1
|
Waldman AT, Sollee JR, Datta R, Lavery AM, Liu G, Aleman TS, Banwell BL, Gaetz WC. Structural correlates of atypical visual and motor cortical oscillations in pediatric-onset multiple sclerosis. Hum Brain Mapp 2020; 41:4299-4313. [PMID: 32648649 PMCID: PMC7502834 DOI: 10.1002/hbm.25126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/18/2020] [Accepted: 06/23/2020] [Indexed: 01/05/2023] Open
Abstract
We have previously demonstrated that pediatric‐onset multiple sclerosis (POMS) negatively impacts the visual pathway as well as motor processing speed. Relationships between MS‐related diffuse structural damage of gray and white matter (WM) tissue and cortical responses to visual and motor stimuli remain poorly understood. We used magnetoencephalography in 14 POMS patients and 15 age‐ and sex‐matched healthy controls to assess visual gamma (30–80 Hz), motor gamma (60–90 Hz), and motor beta (15–30 Hz) cortical oscillatory responses to a visual‐motor task. Then, 3T MRI was used to: (a) calculate fractional anisotropy (FA) of the posterior visual and corticospinal motor WM pathways and (b) quantify volume and thickness of the cuneus and primary motor cortex. Visual gamma band power was reduced in POMS and was associated with reduced FA of the optic radiations but not with loss of cuneus volume or thickness. Activity in the primary motor cortex, as measured by postmovement beta rebound amplitude associated with peak latency, was decreased in POMS, although this reduction was not predicted by structural metrics. Our findings implicate loss of WM integrity as a contributor to reduced electrical responses in the visual cortex in POMS. Future work in larger cohorts will inform on the cognitive implications of this finding in terms of visual processing function and will determine whether the progressive loss of brain volume known to occur in POMS ultimately contributes to both progressive dysfunction in such tasks as well as progressive reduction in cortical electrical responses in the visual cortex.
Collapse
Affiliation(s)
- Amy T Waldman
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology and Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John R Sollee
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Ritobrato Datta
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Amy M Lavery
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Geraldine Liu
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Tomas S Aleman
- Division of Ophthalmology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brenda L Banwell
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology and Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William C Gaetz
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Li J, Guo M, Liu Y, Wu G, Miao L, Zhang J, Zuo Z, Li Y. Both GSK-3β/CRMP2 and CDK5/CRMP2 pathways participate in the protection of dexmedetomidine against propofol-induced learning and memory impairment in neonatal rats. Toxicol Sci 2019; 171:193-210. [PMID: 31187143 DOI: 10.1093/toxsci/kfz135] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/17/2019] [Accepted: 05/29/2019] [Indexed: 11/13/2022] Open
Abstract
Dexmedetomidine has been reported to ameliorate propofol-induced neurotoxicity in neonatal animals. However, the underlying mechanism is still undetermined. Glycogen synthase kinase-3β (GSK-3β), cycline dependent kinase-5 (CDK5) and Rho-kinase (RhoA) pathways play critical roles in neuronal development. The present study is to investigate whether GSK-3β, CDK5 and RhoA pathways are involved in the neuroprotection of dexmedetomidine. Seven-day-old (P7) Sprague-Dawley rats were anesthetized with propofol for 6 h. Dexmedetomidine at various concentrations were administered before propofol exposure. Neuroapoptosis, the neuronal proliferation and the level of neurotransmitter in the hippocampus were evaluated. The effects of GSK-3β inhibitor SB415286, CDK5 inhibitor roscovitine or RhoA inhibitor Y276321 on propofol-induced neurotoxicity were assessed. Propofol induced apoptosis in the hippocampal neurons and astrocytes, inhibited neuronal proliferation in the DG region, down-regulated the level of γ-aminobutyric acid (GABA) and glutamate in the hippocampus, and impaired long-term cognitive function. These harmful effects were reduced by pretreatment with 50 μg·kg-1 dexmedetomidine. Moreover, propofol activated GSK-3β and CDK5 pathways, but not RhoA pathway, by reducing the phosphorylation of GSK-3β (ser 9), increasing the expression of CDK5 activator P25 and increasing the phosphorylation of their target sites on CRMP2 shortly after exposure. These effects were reversed by pretreatment with 50 μg·kg-1 dexmedetomidine. Furthermore, SB415286 and roscovitine, not Y276321, attenuated the propofol-induced neuroapoptosis, brain cell proliferation inhibition, GABA and glutamate downregulation, and learning and memory dysfunction. Our results indicate that dexmedetomidine reduces propofol-induced neurotoxicity and neurocognitive impairment via inhibiting activation of GSK-3β/CRMP2 and CDK5/CRMP2 pathways in the hippocampus of neonatal rats.
Collapse
Affiliation(s)
- Junhua Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minyan Guo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yafang Liu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guiyun Wu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liping Miao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia, 22908-0710, USA
| | - Yujuan Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Linck PA, Kuchcinski G, Munsch F, Griffier R, Lopes R, Okubo G, Sagnier S, Renou P, Asselineau J, Perez P, Dousset V, Sibon I, Tourdias T. Neurodegeneration of the Substantia Nigra after Ipsilateral Infarct: MRI R2* Mapping and Relationship to Clinical Outcome. Radiology 2019; 291:438-448. [PMID: 30860451 DOI: 10.1148/radiol.2019182126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background The substantia nigra (SN) is suspected to be affected after remote infarction, in view of its large array of connections with the supratentorial brain. Whether secondary involvement of SN worsens overall clinical outcome after a supratentorial stroke has not previously been studied. Purpose To assess longitudinal changes in SN R2* by using MRI in the setting of ipsilesional supratentorial infarct and the relationship of SN signal change to clinical outcome. Materials and Methods Participants prospectively included from 2012 to 2015 were evaluated at 24-72 hours (baseline visit) and at 1 year with MRI to quantify R2*. The SN was segmented bilaterally to calculate an R2* asymmetry index (SN-AI); greater SN-AI indicated greater relative R2* in the ipsilateral compared with contralateral SN. The 95th percentile of R2* (hereafter, SN-AI95) was compared according to infarct location with mixed linear regression models. We also conducted voxel-based comparisons of R2* and identified individual infarcted voxels associated with high SN-AI95 through voxel-based lesion-symptom mapping. Multivariable regression models tested the association between SN-AI95 and clinical scores. Results A total of 181 participants were evaluated (127 men, 54 women; mean age ± standard deviation, 64.2 years ± 13.1; 75 striatum infarcts, 106 other locations). Visual inspection, SN-AI95, and average maps consistently showed higher SN R2* at 1 year if ipsilateral striatum was infarcted than if it was not (SN-AI95, 4.25 vs -0.88; P < .001), but this was not observed at baseline. The striatal location of the infarct was associated with higher SN-AI95 at 1 year independently from infarct volume, SN-AI95 at baseline, microbleeds, age, and sex (β = 4.99; P < .001). Voxel-based lesion-symptom mapping confirmed that striatum but also insula, internal capsule, and external capsule were associated with higher SN-AI95 at 1 year. SN-AI95 was an independent contributor of poor motor outcome (Box and Block Test, β = -.62 points; P = .01). Conclusion In patients with stroke, greater substantia nigra R2*, likely reflective of greater iron content, can be observed at 1 year ipsilateral from remote infarcts of specific location, which is associated with worse motor function. © RSNA, 2019 Online supplemental material is available for this article. See also the editorial by Vernooij in this issue.
Collapse
Affiliation(s)
- Pierre Antoine Linck
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Gregory Kuchcinski
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Fanny Munsch
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Romain Griffier
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Renaud Lopes
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Gosuke Okubo
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Sharmila Sagnier
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Pauline Renou
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Julien Asselineau
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Paul Perez
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Vincent Dousset
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Igor Sibon
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| | - Thomas Tourdias
- From the Centre Hospitalier Universitaire (CHU) de Bordeaux, Department of Radiology and Diagnostic Neuroimaging, Bordeaux, France (P.A.L., F.M., V.D., T.T.); University of Bordeaux, Bordeaux, France (P.A.L., F.M., G.O., S.S., V.D., I.S., T.T.); CHU de Lille, Department of Neuroradiology, Lille, France (G.K., R.L.); University of Lille, Lille, France (G.K., R.L.); CHU de Bordeaux, Public Health Center, Methodological Support Unit for Clinical and Epidemiological Research, Bordeaux, France (R.G., J.A., P.P.); CHU de Bordeaux, Neurovascular Unit, Bordeaux, France (S.S., P.R., I.S.); and Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, Bordeaux, France (V.D., T.T.)
| |
Collapse
|
4
|
Ohtomo G, Mano T, Seto A, Tsuji S. Degeneration of the Substantia Nigra Following Ipsilateral Striatal Infarction. Intern Med 2018; 57:767-768. [PMID: 29151528 PMCID: PMC5874359 DOI: 10.2169/internalmedicine.9442-17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Gaku Ohtomo
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Tatsuo Mano
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Akiko Seto
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Shoji Tsuji
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Japan
| |
Collapse
|
5
|
Tsai RJ, Hsieh LC, Cheng SJ, Chen CY. Secondary pallidonigral degeneration mimicking recurrent acute stroke in clinical presentation and magnetic resonance imaging: a case report. BMC Neurol 2017; 17:215. [PMID: 29228907 PMCID: PMC5725823 DOI: 10.1186/s12883-017-1000-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/04/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Secondary pallidonigral transneuronal degeneration after a remote primary cerebral infarct can mimic recurrent stroke at clinical presentation. We describe a patient with secondary pallidonigral degeneration following a previous putaminal infarct, which was diagnosed through diffusion-weighted (DWI) and T2-weighted imaging (T2WI). CASE PRESENTATION A 64-year-old man complained of an acute relapse of right-lower-limb weakness following a cerebral infarction 2 months before presentation. Recurrent cerebral stroke was initially diagnosed in the emergency room. DWI of the brain revealed a subacute to chronic infarct in the left putamen and new acute cytotoxic edema in the left substantia nigra (SN) and globus pallidus while T2WI also showed hyperintensity in the same regions. The SN was outside the aforementioned middle cerebral arterial territory, which includes the putamen. These findings are compatible with the diagnosis of acute pallidonigral injury secondary to striatal infarction. The patient had fully recovered from his right-lower-limb weakness after 1 month. CONCLUSIONS Secondary pallidonigral degeneration may mimic recurrent stroke. DWI along with T2WI facilitates elucidation of this clinicopathological entity, and thus unnecessary treatment can be avoided.
