, in the border zone adjacent to the infarct of 3-month-old and 20 month-old male Sprague-Dawley rats. The upregulation of these key cytologic elements generally was diminished in aged rats compared with young animals, although the morphologic features of fibers in the infarct border zone were similar in both age groups. These results suggest that the regenerative potential of the aged rat brain appears to be competent, although attenuated, at least with respect to MAP1B and MAP2 expression up to 20 months of age.
Collapse
Affiliation(s)
- A Popa-Wagner
- Department of Neurology, University of Greifswald, Germany
| | | | | | | | | |
Collapse
|
21
|
Kitagawa K, Matsumoto M, Saido TC, Ohtsuki T, Kuwabara K, Yagita Y, Mabuchi T, Yanagihara T, Hori M. Species differences in fodrin proteolysis in the ischemic brain. J Neurosci Res 1999; 55:643-9. [PMID: 10082086 DOI: 10.1002/(sici)1097-4547(19990301)55:5<643::aid-jnr11>3.0.co;2-v] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
There has been growing evidence that the breakdown of cytoskeletal proteins is an important biochemical change leading to ischemic neuronal death. In the present study, we investigated species differences in the susceptibility of fodrin to calpain activation induced by cerebral ischemia in gerbils, rats, and mice. In vivo fodrin proteolysis and degradation of microtubule-associated protein 2 after complete ischemia occurred more rapidly in the hippocampus and cerebral cortex of the gerbil brain than in the corresponding area of the rat and mouse brain. The N-methyl-D-aspartate (NMDA) receptor antagonist MK-801 injected intraperitoneally before ischemia did not diminish fodrin degradation in the gerbil hippocampus. In vivo fodrin proteolysis was inhibited at 33 degrees C and enhanced at 41 degrees C compared with proteolysis at 37 degrees C during ischemia. However, in vitro fodrin proteolysis after addition of Ca2+ into the crude membrane fraction did not show any differences among three species. Although it is highly unlikely that the difference in the sensitivity of NMDA receptor or the sensitivity of calpain activation to calcium was the crucial determinant of susceptibility of fodrin degradation in the gerbil brain, the present study clearly demonstrated that fodrin in the gerbil brain was more susceptible to calpain activation induced by ischemia than that in the rat and mouse brains. Enhanced proteolysis may be one of the reasons neurons in the gerbil brain are highly vulnerable to ischemia.
Collapse
Affiliation(s)
- K Kitagawa
- First Department of Internal Medicine, Osaka University Medical School, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Dijkhuizen RM, de Graaf RA, Tulleken KA, Nicolay K. Changes in the diffusion of water and intracellular metabolites after excitotoxic injury and global ischemia in neonatal rat brain. J Cereb Blood Flow Metab 1999; 19:341-9. [PMID: 10078886 DOI: 10.1097/00004647-199903000-00012] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The reduction of the apparent diffusion coefficient (ADC) of brain tissue water in acute cerebral ischemia, as measured by diffusion-weighted magnetic resonance imaging, is generally associated with the development of cytotoxic edema. However, the underlying mechanism is still unknown. Our aim was to elucidate diffusion changes in the intracellular environment in cytotoxic edematous tissue. The ADC of intracellular metabolites was measured by use of diffusion-weighted 1H-magnetic resonance spectroscopy after (1) unilateral N-methyl-D-aspartate (NMDA) injection and (2) cardiac arrest-induced global ischemia in neonatal rat brain. The distinct water ADC drop early after global ischemia was accompanied by a significant reduction of the ADC of all measured metabolites (P < 0.01, n = 8). In the first hours after excitotoxic injury, the ADC of water and the metabolites taurine and N-acetylaspartate dropped significantly (P < 0.05, n = 8). At 24 and 72 hours after NMDA injection brain metabolite levels were diminished and metabolite ADC approached contralateral values. Administration of the NMDA-antagonist MK-801 1.5 hours after NMDA injection completely normalized the water ADC but not the metabolite ADC after 1 to 2 hours (n = 8). No damage was detected 72 hours later and, water and metabolite ADC had normal values (n = 8). The contribution of brain temperature changes (calculated from the chemical shift between the water and N-acetylaspartate signals) and tissue deoxygenation to ischemia-induced intracellular ADC changes was minor. These data lend support to previous suggestions that the ischemia-induced brain water ADC drop may partly be caused by reduced diffusional displacement of intracellular water, possibly involving early alterations in intracellular tortuosity, cytoplasmic streaming, or intracellular molecular interactions.
Collapse
Affiliation(s)
- R M Dijkhuizen
- Department of Neurosurgery, University Hospital Utrecht, The Netherlands
| | | | | | | |
Collapse
|
23
|
Zhang Z, Davies K, Prostak J, Fenstermacher J, Chopp M. Quantitation of microvascular plasma perfusion and neuronal microtubule-associated protein in ischemic mouse brain by laser-scanning confocal microscopy. J Cereb Blood Flow Metab 1999; 19:68-78. [PMID: 9886357 DOI: 10.1097/00004647-199901000-00008] [Citation(s) in RCA: 46] [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: 11/25/2022]
Abstract
In an exposition of the technique of calculating distribution volumes from laser-scanning confocal microscopic (LSCM) data, three-dimensional images of the distribution of one or two fluorescent markers in mouse brain specimens were generated by LSCM and processed by a system developed for morphometric analysis of fixed and stained serial brain histologic samples. To determine the volume of perfused cerebral capillaries, one of two fluorescent plasma markers, either fluorescein isothiocyanate (FITC)-dextran or Evans blue, was intravenously administered to mice subjected to 1 hour of embolic middle cerebral artery (MCA) occlusion (n = 9) and to mice that were not operated on (n = 3); after 1 minute of circulation, brains were removed, immersion-fixed, and processed for LSCM. In some of these animals (n = 5), the volume of endogenous microtubule-associated protein-2 (MAP2) fluorescence was also determined using immunohistochemical staining. For mice that were not operated on, this methodology yielded highly localized volumes of (1) microvascular plasma, which agree with those determined for rodents by other techniques, and (2) MAP2 expression, which appears physiologically and morphologically reasonable. After 1 hour of MCA occlusion, the MAP2 volumes of distribution were less than 10% of normal in the ipsilateral hemisphere in which plasma perfusion essentially ceased. In conclusion, precise colocalization and quantitation of early ischemic neuronal damage and cerebral plasma perfusion deficit can be done with this three-dimensional, microphysiologic and microanatomic methodology.