Collapse
Affiliation(s)
- Ruei-Je Tsai
- Department of Medical Imaging, Taipei Medical University Hospital, 252 Wu Hsing Street, Taipei, 110, Taiwan
| | - Li-Chun Hsieh
- Department of Medical Imaging, Taipei Medical University Hospital, 252 Wu Hsing Street, Taipei, 110, Taiwan.,Translational Imaging Research Center (TIRC), College of Medicine, Taipei Medical University, 252 Wu Hsing Street, Taipei, 110, Taiwan
| | - Sho-Jen Cheng
- Department of Medical Imaging, Taipei Medical University Hospital, 252 Wu Hsing Street, Taipei, 110, Taiwan
| | - Cheng-Yu Chen
- Department of Medical Imaging, Taipei Medical University Hospital, 252 Wu Hsing Street, Taipei, 110, Taiwan. .,Translational Imaging Research Center (TIRC), College of Medicine, Taipei Medical University, 252 Wu Hsing Street, Taipei, 110, Taiwan. .,Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, 252 Wu Hsing Street, Taipei, 110, Taiwan.
| |
Collapse
|
6
|
Transneuronal Degeneration of Thalamic Nuclei following Middle Cerebral Artery Occlusion in Rats. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3819052. [PMID: 27597962 PMCID: PMC4997075 DOI: 10.1155/2016/3819052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 06/22/2016] [Accepted: 07/08/2016] [Indexed: 12/11/2022]
Abstract
Objective. Postinfarction transneuronal degeneration refers to secondary neuronal death that occurs within a few days to weeks following the disruption of input or output to synapsed neurons sustaining ischemic insults. The thalamus receives its blood supply from the posterior circulation; however, infarctions of the middle cerebral arterial may cause secondary transneuronal degeneration in the thalamus. In this study, we presented the areas of ischemia and associated transneuronal degeneration following MCAo in a rat model. Materials and Methods. Eighteen 12-week-old male Sprague-Dawley rats were randomly assigned to receive middle cerebral artery occlusion surgery for 1, 7, and 14 days. Cerebral atrophy was assessed by 2,3,5-triphenyltetrazolium hydrochloride staining. Postural reflex and open field tests were performed prior to animal sacrifice to assess the effects of occlusion on behavior. Results. Myelin loss was observed at the lesion site following ischemia. Gliosis was also observed in thalamic regions 14 days following occlusion. Differential degrees of increased vascular endothelial growth factor expression were observed at each stage of infarction. Increases in myelin basic protein levels were also observed in the 14-day group. Conclusion. The present rat model of ischemia provides evidence of transneuronal degeneration within the first 14 days of occlusion. The observed changes in protein expression may be associated with self-repair mechanisms in the damaged brain.
Collapse
|
7
|
Zhang R, Yang N, Ji C, Zheng J, Liang Z, Hou CY, Liu YY, Zuo PP. Neuroprotective effects of Aceglutamide on motor function in a rat model of cerebral ischemia and reperfusion. Restor Neurol Neurosci 2016; 33:741-59. [PMID: 26444640 DOI: 10.3233/rnn-150509] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE To investigate the effect and underlying mechanism of Aceglutamide on motor dysfunction in rats after cerebral ischemia-reperfusion. METHODS Adult male Sprague-Dawley rats were subjected to 2 h transient middle cerebral artery occlusion (MCAO). Aceglutamide or vehicle was intraperitoneally given to rats at 24 h after reperfusion and lasted for 14 days. Subsequently functional recovery was assessed and number of tyrosine hydroxylase (TH)-positive neurons in substantia nigra (SN) was analyzed. Tumor necrosis factor receptor-associated factor 1(TRAF1), P-Akt and Bcl-2/Bax were determined in mesencephalic tissue by Western blot method. PC12 cells and primary cultured mesencephalic neurons were employed to further investigate the mechanism of Aceglutamide. RESULTS Aceglutamide treatment improved behavioral functions, reduced the infarction volume, and elevated the number of TH-positive neurons in the SN. Moreover, Aceglutamide significantly attenuated neuronal apoptosis in the SN. Meanwhile Aceglutamide treatment significantly inhibited the expression of TRAF1 and up-regulated the expression of P-Akt and Bcl-2/Bax ratio both in vitro and in vivo. CONCLUSIONS Aceglutamide ameliorated motor dysfunction and delayed neuronal death in the SN after ischemia, which involved the inhibition of pro-apoptotic factor TRAF1 and activation of Akt/Bcl-2 signaling pathway. These data provided experimental information for applying Aceglutamide to ischemic stroke treatment.
Collapse
|
8
|
Abstract
Ischemic brain injury produced by stroke or cardiac arrest is a major cause of human neurological disability. Steady advances in the neurosciences have elucidated the pathophysiological mechanisms of brain ischemia and have suggested many therapeutic approaches to achieve neuroprotection of the acutely ischemic brain that are directed at specific injury mechanisms. In the second portion of this two-part review, the following potential therapeutic approaches to acute ischemic injury are considered: 1) modulation of nonglutamatergic neurotransmission, including monoaminergic systems (dopamine, norepinephrine, serotonin), γ-aminobutyric acid, and adenosine; 2) mild-to-moderate therapeutic hypothermia; 3) calcium channel antagonism; 4) an tagonism of oxygen free radicals; 5) modulation of the nitric oxide system; 6) antagonism of cytoskeletal proteolysis; 7) growth factor administration; 8) therapy directed at cellular mediators of injury; and 9) the rationale for combination pharmacotherapy. The Neuroscientist 1:164-175, 1995
Collapse
Affiliation(s)
- Myron D. Ginsberg
- Cerebral Vascular Disease Research Center Department
of Neurology University of Miami School of Medicine Miami, Florida
| |
Collapse
|
9
|
Cawley N, Solanky BS, Muhlert N, Tur C, Edden RAE, Wheeler-Kingshott CAM, Miller DH, Thompson AJ, Ciccarelli O. Reduced gamma-aminobutyric acid concentration is associated with physical disability in progressive multiple sclerosis. Brain 2015; 138:2584-95. [PMID: 26304151 DOI: 10.1093/brain/awv209] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neurodegeneration is thought to be the major cause of ongoing, irreversible disability in progressive stages of multiple sclerosis. Gamma-aminobutyric acid is the principle inhibitory neurotransmitter in the brain. The aims of this study were to investigate if gamma-aminobutyric acid levels (i) are abnormal in patients with secondary progressive multiple sclerosis compared with healthy controls; and (ii) correlate with physical and cognitive performance in this patient population. Thirty patients with secondary progressive multiple sclerosis and 17 healthy control subjects underwent single-voxel MEGA-PRESS (MEscher-GArwood Point RESolved Spectroscopy) magnetic resonance spectroscopy at 3 T, to quantify gamma-aminobutyric acid levels in the prefrontal cortex, right hippocampus and left sensorimotor cortex. All subjects were assessed clinically and underwent a cognitive assessment. Multiple linear regression models were used to compare differences in gamma-aminobutyric acid concentrations between patients and controls adjusting for age, gender and tissue fractions within each spectroscopic voxel. Regression was used to examine the relationships between the cognitive function and physical disability scores specific for these regions with gamma-aminobuytric acid levels, adjusting for age, gender, and total N-acetyl-aspartate and glutamine-glutamate complex levels. When compared with controls, patients performed significantly worse on all motor and sensory tests, and were cognitively impaired in processing speed and verbal memory. Patients had significantly lower gamma-aminobutyric acid levels in the hippocampus (adjusted difference = -0.403 mM, 95% confidence intervals -0.792, -0.014, P = 0.043) and sensorimotor cortex (adjusted difference = -0.385 mM, 95% confidence intervals -0.667, -0.104, P = 0.009) compared with controls. In patients, reduced motor function in the right upper and lower limb was associated with lower gamma-aminobutyric acid concentration in the sensorimotor cortex. Specifically for each unit decrease in gamma-aminobutyric acid levels (in mM), there was a predicted -10.86 (95% confidence intervals -16.786 to -4.482) decrease in grip strength (kg force) (P < 0.001) and -8.74 (95% confidence intervals -13.943 to -3.015) decrease in muscle strength (P < 0.006). This study suggests that reduced gamma-aminobutyric acid levels reflect pathological abnormalities that may play a role in determining physical disability. These abnormalities may include decreases in the pre- and postsynaptic components of gamma-aminobutyric acid neurotransmission and in the density of inhibitory neurons. Additionally, the reduced gamma-aminobutyric acid concentration may contribute to the neurodegenerative process, resulting in increased firing of axons, with consequent increased energy demands, which may lead to neuroaxonal degeneration and loss of the compensatory mechanisms that maintain motor function. This study supports the idea that modulation of gamma-aminobutyric acid neurotransmission may be an important target for neuroprotection in multiple sclerosis.See De Stefano and Giorgio (doi:10.1093/brain/awv213) for a scientific commentary on this article.