Collapse
Affiliation(s)
- Z Zhang
- Department of Neurology, Henry Ford Health Sciences Center, Detroit, Michigan 48202, USA
| | | | | | | | | |
Collapse
|
24
|
Teasdale GM, Graham DI. Craniocerebral trauma: protection and retrieval of the neuronal population after injury. Neurosurgery 1998; 43:723-37; discussion 737-8. [PMID: 9766298 DOI: 10.1097/00006123-199810000-00001] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To review the consequences of mechanical injury to the brain with an emphasis on factors that may explain the variability of outcomes and how this might be influenced. METHODS Information regarding the pathophysiology of traumatic brain damage contained in original scientific reports and in review articles published in recent years was reviewed from the perspective of a clinical neurosurgeon and a neuropathologist, each with major research interests in traumatic brain damage. The information was compiled on the basis of the knowledge of and personal selection of articles that were identified through selective literature searches and current awareness profiles. A systematic literature review was not conducted. RESULTS Mechanical input affects neuronal and vascular elements and is translated into biological effects on the brain through a complex series of interacting cellular and molecular events. Whether these lead to permanent structural damage or to resolution and recovery is determined by the balance between processes that, on the one hand, mediate the effects of initial injury and subsequent secondary insults and, on the other, are manifestations of the brain's protective, reparative response. Experimental and clinical research has identified opportunities for altering the balance in a way that might promote recovery, but data demonstrating that this can lead to substantial clinical benefit are lacking. Recent evidence of genetically determined, individual susceptibility to the effects of injury may explain some of the puzzling variability in outcome after apparently similar insults and may also provide new opportunities for treatment. CONCLUSION The understanding of traumatic brain damage that is being gained from recent research is widening and broadening perspectives from the traditional focus on mechanical, vascular, and metabolic effects to encompass wider, neurobiological issues, drawn from the fields of neurodevelopment, neuroplasticity, neurodegeneration, and neurogenetics. Neurotrauma is a fascinating area of neuroscience research, with promise for the translation of knowledge to improved clinical management and outcome.
Collapse
Affiliation(s)
- G M Teasdale
- Institute of Neurological Sciences, Southern General Hospital NHS Trust, Glasgow, Scotland
| | | |
Collapse
|
25
|
Neumar RW, DeGracia DJ, Konkoly LL, Khoury JI, White BC, Krause GS. Calpain mediates eukaryotic initiation factor 4G degradation during global brain ischemia. J Cereb Blood Flow Metab 1998; 18:876-81. [PMID: 9701348 DOI: 10.1097/00004647-199808000-00007] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Global brain ischemia and reperfusion result in the degradation of the eukaryotic initiation factor (eIF) 4G, which plays a critical role in the attachment of the mRNA to the ribosome. Because eIF-4G is a substrate of calpain, these studies were undertaken to examine whether calpain I activation during global brain ischemia contributes to the degradation of eIF-4G in vivo. Immunoblots with antibodies against calpain I and eIF-4G were prepared from rat brain postmitochondrial supernatant incubated at 37 degrees C with and without the addition of calcium and the calpain inhibitors calpastatin or MDL-28,170. Addition of calcium alone resulted in calpain I activation (as measured by autolysis of the 80-kDa subunit) and degradation of eIF-4G; this effect was blocked by either 1 micromol/L calpastatin or 10 micromol/L MDL-28,170. In rabbits subjected to 20 minutes of cardiac arrest, immunoblots of brain postmitochondrial supernatants showed that the percentage of autolyzed calpain I increased from 1.9% +/- 1.1% to 15.8% +/- 5.0% and that this was accompanied by a 68% loss of eIF-4G. MDL-28,170 pretreatment (30 mg/kg) decreased ischemia-induced calpain I autolysis 40% and almost completely blocked eIF-4G degradation. We conclude that calpain I degrades eIF-4G during global brain ischemia.
Collapse
Affiliation(s)
- R W Neumar
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | | | | |
Collapse
|
26
|
Folkerts MM, Berman RF, Muizelaar JP, Rafols JA. Disruption of MAP-2 immunostaining in rat hippocampus after traumatic brain injury. J Neurotrauma 1998; 15:349-63. [PMID: 9605349 DOI: 10.1089/neu.1998.15.349] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The effects of diffuse brain injury on dendritic morphology in rat hippocampus and cortex were examined in this study using the recently described impact acceleration model of traumatic brain injury (Marmarou et al., 1994). Dendritic structure was visualized using immunostaining of microtubule associated protein-2 (MAP-2). Brains were studied 24, 48, and 72 h after brain injury. Results from immunohistochemistry and light microscopy indicated a time-dependent disruption of dendritic cytoarchitecture in the CA1 subregion and in the hilus of the hippocampus but not in the dentate gyrus or CA3 subregion. Similar disruption was observed in the cortical mantle overlying the hippocampus. Although disruption of dendritic structure was observed at 24 h, the most severe damage was at 48 h after injury with evidence of at least partial recovery of MAP-2 immunostaining by 72 h. In the most severe damage, dendrites appeared to be fragmented, scattered, and unaligned, consisting of irregularly spaced and darkly stained swollen segments. A mixed pattern of immunostaining was observed in somata of hilar cells, with some appearing normal while others stained only faintly, appearing to have lost their typical polygonal shape. Semiquantitative rankings confirmed these qualitative findings. Immediate post-injury behavioral evaluations of injury severity were compared to the degree of disruption of MAP-2 immunostaining. The results of this study indicate that diffuse brain injury is associated not only with axonal damage but also with injury to dendrites.
Collapse
Affiliation(s)
- M M Folkerts
- University of California, Davis, Neurotrauma Research Laboratories, Department of Neurological Surgery, 95616, USA
| | | | | | | |
Collapse
|
27
|
Iijima T, Shimase C, Sawa H, Sankawa H. Spreading depression induces depletion of MAP2 in area CA3 of the hippocampus in a rat unilateral carotid artery occlusion model. J Neurotrauma 1998; 15:277-84. [PMID: 9555973 DOI: 10.1089/neu.1998.15.277] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Traumatic brain injury (TBI) induces neuronal cell loss in area CA3 of the hippocampus. However, it has not yet been established why traumatic injury of the cortex induces neuronal damage in more remote areas. Spreading depression (SD) may be one potential mechanism for this pathophysiology. The present study evaluated whether SD on the cortex evokes a pathological change in the hippocampus. Forty-two Fisher rats were assigned to four groups: Group I: sham operation (n = 7), Group II: right carotid occlusion (UO) for 7 days (n = 7), Group III: repeated induction of SD by KCl application on dura for 7 days (n = 7), Group III' for 3 h (n = 7), Group IV: SD induction and UO for 7 days (n = 14) Group IV' for 3 h (n = 7). In 5 out of 7 animals in Groups III' and IV', cerebral blood flow (CBF) was monitored using laser Doppler flowmetry for 3 h during the passage of SD. The brains were processed for immunohistochemical analysis of microtubule-associated protein 2 (MAP2). Reactive hyperemia induced by SD was not significantly suppressed by right carotid occlusion (194 +/- 25% and 181 +/- 42% UO in Groups III and IV, respectively). In 6 out of 7 animals in a 7-day model of Group IV, and 3 animals in a 7-day model of Group III, MAP2 depletion in the CA3 area of the hippocampus (partly including CA2) was observed, although no change in the hippocampus was observed in other groups. In conclusion, SD in combination with UO yielded reproducible lesions in CA3. Neuronal injury in the hippocampus after brain trauma may be attributable to SD in combination with the blood flow restriction.
Collapse
Affiliation(s)
- T Iijima
- Department of Anesthesiology, Kyorin University School of Medicine, Mitaka City, Tokyo, Japan
| | | | | | | |
Collapse
|
28
|
Abstract
Growth inhibitory factor (GIF) is a small (7 kDa), heat-stable, acidic, hydrophilic metallothionein (MT)-like protein. GIF inhibits the neurotrophic activity in Alzheimer's disease (AD) brain extracts on neonatal rat cortical neurons in culture. GIF has been shown to be drastically reduced and down-regulated in AD brains. In neurodegenerative diseases in humans, GIF expression levels are reduced whereas GFAP expression levels are markedly induced in reactive astrocytes. Both GIF and GIF mRNA are present at high levels in reactive astrocytes following acute experimental brain injury. In chronological observations the level of GIF was found to increase more slowly and remain elevated for longer periods than that of glial fibrillary acidic protein (GFAP). These differential patterns and distribution of GIF and GFAP seem to be important in understanding the mechanism of brain tissue repair. The most important point concerning GIF in AD is not simply the decrease in the level of expression throughout the brain, but the drastic decrease in the level of expression in reactive astrocytes around senile plaques in AD. Although what makes the level of GIF decrease drastically in reactive astrocytes in AD is still unknown, supplements of GIF may be effective for AD, based on a review of current evidence. The processes of tissue repair following acute brain injury are considered to be different from those in AD from the viewpoint of reactive astrocytes.
Collapse
Affiliation(s)
- I Hozumi
- Department of Neurology, Brain Research Institute, Niigata University, Niigata City, Japan
| | | | | |
Collapse
|
29
|
Matsushita K, Matsuyama T, Kitagawa K, Matsumoto M, Yanagihara T, Sugita M. Alterations of Bcl-2 family proteins precede cytoskeletal proteolysis in the penumbra, but not in infarct centres following focal cerebral ischemia in mice. Neuroscience 1998; 83:439-48. [PMID: 9460752 DOI: 10.1016/s0306-4522(97)00391-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Apoptosis has drawn attention in ischemic neuronal death recently. However, studies of apoptosis in cerebral ischemia have concentrated largely in DNA fragmentation, a late phase in apoptotic nuclei, at the expense of possible primary ischemic targets at the subcellular level and of upstream apoptotic signalling. To assess those issues, we used an intraluminal middle cerebral artery occlusion model in mice with or without reperfusion, and examined sequential changes of Bcl-2 family proteins modulating apoptotic signalling immunohistochemically and studied nuclear DNA fragmentation, to compare their chronology in relation to the development of infarct as detected by loss of microtubule-associated protein-2, an early marker of cytoplasmic damage. In the centre of the lesion, Bax protein increased and Bcl-2 and Bcl-x proteins decreased after loss of microtubule-associated protein-2 antigenicity occurred, but at the border of the lesion, the former changes preceded loss of microtubule-associated protein-2 antigenicity. Additionally, close morphologic analysis of DNA fragmentation in situ indicated that transient ischemia predominantly induced apoptotic cells but permanent ischemia produced necrosis of cells in the centre of the lesion. The contrasting cell death mechanisms, apoptosis and necrosis, are selectively involved in the pathology of cerebral ischemia, depending on its severity.
Collapse
Affiliation(s)
- K Matsushita
- Fifth Department of Internal Medicine, Hyogo College of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Sugaya T, Kitani Y. Nitrous oxide attenuates the protective effect of isoflurane on microtubule-associated protein2 degradation during forebrain ischemia in the rat. Brain Res Bull 1997; 44:307-9. [PMID: 9323446 DOI: 10.1016/s0361-9230(97)00142-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Recently, attention has been focused on the degradation of cytoskeletal proteins in animal models of cerebral ischemia, as the collapse of cytoskeletal proteins may be closely related to cytoskeletal disintegration and ultimate neuronal cell death. Among these proteins, microtubule-associated protein 2 (MAP2) has been shown to be highly vulnerable to ischemic injuries. To determine the degree of anesthetic effect on the collapse of cytoskeletal proteins, we compared the effect of three inhalation anesthetics; isoflurane, halothane, and nitrous oxide (N2O), on MAP2 degradation during 20 min of forebrain ischemia in the rat. Under equipotent anesthesia, forebrain ischemia was induced by the occlusion of the bilateral common carotid artery (CCA) combined with a lowering of mean arterial pressure (mAP) to 50 mmHg. After 20 min of ischemia, three regions of the brain, the frontoparietal cortex, brainstem, and hippocampus, were removed and separately homogenized. Subsequently, MAP2 of each region was measured using an enzyme-linked immunosorbent assay (ELISA). In the frontoparietal cortex and hippocampus, MAP2 was significantly protected from degradation when isoflurane was used combined with nitrogen (N2). However, the protective effects of isoflurane were drastically reduced when N2O was given instead of N2. These results suggest that the use of N2O should be discontinued when severe cerebral ischemia is accidentally incurred during anesthetic management.