Collapse
Affiliation(s)
- Niamh Cawley
- 1 NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Bhavana S Solanky
- 1 NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Nils Muhlert
- 1 NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK 2 School of Psychology and Cardiff University Brain Research Imaging Centre, Cardiff University, Cardiff, UK 3 School of Psychological Sciences, University of Manchester, Manchester, UK
| | - Carmen Tur
- 1 NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK
| | - Richard A E Edden
- 4 Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA 5 FM Kirby Centre for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Claudia A M Wheeler-Kingshott
- 1 NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK 6 Brain Connectivity Centre, C. Mondino National Neurological Institute, Pavia, Italy
| | - David H Miller
- 1 NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK 7 National Institute of Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre (BRC), London, UK
| | - Alan J Thompson
- 1 NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK 7 National Institute of Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre (BRC), London, UK
| | - Olga Ciccarelli
- 1 NMR Research Unit, Queen Square Multiple Sclerosis Centre, UCL Institute of Neurology, London, UK 7 National Institute of Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre (BRC), London, UK
| |
Collapse
|
10
|
Winter B, Brunecker P, Fiebach JB, Jungehulsing GJ, Kronenberg G, Endres M. Striatal Infarction Elicits Secondary Extrafocal MRI Changes in Ipsilateral Substantia Nigra. PLoS One 2015; 10:e0136483. [PMID: 26325192 PMCID: PMC4556671 DOI: 10.1371/journal.pone.0136483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 12/24/2014] [Indexed: 02/05/2023] Open
Abstract
Focal ischemia may induce pathological alterations in brain areas distant from the primary lesion. In animal models, exofocal neuron death in the ipsilateral midbrain has been described after occlusion of the middle cerebral artery (MCA). Using sequential magnetic resonance imaging (T2- and diffusion-weighted) at 3 Tesla, we investigated acute ischemic stroke patients on days 1, 2, 6, 8, and 10 after stroke onset. Sixteen consecutive patients who had suffered a stroke involving the caudate nucleus and/or putamen of either hemisphere were recruited into the study. Four additional patients with strokes sparing the caudate nucleus and putamen but encompassing at least one-third of the MCA territory served as controls. Ischemic lesions involving striatal structures resulted in hyperintense lesions in ipsilateral midbrain that emerged between days 6 and 10 after stroke and were not present on the initial scans. In contrast, none of the control stroke patients developed secondary midbrain lesions. Hyperintense lesions in the pyramidal tract or the brain stem caused by degeneration of the corticospinal tract could be clearly distinguished from these secondary midbrain gray matter lesions and were detectable from day 2 after ischemia. Co-registration of high-resolution images with a digitized anatomic atlas revealed localization of secondary lesions primarily in the substantia nigra pars compacta. Apparent diffusion coefficient (ADC) values in the secondary lesions showed a delayed sharp decline through day 10. Normalization of ADC values was observed at late measurements. Taken together, our study demonstrates that striatal infarction elicits delayed degenerative changes in ipsilateral substantia nigra pars compacta.
Collapse
Affiliation(s)
- Benjamin Winter
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Peter Brunecker
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
| | - Jochen B. Fiebach
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
| | - Gerhard Jan Jungehulsing
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Jüdisches Krankenhaus Berlin, Heinz-Galinski-Strasse 1, 13347, Berlin, Germany
| | - Golo Kronenberg
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Max-Delbrück Center and Charité Medical Faculty, Experimental and Clinical Research Center, Lindenbergerweg 80, 13125, Berlin, Germany
| | - Matthias Endres
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Excellence Cluster Neurocure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee, 53175, Bonn, Germany
- German Centre for Cardiovascular Research (DZHK), Oudenarder Straße 16, 13347, Berlin, Germany
- * E-mail:
| |
Collapse
|
11
|
Thomsen MS, Andersen MV, Christoffersen PR, Jensen MD, Lichota J, Moos T. Neurodegeneration with inflammation is accompanied by accumulation of iron and ferritin in microglia and neurons. Neurobiol Dis 2015; 81:108-18. [DOI: 10.1016/j.nbd.2015.03.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 03/03/2015] [Accepted: 03/12/2015] [Indexed: 12/22/2022] Open
|
12
|
Characterization of Behaviour and Remote Degeneration Following Thalamic Stroke in the Rat. Int J Mol Sci 2015; 16:13921-36. [PMID: 26090717 PMCID: PMC4490531 DOI: 10.3390/ijms160613921] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/18/2015] [Accepted: 06/11/2015] [Indexed: 11/17/2022] Open
Abstract
Subcortical ischemic strokes are among the leading causes of cognitive impairment. Selective atrophy of remote brain regions connected to the infarct is thought to contribute to deterioration of cognitive functions. The mechanisms underlying this secondary degenerative process are incompletely understood, but are thought to include inflammation. We induce ischemia by unilateral injection of endothelin-I into the rat dorsomedial thalamic nucleus, which has defined reciprocal connections to the frontal cortex. We use a comprehensive test battery to probe for changes in behaviour, including executive functions. After a four-week recovery period, brain sections are stained with markers for degeneration, microglia, astrocytes and myelin. Degenerative processes are localized within the stroke core and along the full thalamocortical projection, which does not translate into measurable behavioural deficits. Significant microglia recruitment, astrogliosis or myelin loss along the axonal projection or within the frontal cortex cannot be detected. These findings indicate that critical effects of stroke-induced axonal degeneration may only be measurable beyond a threshold of stroke severity and/or follow a different time course. Further investigations are needed to clarify the impact of inflammation accompanying axonal degeneration on delayed remote atrophy after stroke.
Collapse
|
13
|
Kovacs SK, Leonessa F, Ling GSF. Blast TBI Models, Neuropathology, and Implications for Seizure Risk. Front Neurol 2014; 5:47. [PMID: 24782820 PMCID: PMC3988378 DOI: 10.3389/fneur.2014.00047] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/26/2014] [Indexed: 12/31/2022] Open
Abstract
Traumatic brain injury (TBI) due to explosive blast exposure is a leading combat casualty. It is also implicated as a key contributor to war related mental health diseases. A clinically important consequence of all types of TBI is a high risk for development of seizures and epilepsy. Seizures have been reported in patients who have suffered blast injuries in the Global War on Terror but the exact prevalence is unknown. The occurrence of seizures supports the contention that explosive blast leads to both cellular and structural brain pathology. Unfortunately, the exact mechanism by which explosions cause brain injury is unclear, which complicates development of meaningful therapies and mitigation strategies. To help improve understanding, detailed neuropathological analysis is needed. For this, histopathological techniques are extremely valuable and indispensable. In the following we will review the pathological results, including those from immunohistochemical and special staining approaches, from recent preclinical explosive blast studies.
Collapse
Affiliation(s)
- S Krisztian Kovacs
- Laboratory of Neurotrauma, Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Fabio Leonessa
- Laboratory of Neurotrauma, Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Geoffrey S F Ling
- Laboratory of Neurotrauma, Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| |
Collapse
|
14
|
Dirnagl U, Endres M. Found in translation: preclinical stroke research predicts human pathophysiology, clinical phenotypes, and therapeutic outcomes. Stroke 2014; 45:1510-8. [PMID: 24652307 DOI: 10.1161/strokeaha.113.004075] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Ulrich Dirnagl
- From the Departments of Neurology and Experimental Neurology, Center for Stroke Research Berlin, and Excellence Cluster NeuroCure, Charité-Universitätsmedizin Berlin, Berlin, Germany (U.D., M.E.); German Center for Neurodegeneration Research (DZNE), Partner Site, Berlin, Germany (U.D.); and German Center for Cardiovascular Diseases (DZHK), Partner Site, Berlin, Germany (U.D., M.E.)
| | | |
Collapse
|
15
|
Selective neuronal loss in ischemic stroke and cerebrovascular disease. J Cereb Blood Flow Metab 2014; 34:2-18. [PMID: 24192635 PMCID: PMC3887360 DOI: 10.1038/jcbfm.2013.188] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 01/23/2023]
Abstract
As a sequel of brain ischemia, selective neuronal loss (SNL)-as opposed to pannecrosis (i.e. infarction)-is attracting growing interest, particularly because it is now detectable in vivo. In acute stroke, SNL may affect the salvaged penumbra and hamper functional recovery following reperfusion. Rodent occlusion models can generate SNL predominantly in the striatum or cortex, showing that it can affect behavior for weeks despite normal magnetic resonance imaging. In humans, SNL in the salvaged penumbra has been documented in vivo mainly using positron emission tomography and (11)C-flumazenil, a neuronal tracer validated against immunohistochemistry in rodent stroke models. Cortical SNL has also been documented using this approach in chronic carotid disease in association with misery perfusion and behavioral deficits, suggesting that it can result from chronic or unstable hemodynamic compromise. Given these consequences, SNL may constitute a novel therapeutic target. Selective neuronal loss may also develop at sites remote from infarcts, representing secondary 'exofocal' phenomena akin to degeneration, potentially related to poststroke behavioral or mood impairments again amenable to therapy. Further work should aim to better characterize the time course, behavioral consequences-including the impact on neurological recovery and contribution to vascular cognitive impairment-association with possible causal processes such as microglial activation, and preventability of SNL.
Collapse
|
16
|
Da Silva AM, Willmore LJ. Posttraumatic epilepsy. HANDBOOK OF CLINICAL NEUROLOGY 2012; 108:585-99. [PMID: 22939055 DOI: 10.1016/b978-0-444-52899-5.00017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Affiliation(s)
- A Martins Da Silva
- Hospital Geral de Santo António and Biomedical Science Institute, University of Porto, Porto, Portugal
| | | |
Collapse
|
17
|
Nogo-A is associated with secondary degeneration of substantia nigra in hypertensive rats with focal cortical infarction. Brain Res 2012; 1469:153-63. [DOI: 10.1016/j.brainres.2012.06.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 06/23/2012] [Accepted: 06/26/2012] [Indexed: 10/28/2022]
|
18
|
Zuhayra M, Zhao Y, von Forstner C, Henze E, Gohlke P, Culman J, Lützen U. Activation of cerebral peroxisome proliferator-activated receptors γ (PPARγ) reduces neuronal damage in the substantia nigra after transient focal cerebral ischaemia in the rat. Neuropathol Appl Neurobiol 2012; 37:738-52. [PMID: 21366664 DOI: 10.1111/j.1365-2990.2011.01169.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
AIM The function of brain (neuronal) peroxisome proliferator-activated receptor(s) γ (PPARγ) in the delayed degeneration and loss of neurones in the substantia nigra (SN) was studied in rats after transient occlusion of the middle cerebral artery (MCAO). METHODS The PPARγ agonist, pioglitazone, or vehicle was infused intracerebroventricularly over a 5-day period before, during and 5 days after MCAO (90 min). The neuronal degeneration in the SN pars reticularis (SNr) and pars compacta (SNc), the analysis of the number of tyrosine hydroxylase-immunoreactive (TH-IR) neurones and the expression of the PPARγ in these neurones were studied by immunohistochemistry and immunofluorescence staining. The effects of PPARγ activation on excitotoxic and oxidative neuronal damage induced by glutamate and 6-hydroxydopamine were investigated in primary cortical neurones expressing PPARγ. RESULTS Pioglitazone reduced the total and striatal infarct size, neuronal degeneration in both parts of the ipsilateral SN, the loss of TH-IR neurones in the SNc and increased the number of PPARγ-positive TH-IR neurones. Pioglitazone protected primary cortical neurones against oxidative and excitotoxic damage, prevented the loss of neurites and supported the formation of synaptic networks in neurones exposed to glutamate or 6-hydroxydopamine by a PPARγ-dependent mechanism. CONCLUSIONS Activation of cerebral PPARγ confers neuroprotection after ischaemic stroke by preventing both, neuronal damage within the peri-infarct zone and delayed degeneration of neurones and neuronal death in areas remote from the site of ischaemic injury. Pioglitazone and other PPARγ agonists may be useful therapeutic agents to prevent progression of brain damage after cerebral ischaemia.
Collapse
Affiliation(s)
- M Zuhayra
- Department of Nuclear Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | | | | | | | | | | |
Collapse
|
19
|
Ohe Y, Uchino A, Horiuchi Y, Maruyama H, Deguchi I, Fukuoka T, Kato Y, Nagoya H, Dembo T, Tanahashi N. Magnetic resonance imaging investigation of secondary degeneration of the mesencephalic substantia nigra after cerebral infarction. J Stroke Cerebrovasc Dis 2011; 22:58-65. [PMID: 21784662 DOI: 10.1016/j.jstrokecerebrovasdis.2011.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 06/07/2011] [Accepted: 06/11/2011] [Indexed: 01/25/2023] Open
Abstract
Secondary degeneration of the mesencephalic substantia nigra after cerebral infarction is widely known to occur in animal experiments, but has yet to be sufficiently investigated in human cerebral infarction. This study investigated the background and features of patients exhibiting secondary degeneration of the mesencephalic substantia nigra. The subjects comprised 43 patients admitted to our hospital for cerebral infarction between April 2007 and October 2010 showing secondary degeneration of the mesencephalic substantia nigra on cranial magnetic resonance imaging (MRI). We investigated clinical disease type, location of vascular occlusion, lesion site, and time from onset of symptoms to lesion identification by MRI. The clinical disease type was cardiogenic embolism in 29 patients (67%), atheromatous embolism (artery to artery) in 8 patients (19%), embolism (origin unknown) in 2 patients (5%), infarction after coil embolization for internal carotid aneurysm in 1 patient (2%), arterial dissection in 2 patients (5%), and vasculitis due to Takayasu disease in 1 patient (2%). Magnetic resonance angiography (MRA) identified the occluded vessel as the internal carotid artery in 19 patients (44%), the middle cerebral artery (M1) in 20 patients (47%), and the middle cerebral artery (M2) in 3 patients (7%); MRA was not performed in 1 patient (2%). The cerebral infarctions were striatal in 7 patients (16%) and striatal and cortical in 36 patients (84%). Hyperintense regions in the mesencephalic substantia nigra were observed in all patients after 7-28 days (mean, 13.3 days) on diffusion-weighted imaging or fluid-attenuated inversion recovery and T2-weighted MRI. Most patients with secondary degeneration of the substantia nigra demonstrated clinical disease comprising vascular occlusion of the internal carotid artery or the neighborhood of the middle cerebral artery, which was envisaged to cause a sudden drop in brain circulation across a wide area. Striatal infarctions were observed in all patients. Secondary degeneration of the substantia nigra appeared at 1-4 weeks after onset and disappeared after several months.
Collapse
Affiliation(s)
- Yasuko Ohe
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Saitama, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Rekik L, Daguin-Nerrière V, Petit JY, Brachet P. γ-Aminobutyric acid type B receptor changes in the rat striatum and substantia nigra following intrastriatal quinolinic acid lesions. J Neurosci Res 2011; 89:524-35. [PMID: 21290407 DOI: 10.1002/jnr.22574] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Revised: 10/25/2010] [Accepted: 11/05/2010] [Indexed: 12/16/2023]
Abstract
Changes in the regional distribution of the metabotropic GABA type B receptors (GABA(B)) were investigated in a rat model of Huntington's disease. Animals received a unilateral intrastriatal injection of quinolinic acid (QA), and GABA(B) immunoreactivity was monitored 3, 11, and 21 days postinjection in the striatum and substantia nigra (SN). Two antibodies, recognizing either the GABA(B1) or the GABA(B2) receptor subtypes, were used. QA injection rapidly induced a protracted increase in GABA(B1) or GABA(B2) immunoreactivity in the lesioned striatum, despite the neuronal loss. In the SN, a continuous increase in GABA(B1) and GABA(B2) immunoreactivity was observed at all time points in the ipsilateral pars reticulata (SNr), whereas the pars compacta (SNc) was unaffected by this phenomenon. This increase was supported by a densitometric analysis. At day 21 postlesion induction, intensely labeled stellate cells and processes were found in the ipsilateral SNr, in addition to immunoreactive neurons. Double labeling of GABA(B1) and glial fibrillary acidic protein (GFAP) showed that the stellate cells were reactive astrocytes. Hence, part of the sustained increase in GABA(B) immunoreactivity that takes place in the SNr and possibly the striatum may be ascribed to reactive astrocytes. It is suggested that GABA(B) receptors are up-regulated in these reactive astrocytes and that agonists might influence the extent of this astroglial reaction.
Collapse
Affiliation(s)
- Letaïef Rekik
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
| | | | | | | |
Collapse
|
21
|
Uchida H, Yokoyama H, Kimoto H, Kato H, Araki T. Long-term changes in the ipsilateral substantia nigra after transient focal cerebral ischaemia in rats. Int J Exp Pathol 2010; 91:256-66. [PMID: 20353427 DOI: 10.1111/j.1365-2613.2010.00712.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Transient focal cerebral ischaemia can cause neuronal damage in remote areas, including the ipsilateral thalamus and subsutantia nigra, as well as in the ischaemic core. In the present study, we investigated long-term changes in the ipsilateral substantia nigra from 1 up to 20 weeks after 90 min of transient focal cerebral ischaemia in rats, using tyrosine hydroxylase (TH), neuronal nuclei (NeuN), Iba-1, glial fibrillary acidic protein (GFAP) and brain-derived neurotrophic factor (BDNF) immunostaining. These results show that transient focal cerebral ischaemia in rats can cause a severe and prolonged neuronal damage in the ipsilateral striatum. Our results with TH and NeuN immunostaining also demonstrate that the atrophy of the ipsilateral substantia nigra after transient focal cerebral ischaemia was not static but progressive. Furthermore, our double-labelled immunohistochemical study suggests that BDNF released by GFAP-positive astrocytes may play a key role in the survival of dopaminergic neurones in the ipsilateral substantia nigra at the chronic stage after transient focal cerebral ischaemia, although the areas of the ipsilateral substantia nigra are decreased progressively after ischaemia. Thus our study provides further valuable information for the pathogenesis of neuronal damage after transient focal cerebral ischaemia.
Collapse
Affiliation(s)
- Hiroto Uchida
- Department of Neurobiology and Therapeutics, Graduate School and Faculty of Pharmaceutical Sciences, The University of Tokushima, Tokushima, Japan
| | | | | | | | | |
Collapse
|
22
|
Abstract
AbstractThe effects of lesioning the ventral tegmental area (VTA) or substantia nigra (SN) neurons by means of bilateral stereotaxic microinjections of kainic acid (KA) (0.4 mM) were investigated to clarify the role of the VTA and the SN neurons in learning and memory processes. The present study demonstrates that KA in the SN and the VTA lesioned rats significantly decreased spontaneous alternation in Y-maze task, working memory and reference memory in radial 8 arm-maze task, suggesting effects on spatial memory performance. Our findings provide further support for the role of the VTA and the SN neurons in processing and storage of information.
Collapse
|
23
|
Willmore LJ, Ueda Y. Posttraumatic epilepsy: hemorrhage, free radicals and the molecular regulation of glutamate. Neurochem Res 2008; 34:688-97. [PMID: 18785008 DOI: 10.1007/s11064-008-9841-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 08/22/2008] [Indexed: 11/29/2022]
Abstract
Traumatic brain injury causes development of posttraumatic epilepsy. Bleeding within neuropil is followed by hemolysis and deposition of hemoglobin in neocortex. Iron from hemoglobin and transferring is deposited in brains of patients with posttraumatic epilepsy. Iron compounds form reactive free radical oxidants. Microinjection of ferric ions into rodent brain results in chronic recurrent seizures and liberation of glutamate into the neuropil, as is observed in humans with epilepsy. Termination of synaptic effects of glutamate is by removal via transporter proteins. EAAC-1 is within neurons while GLT-1 and GLAST are confined to glia. Persistent down regulation of GLAST production is present in hippocampal regions in chronic seizure models. Down regulation of GLAST may be fundamental to a sequence of free radical reactions initiated by brain injury with hemorrhage. Administration of antioxidants to animals causes interruption of the sequence of brain injury responses induced by hemorrhage, suggesting that such a strategy needs to be evaluated in patients with traumatic brain injury.