Collapse
Affiliation(s)
- T Sugaya
- Department of Anesthesiology and Reanimatology, Gunma University School of Medicine, Japan
| | | |
Collapse
|
31
|
Blomgren K, McRae A, Elmered A, Bona E, Kawashima S, Saido TC, Ono T, Hagberg H. The calpain proteolytic system in neonatal hypoxic-ischemia. Ann N Y Acad Sci 1997; 825:104-19. [PMID: 9369979 DOI: 10.1111/j.1749-6632.1997.tb48420.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neonatal rats were subjected to transient cerebral hypoxic-ischemia (HI, unilateral occlusion of the common carotid artery +7.70% O2 for 100 min) and allowed to recover for up to 14 days. Calpain caseinolytic activity was found to increase in both hemispheres for at least 20 hr. Hypoxic exposure per se increased the activity of calpains, more pronounced in a membrane-associated fraction, probably through interaction with cellular components, whereas HI introduced a loss of activity, most likely through consumption and loss of proteases. Consecutive tissue sections were stained with antibodies against calpastatin, alpha-fodrin, the 150-kDa breakdown product of alpha-fodrin (FBDP, marker of calpain proteolysis) or microtubule-associated protein 2 (MAP-2, marker of dendrosomatic neuronal injury). Areas with brain injury displayed a distinct loss of MAP-2, which clearly delineated the infarct. FBDP accumulated in injured and borderline regions ipsilaterally, and a less conspicuous, transient increase in FBDP also occurred in the contralateral hemisphere, especially in the white matter. The cytosolic fraction (CF) and the membrane and microsomal fraction (MMF) of cortical tissue were subjected to Western blotting and stained with antibodies against calpain, calpastatin and the 150-kDa breakdown product of alpha-fodrin (FBDP). Calpain immunoreactivity decreased bilaterally in the CF during the insult (62-68% of controls) and remained significantly lower during early recovery, whereas the MMF showed no significant changes. This translocation of calpains coincided with the appearance of FBDP in the ipsilateral, HI hemisphere, displaying a significantly higher level of FBDP from immediately after the insult until at least 1 day of recovery (204-292% of controls). No significant changes in FBDP were found in the contralateral, undamaged hemisphere, despite translocation of calpains in both hemispheres, a prerequisite for calpain activation. This discrepancy may be related to changes in the endogenous inhibitor, calpastatin. Calpastatin protein was found to decrease during and shortly after HI in the ipsilateral, but not the contralateral, hemisphere. The inhibitory activity of calpastatin also tended to decrease after HI, indicating that a reduction of calpastatin may be necessary for extensive calpain activation to occur. The mRNA of m-calpain increased in the HI hemisphere 48 hr after the insult (167%, p < 0.001), a time point when the protein was also increased. In summary, our findings indicate that calpains are activated during HI and in the early phase of reperfusion after HI, preceding neuronal death.
Collapse
Affiliation(s)
- K Blomgren
- Institute of Anatomy and Cell Biology, University of Göteborg, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Lee KS, Yanamoto H, Fergus A, Hong SC, Kang SD, Cappelletto B, Toyoda T, Kassell NF, Bavbek M, Kwan AL. Calcium-activated proteolysis as a therapeutic target in cerebrovascular disease. Ann N Y Acad Sci 1997; 825:95-103. [PMID: 9369978 DOI: 10.1111/j.1749-6632.1997.tb48419.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- K S Lee
- Department of Neurological Surgery, University of Virginia, Charlottesville 22908, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kuramoto H, Hozumi I, Inuzuka T, Sato S. Occurrence of myelin-associated glycoprotein (MAG)-like immunoreactivity in some nervous, endocrine, and immune-related cells of the rat. An immunohistochemical study. MOLECULAR AND CHEMICAL NEUROPATHOLOGY 1997; 31:85-94. [PMID: 9271008 DOI: 10.1007/bf02815163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The occurrence and distribution of myelin-associated glycoprotein (MAG)-like immunoreactivity was investigated in the rat using a polyclonal antibody to MAG purified from rat brain. In the nervous system, MAG immunoreactivity was found in the periaxonal portion of the myelinated fibers and in a small number of oligodendroglia in the cortex, hippocampus, and the spinal cord. The sheath of Schwann cells in unmyelinated fibers and satellite cells in the spinal ganglia were also immunoreactive for MAG. In the endocrine system, the noradrenaline-containing cells in the adrenal medulla and some endocrine cells in the duodenum showed MAG immunoreactivity. In the immune system, numerous reticular cells with slender cytoplasmic processes, which formed a dense network, were immunopositive for MAG within the germinal center in the lymph nodes and spleen. In the thymus, a number of epithelial reticular cells within the medulla showed variation in staining intensity. These findings provide new information on the wide distribution of MAG immunoreactivity in the nervous, endocrine, and immune systems, and may contribute to the further understanding of the biological roles of this protein.
Collapse
Affiliation(s)
- H Kuramoto
- Department of Anatomy, Niigata University School of Medicine, Japan
| | | | | | | |
Collapse
|
34
|
Posmantur R, Kampfl A, Siman R, Liu J, Zhao X, Clifton GL, Hayes RL. A calpain inhibitor attenuates cortical cytoskeletal protein loss after experimental traumatic brain injury in the rat. Neuroscience 1997; 77:875-88. [PMID: 9070759 DOI: 10.1016/s0306-4522(96)00483-6] [Citation(s) in RCA: 142] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The capacity of a calpain inhibitor to reduce losses of neurofilament 200-, neurofilament 68- and calpain 1-mediated spectrin breakdown products was examined following traumatic brain injury in the rat. Twenty-four hours after unilateral cortical impact injury, western blot analyses detected neurofilament 200 losses of 65% (ipsilateral) and 36% (contralateral) of levels observed in naive, uninjured rat cortices. Neurofilament 68 protein levels decreased only in the ipsilateral cortex by 35% relative to naive protein levels. Calpain inhibitor 2, administered 10 min after injury via continuous arterial infusion into the right external carotid artery for 24 h, significantly reduced neurofilament 200 losses to 17% and 3% relative to naive neurofilament 200 protein levels in the ipsilateral and contralateral cortices, respectively. Calpain inhibitor administration abolished neurofilament 68 loss in the ipsilateral cortex and was accompanied by a reduction of putative calpain-mediated neurofilament 68 breakdown products. Spectrin breakdown products mediated by calpain 1 activation were detectable in both hemispheres 24 h after traumatic brain injury and were substantially reduced in animals treated with calpain inhibitor 2 both ipsilaterally and contralaterally to the site of injury. Qualitative immunofluorescence studies of neurofilament 200 and neurofilament 68 confirmed western blot data, demonstrating morphological protection of neuronal structure throughout cortical regions of the traumatically injured brain. Morphological protection included preservation of dendritic structure and reduction of axonal retraction balls. In addition, histopathological studies employing hematoxylin and eosin staining indicated reduced extent of contusion at the injury site. These data indicate that calpain inhibitors could represent a viable strategy for preserving the cytoskeletal structure of injured neurons after experimental traumatic brain injury in vivo.