Collapse
Affiliation(s)
- L J Willmore
- Department of Neurology & Psychiatry, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MI 63104, USA.
| | | |
Collapse
|
24
|
Hirouchi Y, Suzuki E, Mitsuoka C, Jin H, Kitajima S, Kohjimoto Y, Enomoto M, Kugino K. Neuroimaging and histopathological evaluation of delayed neurological damage produced by artificial occlusion of the middle cerebral artery in Cynomolgus monkeys. ACTA ACUST UNITED AC 2007; 59:9-16. [PMID: 17596924 DOI: 10.1016/j.etp.2007.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Accepted: 02/09/2007] [Indexed: 10/23/2022]
Abstract
A monkey model (Cynomolgus) was established to evaluate the delayed neurological damage evident at areas distant from ischemic cerebral foci. In addition to proton magnetic resonance spectroscopy (MRS) monitoring in life, histological examinations of specimens of the brain was conducted on lesions produced 6h and 1, 2, 4 and 8 weeks after unilateral (left) permanent middle cerebral artery occlusion (pMCO) on five monkeys. In addition to the typical images evident at primary ischemic foci around the middle cerebral artery, MRS revealed and enhanced, clearer region, due to edema extending into the reticular and compact area of the left substantia nigra one week after pMCO, inducing right hemiparesis caused by focal cerebral ischemia. Similar histological lesions were also induced in the left thalamus 4 weeks after pMCO. Thereafter, a variety of histological findings including astrocytic activation, reduced number of nerve cells and gliosis were found in the above described areas far apart from the original ischemic cerebral foci. Our monkey model should be suitable for studies elucidating the pathological process in cerebral ischemia as well as for investigating therapeutic strategies involving ischemic stroke in humans.
Collapse
|
25
|
Wright AK, Arbuthnott GW. The influence of the subthalamic nucleus upon the damage to the dopamine system following lesions of globus pallidus in rats. Eur J Neurosci 2007; 26:642-8. [PMID: 17634067 DOI: 10.1111/j.1460-9568.2007.05706.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Lesioning or stimulating the subthalamic nucleus (STN) in patients with Parkinson's disease, or in animal models of parkinsonism, alleviates many of the symptoms and so it is tempting to think of the STN as a part of the cause of Parkinson's disease. The globus pallidus (GP) is thought to have a tonic inhibitory action on the STN. An ibotenic acid injection into the GP in rats removes the cells of the GP and, over the following 6 weeks, a progressive loss of dopamine cells (counted stereologically in sections stained for tyrosine hydroxylase) develops in substantia nigra (SN). In this investigation we show that, when animals have the STN cells destroyed by very small ibotenic acid injections, their dopamine neurons are not damaged. Furthermore, if a lesion to the GP follows a lesion of STN then the dopamine cells also survive this double insult, at least for the first 3 weeks following the lesion. The experiments provide good reason to suspect that, at least in the short term, increased activity in the STN is a contributory cause of the loss of dopamine cells which follows the lesion of the GP in rats. Whether or not this is part of the mechanism of cell loss in Parkinson's disease, the rats with GP lesions at least provide an opportunity to test strategies that might protect dopamine cells from slowly developing damage. Removing the STN seems to be neuroprotective in this new model of dopamine degeneration.
Collapse
Affiliation(s)
- A K Wright
- Division of Neuroscience, University of Edinburgh, EH9 1QH, Scotland, UK
| | | |
Collapse
|
26
|
Yang SH, Shetty RA, Liu R, Sumien N, Heinrich KR, Rutledge M, Thangthaeng N, Brun-Zinkernagel AM, Forster MJ. Endovascular middle cerebral artery occlusion in rats as a model for studying vascular dementia. AGE (DORDRECHT, NETHERLANDS) 2006; 28:297-307. [PMID: 22253496 PMCID: PMC3259150 DOI: 10.1007/s11357-006-9026-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2006] [Revised: 10/20/2006] [Accepted: 10/23/2006] [Indexed: 05/31/2023]
Abstract
Vascular dementia (VaD), incorporating cognitive dysfunction with vascular disease, ranks as the second leading cause of dementia in the United States, yet no effective treatment is currently available. The challenge of defining the pathological substrates of VaD is complicated by the heterogeneous nature of cerebrovascular disease and coexistence of other pathologies, including Alzheimer's disease (AD) types of lesion. The use of rodent models of ischemic stroke may help to elucidate the type of lesions that are responsible for cognitive impairment in humans. Endovascular middle cerebral artery (MCA) occlusion in rats is considered to be a convenient and reliable model of human cerebral ischemia. Both sensorimotor and cognitive dysfunction can be induced in the rat endovascular MCA occlusion model, yet sensorimotor deficits induced by endovascular MCA occlusion may improve with time, whereas data presented in this review suggest that in rats this model can result in a progressive course of cognitive impairment that is consistent with the clinical progression of VaD. Thus far, experimental studies using this model have demonstrated a direct interaction of cerebral ischemic damage and AD-type neuropathologies in the primary ischemic area. Further, coincident to the progressive decline of cognitive function, a delayed neurodegeneration in a remote area, distal to the primary ischemic area, the hippocampus, has been demonstrated in a rat endovascular MCA occlusion model. We argue that this model could be employed to study VaD and provide insight into some of the pathophysiological mechanisms of VaD.
Collapse
Affiliation(s)
- Shao-Hua Yang
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| | - Ritu A. Shetty
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| | - Ran Liu
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| | - Nathalie Sumien
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| | - Kevin R. Heinrich
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| | - Margaret Rutledge
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| | - Nopporn Thangthaeng
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| | - Anne-Marie Brun-Zinkernagel
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| | - Michael J. Forster
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107 USA
| |
Collapse
|
27
|
Dorandeu A, Wingertsmann L, Chrétien F, Delisle M, Vital C, Parchi P, Montagna P, Lugaresi E, Ironside JW, Budka H, Gambetti P, Gray F. Neuronal apoptosis in fatal familial insomnia. Brain Pathol 2006; 8:531-7. [PMID: 9669704 PMCID: PMC8098240 DOI: 10.1111/j.1750-3639.1998.tb00175.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The possibility that neuronal loss in prion diseases occurs through an apoptotic process has been postulated and is consistent with the lack of inflammation in these disorders. In order to test this hypothesis in FFI, in which neuronal loss is the predominant neuropathological feature, we examined samples of thalamus, basal ganglia, cerebral cortex, cerebellum and medulla from 10 subjects with FFI. All the patients had the characteristic 178 N mutation of the PrP gene. Eight subjects were homozygous methionine/methionine at codon 129 and 2 were heterozygous methionine/valine. Apoptotic neurons were identified by in situ end labelling in all the FFI cases and in none of the controls. They were mostly found in damaged regions and their presence and abundance seemed to correlate closely with the neuronal loss. They were particularly abundant in the thalamus and medullary olives. In heterozygous cases who had a longer disease duration and more widespread cerebral changes, apoptotic neurons were also found in the neocortex and striatum. The abundance of apoptotic neurons also correlated well with microglial activation as demonstrated by the expression of major histocompatibility complex class II antigens. PrPres immunostaining was almost invariably negative, consistent with previous data showing the lack of obvious correlation between neuronal loss and PrPres deposits in prion diseases.
Collapse
Affiliation(s)
- Anne Dorandeu
- Laboratoire de Neuropathologie, Faculté de Médecine Paris‐Ouest, Garches, France
| | - Laure Wingertsmann
- Laboratoire d'Histologie, Embryologie et Cytogénétique, Faculté de Médecine Pitié‐Salpêtrière, Paris, France
| | - Fabrice Chrétien
- Laboratoire d'Histologie, Embryologie et Cytogénétique, Faculté de Médecine Pitié‐Salpêtrière, Paris, France
| | | | - Claude Vital
- Laboratoire de Neuropathologie, Université Victor Ségalen, Bordeaux, France
| | - Piero Parchi
- Division of Neuropathology, Institute of Pathology, Case Western University, Cleveland, Ohio, USA
| | - Pasquale Montagna
- Institute of Clinical Neurology, University of Bologna, Bologna, Italy
| | - Elio Lugaresi
- Institute of Clinical Neurology, University of Bologna, Bologna, Italy
| | - James W. Ironside
- Neuropathology Laboratory, Department of Pathology, the University of Edinburgh, Edinburgh, UK
| | - Herbert Budka
- Institute of Neurology, University of Vienna, Vienna, Austria
| | - Pierluigi Gambetti
- Division of Neuropathology, Institute of Pathology, Case Western University, Cleveland, Ohio, USA
| | - Françoise Gray
- Laboratoire de Neuropathologie, Faculté de Médecine Paris‐Ouest, Garches, France
- Laboratoire d'Histologie, Embryologie et Cytogénétique, Faculté de Médecine Pitié‐Salpêtrière, Paris, France
| |
Collapse
|
28
|
Bazille C, Megarbane B, Bensimhon D, Lavergne-Slove A, Baglin AC, Loirat P, Woimant F, Mikol J, Gray F. Brain damage after heat stroke. J Neuropathol Exp Neurol 2005; 64:970-5. [PMID: 16254491 DOI: 10.1097/01.jnen.0000186924.88333.0d] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cerebellar syndromes and radiologic cerebellar atrophy after hyperpyrexia have occasionally been reported, mostly in neuroleptic malignant syndromes, but neuropathologic studies are extremely rare. We studied 3 patients (a 74-year-old woman, a 63-year-old man, and an 80-year-old man) who had heat stroke during heat waves in France. One patient had generalized seizures and died 28 hours after admission. The other patients survived one month and 2 months after admission; both had palatal myoclonus, and in one case, magnetic resonance imaging showed high signal intensity in the cerebral peduncles. The main neuropathology in the 3 cases was severe diffuse loss of Purkinje cells associated with heat shock protein 70 expression by Bergmann glia. In situ end labeling was negative in surviving Purkinje cells, suggesting that the mechanism of neuronal death was not apoptosis. Degeneration of Purkinje cells axons resulted in myelin pallor of the white matter of the folia and of the hilum of the dentate nuclei. DNA internucleosomal breakages were identified by in situ end labeling in the dentate nuclei and centromedian nuclei of the thalamus and were associated with degeneration of the cerebellar efferent pathways: superior cerebellar peduncles, decussation of the superior cerebellar peduncles (Wernekinck commissure), and dentatothalamic tract. These findings suggest that the mechanisms of neuronal death in the dentate nuclei and centromedian nuclei of the thalamus was different from that in Purkinje cells and more likely resulted from deafferentation. Ammon's horn and other areas susceptible to hypoxia were spared. These observations confirm the selective vulnerability of Purkinje cells to heat-induced injury and involvement of the cerebellar efferent pathways in palatal myoclonus.
Collapse
Affiliation(s)
- Céline Bazille
- Service Central d'Anatomie et de Cytologie Pathologiques, Hôpital Lariboisière, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Mátyás F, Yanovsky Y, Mackie K, Kelsch W, Misgeld U, Freund TF. Subcellular localization of type 1 cannabinoid receptors in the rat basal ganglia. Neuroscience 2005; 137:337-61. [PMID: 16289348 DOI: 10.1016/j.neuroscience.2005.09.005] [Citation(s) in RCA: 147] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2005] [Revised: 09/08/2005] [Accepted: 09/09/2005] [Indexed: 11/29/2022]
Abstract
Endocannabinoids, acting via type 1 cannabinoid receptors (CB1), are known to be involved in short-term synaptic plasticity via retrograde signaling. Strong depolarization of the postsynaptic neurons is followed by the endocannabinoid-mediated activation of presynaptic CB1 receptors, which suppresses GABA and/or glutamate release. This phenomenon is termed depolarization-induced suppression of inhibition (DSI) or excitation (DSE), respectively. Although both phenomena have been reported to be present in the basal ganglia, the anatomical substrate for these actions has not been clearly identified. Here we investigate the high-resolution subcellular localization of CB1 receptors in the nucleus accumbens, striatum, globus pallidus and substantia nigra, as well as in the internal capsule, where the striato-nigral and pallido-nigral pathways are located. In all examined nuclei of the basal ganglia, we found that CB1 receptors were located on the membrane of axon terminals and preterminal axons. Electron microscopic examination revealed that the majority of these axon terminals were GABAergic, giving rise to mostly symmetrical synapses. Interestingly, preterminal axons showed far more intense staining for CB1, especially in the globus pallidus and substantia nigra, whereas their terminals were only faintly stained. Non-varicose, thin unmyelinated fibers in the internal capsule also showed strong CB1-labeling, and were embedded in bundles of myelinated CB1-negative axons. The majority of CB1 receptors labeled by immunogold particles were located in the axonal plasma membrane (92.3%), apparently capable of signaling cannabinoid actions. CB1 receptors in this location cannot directly modulate transmitter release, because the release sites are several hundred micrometers away. Interestingly, both the CB1 agonist, WIN55,212-2, as well as its antagonist, AM251, were able to block action potential generation, but via a CB1 independent mechanism, since the effects remained intact in CB1 knockout animals. Thus, our electrophysiological data suggest that these receptors are unable to influence action potential propagation, thus they may not be functional at these sites, but are likely being transported to the terminal fields. The present data are consistent with a role of endocannabinoids in the control of GABA, but not glutamate, release in the basal ganglia via presynaptic CB1 receptors, but also call the attention to possible non-CB1-mediated effects of widely used cannabinoid ligands on action potential generation.
Collapse
Affiliation(s)
- F Mátyás
- Institute of Experimental Medicine, Hungarian Academy of Sciences, PO Box 67, Budapest H-1450, Hungary
| | | | | | | | | | | |
Collapse
|
30
|
Kinoshita T, Moritani T, Shrier DA, Wang HZ, Hiwatashi A, Numaguchi Y, Westesson PLA. Secondary degeneration of the substantia nigra and corticospinal tract after hemorrhagic middle cerebral artery infarction: diffusion-weighted MR findings. Magn Reson Med Sci 2005; 1:175-8. [PMID: 16082141 DOI: 10.2463/mrms.1.175] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Middle cerebral artery (MCA) infarction involving the striatum can cause secondary degeneration of the substantia nigra and corticospinal tract. We present a patient with subacute hemorrhagic MCA infarction in whom diffusion-weighted MR images showed high signal intensity in the ipsilateral substantia nigra and corticospinal tract. A corresponding apparent diffusion coefficient map revealed a uniformly decreased signal in the same area. This represents secondary degeneration and should not be mistaken for other pathological conditions, such as a new infarction.
Collapse
Affiliation(s)
- Toshibumi Kinoshita
- Department of Radiology, Division of Neuroradiology, University of Rochester Medical Center, 601 Elmwood Avenue, Box 648, Rochester, NY 14642, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Block F, Dihné M, Loos M. Inflammation in areas of remote changes following focal brain lesion. Prog Neurobiol 2005; 75:342-65. [PMID: 15925027 DOI: 10.1016/j.pneurobio.2005.03.004] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2004] [Revised: 03/15/2005] [Accepted: 03/31/2005] [Indexed: 11/22/2022]
Abstract
Focal brain lesions can lead to metabolic and structural changes in areas distant from but connected to the lesion site. After focal ischemic or excitotoxic lesions of the cortex and/or striatum, secondary changes have been observed in the thalamus, substantia nigra pars reticulata, hippocampus and spinal cord. In all these regions, inflammatory changes characterized by activation of microglia and astrocytes appear. In the thalamus, substantia nigra pars reticulata and hippocampus, an expression of proinflammatory cytokine like tumor necrosis factor-alpha and interleukin-1beta is induced. However, time course of expression and cellular localisation differ between these regions. Neuronal damage has consistently been observed in the thalamus, substantia nigra and spinal cord. It can be present in the hippocampus depending on the procedure of induction of focal cerebral ischemia. This secondary neuronal damage has been linked to antero- and retrograde degeneration. Anterograde degeneration is associated with somewhat later expression of cytokines, which is localised in neurons. In case of retrograde degeneration, the expression of cytokines is earlier and is localised in astrocytes. Pharmacological intervention aiming at reducing expression of tumor necrosis factor-alpha leads to reduction of secondary neuronal damage. These first results suggest that the inflammatory changes in remote areas might be involved in the pathogenesis of secondary neuronal damage.
Collapse
Affiliation(s)
- F Block
- Department of Neurology UK Aachen, Pauwelsstr. 30, D-52057 Aachen, Germany.
| | | | | |
Collapse
|
32
|
Zhao CS, Puurunen K, Schallert T, Sivenius J, Jolkkonen J. Behavioral effects of photothrombotic ischemic cortical injury in aged rats treated with the sedative-hypnotic GABAergic drug zopiclone. Behav Brain Res 2005; 160:260-6. [PMID: 15863222 DOI: 10.1016/j.bbr.2004.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Revised: 12/07/2004] [Accepted: 12/09/2004] [Indexed: 11/27/2022]
Abstract
Sedative-hypnotic drugs commonly used in the elderly may affect functional recovery following cerebrovascular events. Previous research has shown that prolonged exposure to diazepam can interfere with recovery of function and exaggerate tissue loss after brain injury. The present study evaluated the effect of zopiclone, a widely used hypnotic drug, on functional and histological outcome after cortical photothrombosis in aged rats, which might be particularly vulnerable to brain insults and inhibitory sedative-hypnotic drugs. Aged Wistar rats were treated with zopiclone at a dose of 3 mg/kg (i.p., once a day) beginning 4 days before ischemia induction and continuing for 23 days. Sensorimotor recovery was assessed by a new ledged beam-walking test and spatial learning by the Morris water-maze. After a 7-day washout period all rats were administered a single dose of zopiclone (3 mg/kg, i.p.) and retested. Infarct volumes were measured from nitroblue tetrazolium-stained sections at the end of the experiment. Beam-walking data showed that ischemic rats treated with zopiclone were not more impaired than untreated rats. Indeed, they showed fewer faults with the impaired hindlimb than ischemic controls on post-operative day 16. Water-maze performance was not affected by zopiclone. After the washout period a single dose of zopiclone did not worsen forelimb or hindlimb function, but seemed to improve performance in the water-maze test. Cortical infarct volumes were similar in ischemic controls and ischemic rats treated with zopiclone. In conclusion, zopiclone was not detrimental and even seemed to improve behavioral outcome without affecting ischemic damage in aged rats subjected to cortical photothrombosis.