Collapse
Affiliation(s)
- R Posmantur
- Department of Neurosurgery, University of Texas Houston Health Science Center, 77030, U.S.A
| | | | | | | | | | | | | |
Collapse
|
35
|
Kampfl A, Posmantur RM, Zhao X, Schmutzhard E, Clifton GL, Hayes RL. Mechanisms of calpain proteolysis following traumatic brain injury: implications for pathology and therapy: implications for pathology and therapy: a review and update. J Neurotrauma 1997; 14:121-34. [PMID: 9104930 DOI: 10.1089/neu.1997.14.121] [Citation(s) in RCA: 156] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Much recent research has focused on the pathological significance of calcium accumulation in the central nervous system (CNS) following cerebral ischemia, spinal cord injury (SCI), and traumatic brain injury (TBI). Disturbances in neuronal calcium homeostasis may result in the activation of several calcium-sensitive enzymes, including lipases, kinases, phosphatases, and proteases. One potential pathogenic event in a number of acute CNS insults, including TBI, is the activation of the calpains, calcium-activated intracellular proteases. This article reviews new evidence indicating that overactivation of calpains plays a major role in the neurodegenerative cascade following TBI in vivo. Further, this article presents an overview from in vivo and in vitro models of CNS injuries suggesting that administration of calpain inhibitors during the initial 24-h period following injury can attenuate injury-induced derangements of neuronal structure and function. Lastly, this review addresses the potential contribution of other proteases to neuronal damage following TBI.
Collapse
Affiliation(s)
- A Kampfl
- Department of Neurology, University of Innsbruck, Austria
| | | | | | | | | | | |
Collapse
|
36
|
Morimoto T, Ginsberg MD, Dietrich WD, Zhao W. Hyperthermia enhances spectrin breakdown in transient focal cerebral ischemia. Brain Res 1997; 746:43-51. [PMID: 9037482 DOI: 10.1016/s0006-8993(96)01154-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Calpain-mediated spectrin degradation is triggered by cerebral ischemia and, when persistent, is thought to signal irreversible neuronal injury. Hyperthermia superimposed upon cerebral ischemia may exacerbate the injury process. In this study, we compared the extent of spectrin degradation in the brains of rats subjected to 1 h of transient proximal middle cerebral artery (MCA) clip-occlusion performed under conditions of cranial normothermia (37 degrees C) or mild cranial hyperthermia (39 degrees C). Immunocytochemical localization of spectrin breakdown products was achieved by the use of a rabbit polyclonal antibody which reacted selectively with calpain-generated fragments of brain spectrin. The perfusion times studied were 1, 4 or 24 h. Following normothermic MCA occlusion, spectrin immunoreactivity was present only occasionally and only in scattered cortical neurons immediately upon reperfusion and 1 h later; all normothermic brains showed space immunoreactivity at 4 h of reperfusion; and no immunoreactivity was detected at 24 h. By contrast, following hyperthermic MCA occlusion, moderate-to-intense immunostaining was present in cortical pyramidal neurons even immediately upon reperfusion and persisted at 1 h of reperfusion. At 4 and 24 h, most brains exhibited dense immunoreactivity associated with morphologically shrunken neurons. Following 24 h survival, semi-thick plastic sections revealed intact neuropil and only selective neuronal necrosis in normothermic rats. By contrast, pan-necrosis was evident 24 h after the hyperthermic ischemic insult. These results indicate that mild cranial hyperthermia superimposed upon transient focal ischemia markedly enhances calpain activation and spectrin degradation; this process appears to be an important mechanism by which hyperthermia exacerbates ischemic injury.
Collapse
Affiliation(s)
- T Morimoto
- Department of Neurology, University of Miami School of Medicine, FL 33101, USA
| | | | | | | |
Collapse
|
37
|
Pettigrew LC, Holtz ML, Craddock SD, Minger SL, Hall N, Geddes JW. Microtubular proteolysis in focal cerebral ischemia. J Cereb Blood Flow Metab 1996; 16:1189-202. [PMID: 8898691 DOI: 10.1097/00004647-199611000-00013] [Citation(s) in RCA: 111] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Calpain, a neutral protease activated by calcium, may promote microtubular proteolysis in ischemic brain. We tested this hypothesis in an animal model of focal cerebral ischemia without reperfusion. The earliest sign of tissue injury was observed after no more than 15 min of ischemia, with coiling of apical dendrites immunolabeled to show microtubule-associated protein 2 (MAP2). After 6 h of ischemia, MAP2 immunoreactivity was markedly diminished in the infarct zone. Quantitative Western analysis demonstrated that MAP2 was almost unmeasurable after 24 h of ischemia. An increase in calpain activity, shown by an antibody recognizing calpain-cleaved spectrin fragments, paralleled the loss of MAP2 immunostaining. Double-labeled immunofluorescent studies showed that intraneuronal calpain activity preceded evidence of MAP2 proteolysis. Perikaryal immunolabeling of tau protein became increasingly prominent between 1 and 6 h in neurons located within the transition zone between ischemic and unaffected tissue. Western blot experiments confirmed that dephosphorylation of tau protein occurred during 24 h of ischemia, but was not associated with significant loss of tau antigen. We conclude that focal cerebral ischemia is associated with early microtubular proteolysis caused by calpain.
Collapse
Affiliation(s)
- L C Pettigrew
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington 40536-0230, USA
| | | | | | | | | | | |
Collapse
|
38
|
Lewén A, Li GL, Olsson Y, Hillered L. Changes in microtubule-associated protein 2 and amyloid precursor protein immunoreactivity following traumatic brain injury in rat: influence of MK-801 treatment. Brain Res 1996; 719:161-71. [PMID: 8782876 DOI: 10.1016/0006-8993(96)00081-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We investigated by immunohistochemistry dendritic and axonal changes occurring in the rat brain after mild focal cortical trauma produced by the weight drop technique. One and 3 days after injury, nerve cell bodies and dendrites in the perimeter of the impact site displayed decreased microtubule-associated protein 2 (MAP2) immunoreactivity. Some dendrites in the immediate adjacent region were more intensely stained and distorted. The dentate hilar region of the hippocampus showed a reduction of immunoreactive nerve cell bodies and dendrites. Twenty-one days after injury the strongly stained cortical dendrites and the reduction of immunoreactivity in the hippocampus remained, whereas the reduced staining in the perimeter of the lesion had normalised. These results indicate that there is a long-lasting disturbed dendritic organisation implicating impaired neurotransmission after this type of mild brain trauma. beta-Amyloid precursor protein (APP) immunohistochemistry revealed numerous stained axons in the ipsilateral subcortical white matter and thalamus indicating local and remote axonal injuries with disturbed axonal transport. Twenty-one days after injury, numerous small immunostained profiles appeared in the neuropil of the cortical impact site and in the ipsilateral thalamus. The axonal changes indicate disturbed connectivity between the site of the impact and other brain regions, chiefly the thalamus. The presence of beta-amyloid was investigated 21 days after trauma. There were no signs of beta-amyloid depositions in the brain after injury. Finally, we tested if the non-competitive NMDA receptor antagonist dizocilpine maleate (MK-801) could influence the observed MAP2 and APP changes. Pretreatment with this compound did not affect the early MAP2 and APP alterations. Instead, an increased expression of the APP antigen in the thalamus was observed 21 days after trauma in the MK-801-treated animals. The cause of this phenomenon is not known but may be related to a delayed neurotoxic action of MK-801 treatment.