Collapse
Affiliation(s)
- Chuan-sheng Zhao
- Department of Neuroscience and Neurology, University of Kuopio, Finland
| | | | | | | | | |
Collapse
|
33
|
Burke RE. Ontogenic cell death in the nigrostriatal system. Cell Tissue Res 2004; 318:63-72. [PMID: 15349767 DOI: 10.1007/s00441-004-0908-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Accepted: 04/26/2004] [Indexed: 01/03/2023]
Abstract
Like most neural systems, dopamine neurons of the substantia nigra undergo apoptotic natural cell death during development. In rodents, this occurs largely postnatally and is biphasic with an initial major peak just after birth and a second minor peak on postnatal day 14. As envisioned by classic neurotrophic theory, this event is regulated by interactions with the target of these neurons, the striatum, because a developmental target lesion results in an augmented natural cell death event with fewer nigral dopamine neurons surviving into adulthood. Until recently, the striatal target-derived neurotrophic factors providing developmental support of dopamine neurons were unknown, but there is now growing evidence that glial-cell-line-derived neurotrophic factor (GDNF) serves as a physiologic limiting neurotrophic factor for these neurons during the first phase of natural cell death. During this phase, intrastriatal injection of GDNF diminishes the natural cell death event and neutralizing antibodies augment it. Sustained overexpression of GDNF in the striatum throughout development in a unique double transgenic mouse model results in an increased number of dopamine neurons surviving the first phase of natural cell death. However, this increase does not persist into adulthood. Therefore, other factors or mechanisms must play important roles in the determination of the mature number of nigral dopamine neurons. Further elucidation of these mechanisms will be important in the development of neuroprotective and cell replacement therapies for Parkinson's disease.
Collapse
Affiliation(s)
- Robert E Burke
- Department of Neurology, The College of Physicians and Surgeons, Columbia University, 650 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
34
|
Block F, Loos M, Frohn C, Schwarz M. Association between inflammation and nigral neuronal damage following striatal excitotoxic lesion. Brain Res 2004; 998:29-35. [PMID: 14725965 DOI: 10.1016/j.brainres.2003.10.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We examined the expression of TNF-alpha within the substantia nigra pars reticulata (SNR) following intrastriatal injection of quinolinic acid (QA) and studied the effect of rolipram, a TNF-alpha-inhibitor, on the secondary neuronal damage. QA (240 nmol in 1 microl) was injected stereotactically into the striatum of male Wistar rats. After survival of 1, 3 or 10 days, the animals were sacrificed and immunohistochemical staining with an antibody against TNF-alpha was performed. From day 1 to day 10 after striatal QA injection TNF-alpha positive cells were observed within ipsilateral substantia nigra which were neither present on the contralateral side nor in sham-operated controls. Double labeling with antibodies against TNF-alpha and NeuN, keratan sulfate proteoglycan or GFAP displayed a good overlap between TNF-alpha and NeuN, which suggests that TNF-alpha positive cells are neurons. For the pharmacological approach, three groups of QA rats were treated intraperitoneally with either solvent (n=5), the NMDA receptor antagonist MK 801 (4 mg/kg, n=6) or the TNF-alpha inhibitor rolipram (0.3 mg/kg, n=6), which was started 24 h after QA-injection and continued with daily applications for 14 days. The amount of striatal damage did not differ between the three groups. The number of intact neurons within the ipsilateral substantia nigra of the solvent treated group was reduced by approximately 30% compared to the contralateral side. Both MK 801 and rolipram ameliorated this secondary damage and reduced the number of TNF-alpha positive cells. The observed association between expression of TNF-alpha and secondary neuronal damage within the substantia nigra induced by intrastriatal QA application might hint towards an involvement of this cytokine in transneuronal degeneration.
Collapse
Affiliation(s)
- F Block
- Department of Neurology, University of Aachen, Pauwelsstr. 30, D-52057 Aachen, Germany.
| | | | | | | |
Collapse
|
35
|
Costa C, Leone G, Saulle E, Pisani F, Bernardi G, Calabresi P. Coactivation of GABA
A
and GABA
B
Receptor Results in Neuroprotection During In Vitro Ischemia. Stroke 2004; 35:596-600. [PMID: 14726544 DOI: 10.1161/01.str.0000113691.32026.06] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
The possible neuroprotective effect of endogenous γ-aminobutyric acid (GABA) on the irreversible electrophysiological changes induced by in vitro ischemia on striatal neurons was investigated. In particular, the aim of the study was the characterization of the neuroprotective action of 2 antiepileptic drugs increasing GABAergic transmission such as tiagabine, a GABA transporter inhibitor, and vigabatrin, an irreversible inhibitor of GABA transaminase.
Methods—
Extracellular field potential recordings were obtained from rat corticostriatal slice preparations. In vitro ischemia was delivered by switching to an artificial cerebrospinal fluid solution in which glucose was omitted and oxygen was replaced with N
2
.
Results—
An irreversible loss of the field potentials recorded from striatal neurons was observed after 10 minutes of ischemia in control solution. Conversely, tiagabine and vigabatrin partially prevented the ischemia-induced field potential loss. Surprisingly, both GABA
A
and GABA
B
receptor antagonists blocked these effects. Accordingly, neuroprotection could be obtained only when GABA
A
and GABA
B
receptor agonists were coapplied, but not when a single agonist was given in isolation.
Conclusions—
Antiepileptic drugs targeting GABAergic transmission can exert neuroprotective effects against ischemia by increasing endogenous GABA levels and via the activation of both GABA
A
and GABA
B
receptors.
Collapse
Affiliation(s)
- Cinzia Costa
- Clinica Neurologica, Dipartimento di Neuroscienze, Università di Tor Vergata, Rome, Italy
| | | | | | | | | | | |
Collapse
|
36
|
McBride JL, Behrstock SP, Chen EY, Jakel RJ, Siegel I, Svendsen CN, Kordower JH. Human neural stem cell transplants improve motor function in a rat model of Huntington's disease. J Comp Neurol 2004; 475:211-9. [PMID: 15211462 DOI: 10.1002/cne.20176] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The present study investigated the neuroanatomical and behavioral effects of human stem cell transplants into the striatum of quinolinic acid (QA)-lesioned rats. Twenty-four rats received unilateral QA (200 nM/microl) injections into the striatum. One week later, rats were transplanted with stem cells derived from human fetal cortex (12 weeks postconception) that were either 1) pretreated in culture media with the differentiating cytokine ciliary neurotrophic factor (CNTF; n = 9) or 2) allowed to grow in culture media alone (n=7). Each rat was injected with a total of 200,000 cells. A third group of rats (n=8) was given a sham injection of vehicle. Rats transplanted with human stem cells performed significantly better over the 8 weeks of testing on the cylinder test compared with those treated with vehicle (P < or = 0.001). Stereological striatal volume analyses performed on Nissl-stained sections revealed that rats transplanted with CNTF-treated neurospheres had a 22% greater striatal volume on the lesioned side compared with those receiving transplants of untreated neurospheres (P = 0.0003) and a 26% greater striatal volume compared with rats injected with vehicle (P < or = 0.0001). Numerous human nuclei-positive cells were visualized in the striatum in both transplantation groups. Grafted cells were also observed in the globus pallidus, entopeduncular nucleus, and substantia nigra pars reticulata, areas of the basal ganglia receiving striatal projections. Some of the human nuclei-positive cells coexpressed glial fibrillary acidic protein and NeuN, suggesting that they had differentiated into neurons and astrocytes. Taken together, these data demonstrate that striatal transplants of human fetal stem cells elicit behavioral and anatomical recovery in a rodent model of Huntington's disease.
Collapse
Affiliation(s)
- Jodi L McBride
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Foster JA, Bezin L, Groc L, Christopherson PL, Levine RA. Kainic acid lesion-induced nigral neuronal death. J Chem Neuroanat 2003; 26:65-73. [PMID: 12954531 DOI: 10.1016/s0891-0618(03)00040-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive death of dopamine (DA) neurons in the substantia nigra pars compacta. We report a rat model that exhibits progressive death of nigral neurons following unilateral injection of kainic acid in the striatum. In situ end-labeling revealed significant numbers of dying nigral neurons ipsilateral to the lesion during the first 3 weeks following injection. An indication of the gradual nature of death was that similar small numbers of cells were detected at each time point. These early morphological markers of neuronal death led to a significant reduction (20%) at 5 months of tyrosine hydroxylase-positive neurons and total number of neurons in the ipsilateral substantia nigra compared with the contralateral control. To examine the role of nigrostriatal DA metabolism in the observed nigral neuronal death, we manipulated DA metabolism during the initial 2 weeks following kainic acid lesion. Neurons in the ventral tier of the substantia nigra pars compacta were protected from death by treatment with 2,4-diamino-6-hydroxy-pyrimidine (DAHP), an inhibitor of GTP cyclohydrolase, the initial enzyme in the synthesis of the tyrosine hydroxylase co-substrate, tetrahydrobiopterin (BH(4)). Neurons in both the dorsal and ventral tier of substantia nigra pars compacta were protected from death by treatment with DAHP and L-DOPA. These experiments suggest that intrastriatal kainic acid lesion is an in vivo model of trophic support withdrawal. This experimental procedure is useful for studying mechanisms underlying protracted death of nigral DA neurons and may provide valuable mechanistic information relevant to understanding the etiology of PD.
Collapse
Affiliation(s)
- Jane A Foster
- William T. Gossett Neurology Laboratories, Henry Ford Health System, 1 Ford Place, 4D, Detroit, MI 48202, USA.
| | | | | | | | | |
Collapse
|
38
|
Madden K, Clark W, Lessov N. Failure of ischemic neuroprotection by potentiators of gamma-aminobutyric acid. Clin Med Res 2003; 1:119-24. [PMID: 15931298 PMCID: PMC1069034 DOI: 10.3121/cmr.1.2.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2003] [Accepted: 03/04/2003] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Potentiators of inhibitory neurotransmission may provide a neuroprotective effect on cerebral tissue exposed to ischemia, without inducing toxic side effects. Topiramate and vigabatrin enhance the action of gamma-aminobutyric acid (GABA), and each has side effect profiles known to be well tolerated through their clinical use as anticonvulsant medications. We assessed the potential benefit through GABA activation by these drugs on infarct size and functional recovery following focal cerebral ischemia in mice. METHODS Silicon-coated suture was advanced through the internal carotid artery of 89 halothane-anesthetized mice to temporarily occlude the right middle cerebral artery for either 45 minutes (topiramate), or 120 minutes (vigabatrin). Animals were treated either at the time of reperfusion with topiramate (100 mg/kg, 40 mg/kg, or saline control), or two hours before arterial occlusion with vigabatrin, (1000 mg/kg, 500 mg/kg, or saline control). Neurological outcome was measured 24 hours after ischemia using a 28-point functional examination score. Infarct volume was estimated by summing area maps of stained slices of infarcted hemispheres. RESULTS Functional examination scores at 24 hours were similar between the high dose topiramate group, the low dose topiramate group, and the control group. Similarly, no differences were noted between examination scores of high dose vigabatrin, low dose vigabatrin, and control. Consistent sized right hemisphere infarcts were noted within each group on histological examination. Mean infarct volumes did not differ between groups treated with high dose topiramate, low dose topiramate, or control. Infarct volumes of animals treated with saline control were slightly larger than that of high dose vigabatrin and low dose vigabatrin groups, but the difference did not reach significance. CONCLUSION Treatment with these two potentiators of GABA did not result in significant differences in outcome following focal cerebral ischemia, by either functional or histological measures. These results do not support a substantial neuroprotective role of GABA following ischemia in this mouse suture model.