Collapse
Affiliation(s)
- A Lewén
- Department of Neurosurgery, Uppsala University Hospital, Sweden.
| | | | | | | |
Collapse
|
39
|
Inuzuka T, Hozumi I, Tamura A, Hiraiwa M, Tsuji S. Patterns of growth inhibitory factor (GIF) and glial fibrillary acidic protein relative level changes differ following left middle cerebral artery occlusion in rats. Brain Res 1996; 709:151-31. [PMID: 8869568 DOI: 10.1016/0006-8993(95)01444-6] [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: 02/02/2023]
Abstract
Growth inhibitory factor (GIF) has been identified as a new metallothionein-like protein, the level of which is decreased in the Alzheimer's disease brain. GIF and glial fibrillary acidic protein (GFAP) have been reported to be expressed in reactive astrocytes in the rat brain following stab wounds. Moreover, strong expression of GIF mRNA in reactive astrocytes after ventricular injection of kainic acid has been demonstrated. To clarify the biological functions of GIF and GFAP in repair of the CNS, we examined changes in their relative levels to sham control using a Western blotting technique in the rat left hemisphere following occlusion of the left middle cerebral artery, for 28 days after surgery. The GIF relative level declined to 56% of the sham-operated control value on day 7. Thereafter the GIF relative level increased and returned to the normal relative level by days 21-28. The GFAP relative level increased from day 3 and reached a maximum of 120% of the sham-operated control value on days 14-21. While GIF and GFAP were both detected in reactive astrocytes, an increase in the GFAP relative level occurred prior to an increase in GIF relative level following the ischemia. The patterns of changes in relative expression levels of GIF and GFAP were quite similar to those in our previous studies on effects of cerebral stab wounds in rats, although the changes were more rapid in the previous studies. GIF and GFAP appear to play different roles in the repair of the CNS. The present results also indicated that GIF could play an important role in CNS repair after cerebral ischemia and provide new insights into the mechanism of gliosis investigated mainly from the viewpoint of GFAP.
Collapse
Affiliation(s)
- T Inuzuka
- Department of Neurology, Niigata University, Japan
| | | | | | | | | |
Collapse
|
40
|
Dawson DA, Hallenbeck JM. Acute focal ischemia-induced alterations in MAP2 immunostaining: description of temporal changes and utilization as a marker for volumetric assessment of acute brain injury. J Cereb Blood Flow Metab 1996; 16:170-4. [PMID: 8530550 DOI: 10.1097/00004647-199601000-00020] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The utility of microtubule-associated protein 2 (MAP2) immunostaining as a marker of acute focal ischemic injury was investigated. Permanent middle cerebral artery occlusion (MCAO) elicited a rapid reduction in MAP2 immunostaining that was visible 1 h post-MCAO and that increased in intensity and area encompassed over time. The ischemic lesion borders were well defined by loss of MAP2 immunostaining, but alterations in staining within the lesion were more heterogeneous. Lesion volume increased significantly from 1 to 4 h post-MCAO (from 63.8 +/- 10.8 to 111.3 +/- 19.0 mm3, mean +/- SD). Thus, MAP2 immunostaining is a sensitive, quantifiable indicator of acute brain injury following focal ischemia.
Collapse
Affiliation(s)
- D A Dawson
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
41
|
Okada M, Nakanishi H, Tamura A, Urae A, Mine K, Yamamoto K, Fujiwara M. Long-term spatial cognitive impairment after middle cerebral artery occlusion in rats: no involvement of the hippocampus. J Cereb Blood Flow Metab 1995; 15:1012-21. [PMID: 7593333 DOI: 10.1038/jcbfm.1995.127] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The behavioral and neurochemical changes in the chronic phase of permanent occlusion of the right middle cerebral artery (MCA) in rats were investigated. One month after MCA occlusion, 23 rats were unable to solve a radial eight-arm maze task during an entire 1-month period, whereas seven rats were able to solve this task. Three months after occlusion, 19 MCA-occluded rats failed to solve the task successfully again for at least 1 month (the cognitively impaired rats), whereas 11 MCA-occluded rats were able to solve it (the cognitively unimpaired rats). The rats that underwent behavioral testing were examined for any changes in the acetylcholine (ACh) levels in the hippocampus using HPLC with electrochemical detection or the formation of long-term potentiation (LTP) in the population spike of the hippocampal CA1 field. The immunohistochemical distribution of either the microtubule-associated protein 2 (MAP2) or glial fibrillary acidic protein (GFAP) in the hippocampus of the cognitively impaired rats was also studied. In the cognitively impaired rats, neither the suppression of the induction of LTP, nor the degradation of MAP2, nor the increase in the GFAP immunoreactivity was observed in the hippocampus. The levels of ACh in the hippocampus did not change significantly among the cognitively impaired, unimpaired, and the sham-operated rats. These results suggest that MCA occlusion is capable of producing long-term spatial cognitive disturbance in rats without any evidence of neurobiological damage in the hippocampus.
Collapse
Affiliation(s)
- M Okada
- Department of Physiology and Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Hayes RL, Yang K, Whitson JS, Postmantur R. Cytoskeletal derangements following central nervous system injury: modulation by neurotrophic gene transfection. J Neurotrauma 1995; 12:933-41. [PMID: 8594223 DOI: 10.1089/neu.1995.12.933] [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: 01/31/2023] Open
Abstract
This paper reviews important new evidence indicating that traumatic brain injury can produce more widespread derangements to the neuronal cytoskeleton than previously recognized. Although cytoskeletal derangements in axons have long been documented, recent data suggest that traumatic brain injury can produce structural derangements to dendrites and cell bodies as well. Many of these investigations have employed in vivo models to provide important insights into mechanisms possibly mediating the acute loss of cytoskeletal proteins, including disturbances in calcium homeostasis and activation of calcium-dependent proteolytic enzymes. However, we have little understanding of processes mediating the recovery of cytoskeletal proteins following injury. This paper provides recent evidence from in vitro models of central nervous system injury that neurotrophic proteins can enhance the recovery of the neuronal cytoskeleton. Neurotrophin-based therapy could employ either administration of exogenous neurotrophic proteins and/or transfection of cDNA for appropriate neurotrophins.
Collapse
Affiliation(s)
- R L Hayes
- Department of Neurosurgery, University of Texas Houston Health Science Center, 77030, USA
| | | | | | | |
Collapse
|
43
|
Blomgren K, McRae A, Bona E, Saido TC, Karlsson JO, Hagberg H. Degradation of fodrin and MAP 2 after neonatal cerebral hypoxic-ischemia. Brain Res 1995; 684:136-42. [PMID: 7583214 DOI: 10.1016/0006-8993(95)00398-a] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Neonatal rats were subjected to transient cerebral hypoxic-ischemia (unilateral occlusion of the common carotid artery + 7.70% O2 for 100 min) and allowed to recover for 3 h, 24 h, 2 days or 14 days. Consecutive tissue sections were stained with antibodies against alpha-fodrin, the 150 kDa breakdown product of alpha-fodrin (FBDP, marker of calpain proteolysis) or microtubule associated protein 2 (MAP 2, marker of dendrosomatic neuronal injury). Cortical tissue pieces were subjected to Western blotting using the antibody against the FBDP. Areas with brain injury displayed a distinct loss of MAP 2 which clearly delineated the infarct. FBDP accumulated in injured and borderline regions ipsilaterally and a less conspicuous, transient increase in FBDP also occurred in the contralateral hemisphere, especially in the white matter. A reciprocal staining pattern could be seen in the cerebral cortex, i.e. loss of MAP 2 and accumulation of FBDP, most pronounced 14 days after the insult. Fodrin and MAP 2 are known calpain substrates, and degradation of these proteins preceded neuronal degeneration, indicating that these proteases may be involved in the early events triggering the cascades leading to neuronal death.
Collapse
Affiliation(s)
- K Blomgren
- Institute of Anatomy and Cell Biology, Göteborg University, Sweden
| | | | | | | | | | | |
Collapse
|
44
|
Zhou Q, Abe H, Nowak TS. Immunocytochemical and in situ hybridization approaches to the optimization of brain slice preparations. J Neurosci Methods 1995; 59:85-92. [PMID: 7475255 DOI: 10.1016/0165-0270(94)00197-o] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Methods are described for determining the expression of specific mRNAs and proteins in brain slices, in order to elucidate changes in gene expression during preparation of vibratome slices from hippocampus of adult rats. In situ hybridization with 35S-labeled oligonucleotides was used to evaluate the level and distribution of c-fos and hsp72 mRNAs in 15-microns frozen sections prepared from these slices. Commercially available antibodies were used to examine the distribution of induced Fos and Jun proto-oncogenes as well as expression of the neuronal cytoskeletal protein, microtubule-associated protein 2 (MAP2), in 50-microns vibratome sections from immersion-fixed slices. These studies confirm the induction of c-fos and hsp72 mRNAs during routine incubation, as previously observed in hippocampal slices obtained with a tissue chopper and incubated under somewhat different conditions, indicating that such responses are likely to be common features of many slice preparations. Accumulation of Fos and Jun immunoreactivities in neurons and glia was generally consistent with the distribution of c-fos mRNA induction observed in slices, and the neuronal component of this response was comparable to the expression of these proteins observed after transient ischemia in vivo. MAP2 immunoreactivity detected in the dendritic processes of neurons tended to show an increase in staining intensity during slice incubation, although loss of dendritic staining in specific regions was occasionally observed in association with the absence of Fos and Jun expression and histological evidence of neuron damage. These results support the use of MAP2 immunoreactivity as a sensitive indicator of neuronal integrity in slices.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- Q Zhou
- Department of Neurology, University of Tennessee, Memphis 38163, USA
| | | | | |
Collapse
|
45
|
Kumar K, Wu XL. Expression of beta-actin and alpha-tubulin mRNA in gerbil brain following transient ischemia and reperfusion up to 1 month. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1995; 30:149-57. [PMID: 7609636 DOI: 10.1016/0169-328x(94)00286-n] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The time course of mRNA expressions of two cytoskeletal proteins, beta-actin and alpha-tubulin, was studied by Northern blot analysis and in situ hybridization in the same gerbil brains at various periods of recirculation following 10 min of forebrain ischemia. On Northern blot analysis, beta-actin mRNA in the forebrain showed increase after 6 h and 24 h recirculation. There was wide variation in its expression 3 days postischemia (PI), and by 7 days PI it had returned to control. The alpha-tubulin mRNA in the forebrain was shown to be reduced 6 h PI in our previous study. In the present analysis of Northern blots of delayed postischemic periods, there was no significant change in its expression even though there were variations. In situ hybridization revealed a decline in the mRNA expressions of both alpha-tubulin and beta-actin in the CA1 region as early as 6-24 h PI with the reductions being prominent at 3 days PI. By 7 days PI, beta-actin was only faintly visible while alpha-tubulin was completely absent in the CA1 region. Neither RNA was detectable in CA1 1 month PI. The heat shock-70 protein was expressed by 1 h PI, and it continued to be expressed up to 24 h, returning to control by 3 days PI. These results indicate that ischemia inhibits mRNA expressions of cytoskeletal protein in the selectively vulnerable region of the brain, i.e. CA1. The time course of the reduction of the two mRNAs coincides with delayed neuronal death suggesting that the cytoskeletal proteins may play important roles in selective postischemic neuronal injury.
Collapse
Affiliation(s)
- K Kumar
- Department of Pathology, Michigan State University, East Lansing 48824, USA
| | | |
Collapse
|
46
|
Hicks RR, Smith DH, McIntosh TK. Temporal response and effects of excitatory amino acid antagonism on microtubule-associated protein 2 immunoreactivity following experimental brain injury in rats. Brain Res 1995; 678:151-60. [PMID: 7620884 DOI: 10.1016/0006-8993(95)00179-t] [Citation(s) in RCA: 83] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Alterations in microtubule-associated protein 2 (MAP2) immunoreactivity following lateral fluid-percussion (FP) brain injury were investigated in rats with survival times ranging between 10 min and 7 days. MAP2 immunoreactivity was profoundly diminished in the cortex and hippocampus ipsilateral to the site of injury by 10 min and remained diminished up to 7 days after injury. Nissl staining and silver impregnation histochemistry demonstrated a correlation between the loss of MAP2 and neuronal degeneration. The effect of excitatory amino acid receptor antagonism on MAP2 immunoreactivity was evaluated by administering kynurenate or buffer 15 min after FP injury. Administration of kynurenate significantly attenuated the loss of MAP2 observed in the cortex two weeks after injury when compared to buffer treated control animals (P < 0.02). We conclude that significant and prolonged cytoskeletal changes occur following lateral FP brain injury, and that these alterations can be attenuated by blocking excitatory amino acid receptors.
Collapse
Affiliation(s)
- R R Hicks
- University of Kentucky, Division of Physical Therapy, Lexington 40536-0079, USA
| | | | | |
Collapse
|
47
|
Saito N, Kawai K, Nowak TS. Reexpression of developmentally regulated MAP2c mRNA after ischemia: colocalization with hsp72 mRNA in vulnerable neurons. J Cereb Blood Flow Metab 1995; 15:205-15. [PMID: 7860654 DOI: 10.1038/jcbfm.1995.26] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Levels of mRNAs encoding the microtubule-associated proteins MAP2b and MAP2c as well as the 70-kDa stress protein [72-kDa heat shock protein (hsp72)] were evaluated in postischemic rat brain by in situ hybridization with oligonucleotide probes corresponding to the known rat sequences. Rats were subjected to 10-min cardiac arrest, produced by compression of major thoracic vessels, followed by resuscitation. The normally expressed MAP2b mRNA showed transient twofold elevations in all hippocampal neuron populations at 6-h recirculation, followed by a return to control levels by 24 h. MAP2b hybridization was progressively lost thereafter from the vulnerable CA1 and outer cortical layers, preceding both the fall in immunoreactive MAP2b and the eventual cell loss in these regions. The depletion of MAP2b mRNA coincided with an increase in the alternatively spliced MAP2c in vulnerable regions during 12-48 h of recirculation, precisely overlapping the late component of hsp72 expression that persisted in these cell populations. Previous studies have suggested that the initial induction of hsp72 provides an index of potential postischemic injury in neuron populations that may or may not be injured, while lasting hsp72 mRNA expression is associated with cell damage. In contrast, the present results demonstrate that MAP2c expression under these conditions occurs uniquely in neuron populations subject to injury. Available evidence suggests that MAP2c expression represents a plastic response in subpopulations of neurons that will survive in these regions, although it remains to be explicitly determined whether it may also be transiently expressed in dying cells. In any case, these observations demonstrate that reexpression of developmentally regulated MAP2c mRNA is a relatively late postischemic response in vulnerable cell populations, indicating that pathways regulating MAP2 splicing may be closely associated with mechanisms of neuron injury and/or recovery.
Collapse
Affiliation(s)
- N Saito
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
48
|
McRae A, Gilland E, Bona E, Hagberg H. Microglia activation after neonatal hypoxic-ischemia. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1995; 84:245-52. [PMID: 7743644 DOI: 10.1016/0165-3806(94)00177-2] [Citation(s) in RCA: 164] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The inflammatory response following hypoxic-ischemia (HI) in the neonate is largely unknown. Presently, the expression of microglial antigens and the beta-amyloid precursor protein (APP) were studied in relation to a dendrosomatic marker of neuronal injury (microtubule associated protein II; MAP II). HI was induced in 7-day-old rats by the combined unilateral carotid ligation and hypoxia. The pups (n = 23) were perfusion fixed 2-3 h, 24 h, 2-4 days and 14 days after HI and compared to sham-operated controls (n = 6). Antibodies were used for detection of the major histocompatibility complex II (OX-6), major histocompatibility complex I (OX-18) and complement receptor type 3 (OX-42), APP (APP 676-695) and MAP II (monoclonal MAP II) antigens. There was a transient APP expression 2-3 h after HI. A slight increase of microglial antigens (OX-18) was seen in the white matter 2 h after HI followed by a marked increase of OX-18, OX-6, OX-42 antigens 24 h-3-4 days in most injured regions with exception of the thalamus where a delayed (14 days) microglial response was seen. The latter event was parallelled by a delayed loss of MAP II. In conclusion, intense microglial expression occurs after neonatal HI either with an acute or delayed time-course depending on brain region.
Collapse
Affiliation(s)
- A McRae
- Department of Anatomy and Cell Biology, University of Göteborg, Sweden
| | | | | | | |
Collapse
|
49
|
Aoki M, Abe K, Yoshida T, Hattori A, Kogure K, Itoyama Y. Early immunohistochemical changes of microtubule based motor proteins in gerbil hippocampus after transient ischemia. Brain Res 1995; 669:189-96. [PMID: 7712174 DOI: 10.1016/0006-8993(94)01227-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Changes of immunoreactivities for microtubule based motor proteins, kinesin and cytoplasmic dynein, and non-motor protein, microtubule associated protein (MAP) 2 were investigated in gerbil hippocampus after transient ischemia. The immunoreactivities for kinesin showed a progressive decrease in hippocampal CA1 cells from 8 h after transient 5 or 15 min of ischemia that is lethal to the CA1 cells, while it showed no change after 2 min of ischemia that is non-lethal to the cells. The immunoreactivities for cytoplasmic dynein showed a decrease from 3 or 1 h of reperfusion in the CA1 cells after 5 or 15 min of ischemia, respectively. In contrast, the immunoreactivity for MAP2 remained normal until 2 days in the CA1 cells after 5 min of ischemia. These results showed an early changes of microtubule based motor proteins, such as kinesin and cytoplasmic dynein in vulnerable CA1 neurons. These changes may affect the mitochondrial shuttle system between neuronal cell body and the peripheries such as axon terminal and dendrites. This early disturbance may cause a failure to obtain newly synthesized nuclear encoded mitochondrial protein, and result in mitochondrial dysfunctions and the subsequent cell death.
Collapse
Affiliation(s)
- M Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Rosenstein JM. Diminished Expression of Microtubule-Associated Protein (MAP-2) and β-Tubulin as a Putative Marker for Ischemic Injury in Neocortical Transplants. Cell Transplant 1995; 4:83-91. [PMID: 7728337 DOI: 10.1177/096368979500400112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The present study examined the immunoexpression of the neuronal cytoskeletal proteins, MAP-2 and β-tubulin within a timed series of rat fetal neocortical transplants. β-tubulin is a major component of microtubules and MAP-2 regulates the assembly and stability of neuronal microtubules and is a major site for the phosphorylation cAMP dependent protein kinase in neurons. Both proteins are strongly expressed in the soma and dendrites of normal neurons. MAP-2 has been shown to be a sensitive marker for ischemia in neurons and is downregulated in this form of injury. Immunoexpression of both MAP-2 and β-tubulin in grafted cortical neurons was markedly reduced when compared to age-matched or even perinatal specimens at all postoperative times. Dendritic staining was confined to random, thin processes with no laminar patterns and staining within somata was very weak. In some specimens, somatic expression was increased and dendrites were more robustly stained when a portion of the graft was juxtaposed to a fiber tract even though in other regions of the same graft there was very weak immunostaining. The present results corroborate previous studies of cortical transplants indicating an immature structure and metabolism, and it is suggested here that the primary factor is a sublethal form of ischemic injury. Another possibility for the relative paucity of cytoskeletal protein expression could be that transplanted neurons undergo a new developmental scheme (neodevelopment) that is brought about by truncated migration patterns and abnormal synaptic connections.
Collapse
Affiliation(s)
- J M Rosenstein
- Department of Anatomy, George Washington University Medical Center, Washington, DC 20037, USA
| |
Collapse
|