Collapse
Affiliation(s)
- Ken Madden
- Department of Neurology, Marshfield Clinic, Marshfield, Wisconsin 54449, USA.
| | | | | |
Collapse
|
39
|
Michelot D, Melendez-Howell LM. Amanita muscaria: chemistry, biology, toxicology, and ethnomycology. MYCOLOGICAL RESEARCH 2003; 107:131-46. [PMID: 12747324 DOI: 10.1017/s0953756203007305] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The fly agaric is a remarkable mushroom in many respects; these are its bearing, history, chemical components and the poisoning that it provokes when consumed. The 'pantherina' poisoning syndrome is characterized by central nervous system dysfunction. The main species responsible are Amanita muscaria and A. pantherina (Amanitaceae); however, some other species of the genus have been suspected for similar actions. Ibotenic acid and muscimol are the active components, and probably, some other substances detected in the latter species participate in the psychotropic effects. The use of the mushroom started in ancient times and is connected with mysticism. Current knowledge on the chemistry, toxicology, and biology relating to this mushroom is reviewed, together with distinctive features concerning this unique species.
Collapse
Affiliation(s)
- Didier Michelot
- Muséum National d'Histoire Naturelle, Institut Régulation et Développement, Diversité Moléculaire, Chimie et Biochimie des Substances Naturelles, USM 502 UMR 8041 C.N.R.S., 63 rue de Buffon, F-75005 Paris, France.
| | | |
Collapse
|
40
|
Loos M, Dihné M, Block F. Tumor necrosis factor-α expression in areas of remote degeneration following middle cerebral artery occlusion of the rat. Neuroscience 2003; 122:373-80. [PMID: 14614903 DOI: 10.1016/s0306-4522(03)00498-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Remote areas undergoing delayed neuronal degeneration after focal brain ischemia display a preceding glial activation. The expression of proinflammatory cytokines there has not been examined so far. We examined the expression of TNFalpha in the thalamus and the substantia nigra pars reticulata (SNr) 1, 3 and 7 days after transient middle cerebral artery occlusion (MCAO) of the rat. We used antibodies against glial fibrillary acidic protein (GFAP), OX-42, NeuN and tumor necrosis factor-alpha (TNFalpha) for immunohistochemistry/double-immunofluorescent labeling to investigate the time course of glial activation and the cellular localization of TNFalpha. Neuronal degeneration was measured by means of cell counting in Nissl-stained sections. In the ipsilateral thalamus, TNFalpha was upregulated already 1 day after MCAO. Microglia and astroglia were activated after 3 days. A cellular colocalisation of GFAP and TNFalpha was observed. Neuronal degeneration was evident at day 14. In the SNr, TNFalpha expression was enhanced 3 days after MCAO. Microglia was activated after 3 days and astroglia after 7 days. A cellular colocalisation of NeuN and TNFalpha was observed. Neuronal degeneration was evident at day 14. Thus, in both areas, expression of TNFalpha precedes astrogliosis and neuronal degeneration. The different patterns of TNFalpha upregulation of the substantia nigra pars reticulata and the thalamus following middle cerebral artery occlusion may reflect different pathophysiological mechanisms leading to remote neuronal degeneration.
Collapse
Affiliation(s)
- M Loos
- Department of Neurology, Rheinisch Westfälische Technische Hochschule, Aachen, Pauwelsstrasse 30, D-52057 Aachen, Germany.
| | | | | |
Collapse
|
41
|
Dihné M, Grommes C, Lutzenburg M, Witte OW, Block F. Different mechanisms of secondary neuronal damage in thalamic nuclei after focal cerebral ischemia in rats. Stroke 2002; 33:3006-11. [PMID: 12468804 DOI: 10.1161/01.str.0000039406.64644.cb] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE After focal cerebral ischemia, depending on its localization and extent, secondary neuronal damage may occur that is remote from the initial lesion. In this study differences in secondary damage of the ventroposterior thalamic nucleus (VPN) and the reticular thalamic nucleus (RTN) were investigated with the use of different ischemia models. METHODS Transient middle cerebral artery occlusion (MCAO) leads to cortical infarction, including parts of the basal ganglia such as the globus pallidus, and to widespread edema. Photothrombotic ischemia generates pure cortical infarcts sparing the basal ganglia and with only minor edema. Neuronal degeneration was quantified within the ipsilateral RTN and VPN 14 days after ischemia. Glial reactions were studied with the use of immunohistochemistry. RESULTS MCAO resulted in delayed neuronal cell loss of the ipsilateral VPN and RTN. Glial activation occurred in both nuclei beginning after 24 hours. Photothrombotic ischemia resulted in delayed neuronal cell loss only within the VPN. Even 2 weeks after photothrombotic ischemia, glial activation could only be seen within the VPN. CONCLUSIONS Pure cortical infarcts after photothrombotic ischemia, without major edema and without effects on the globus pallidus of the basal ganglia, only lead to secondary VPN damage that is possibly due to retrograde degeneration. MCAO, which results in infarction of cortex and globus pallidus and which causes widespread edema, leads to secondary damage in the VPN and RTN. Thus, additional RTN damage may be due to loss of protective GABAergic input from the globus pallidus to the RTN or due to the extensive edema. Retrograde degeneration is not possible because the RTN, in contrast to the VPN, has no efferents to the cortex.
Collapse
Affiliation(s)
- Marcel Dihné
- Department of Neurology, Rheinisch Westfälische Technische Hochschule-Aachen, Aachen, Germany
| | | | | | | | | |
Collapse
|
42
|
Kanemitsu H, Nakagomi T, Tamura A, Tsuchiya T, Kono G, Sano K. Differences in the extent of primary ischemic damage between middle cerebral artery coagulation and intraluminal occlusion models. J Cereb Blood Flow Metab 2002; 22:1196-204. [PMID: 12368658 DOI: 10.1097/01.wcb.0000037992.07114.95] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The authors studied the differences between heat-shock/stress protein 70 (hsp70) gene expression and protein synthesis in the unilateral middle cerebral artery (MCA) microsurgical direct occlusion (Tamura's) model and the unilateral intraluminal occlusion model. In Tamura's model, expression of hsp70 mRNA and HSP70 protein and decreased protein synthesis were detected in the ischemic areas, including the ipsilateral cortex and caudate. These phenomena, however, were not observed in the areas outside the MCA territory, including the ipsilateral thalamus, hippocampus, and substantia nigra. These results were consistent among the experimental rats. In the intraluminal occlusion model, however, induction of both hsp70 mRNA and HSP70 protein and impairment of protein synthesis were noted in the areas outside the MCA territory, including the ipsilateral thalamus, hypothalamus, hippocampus, and substantia nigra, as well as in the MCA territory, including the ipsilateral cortex and caudate. These results were not consistent among the experimental rats. These different results might be due to widespread damage resulting from internal carotid artery (ICA) occlusion in the intraluminal occlusion model. Accordingly, the authors suggest that this model be called an ICA occlusion model, rather than a pure MCA occlusion model.
Collapse
Affiliation(s)
- Hideaki Kanemitsu
- Department of Neurosurgery, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
Sharshar T, Gray F, Poron F, Raphael JC, Gajdos P, Annane D. Multifocal necrotizing leukoencephalopathy in septic shock. Crit Care Med 2002; 30:2371-5. [PMID: 12394971 DOI: 10.1097/00003246-200210000-00031] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Multifocal necrotizing leukoencephalopathy, characterized by multiple microscopic foci of necrosis involving the white matter of the pons, has been described mainly after chemotherapy or radiotherapy for brain cancer and in HIV infection. The role of circulating cytokines has been suggested but remains to be assessed. DESIGN Prospective case series. SETTING A 26-bed general medical intensive care unit at a university hospital. PATIENTS Septic shock patients. MEASUREMENTS AND PATIENTS: In three patients who died from septic shock, careful postmortem examination of the brain was performed, including studies of neuronal apoptosis and cytokine expression. MAIN RESULTS In one patient, typical lesions of multifocal necrotizing leukoencephalopathy were seen. As compared with control 1 and control 2 who did not have multifocal necrotizing leukoencephalopathy, marked lesions of the pons, including vacuolization, apoptosis, microglial activation, and expression of tumor necrosis factor-alpha and interleukin-1beta, were observed in the case. Simultaneously, case 1 had markedly increased circulating levels for tumor necrosis factor-alpha, interleukin-1beta, interleukin-6, interleukin-8, interleukin-10, soluble tumor necrosis factor receptor II, and for interleukin-1 receptor antagonist. CONCLUSION Septic shock is a newly described cause of multifocal necrotizing leukoencephalopathy, probably mediated by an excessive systemic inflammatory response.
Collapse
Affiliation(s)
- Tarek Sharshar
- Service de Réanimation Médiale, Hôpital Raymond Poincaré--Faculté de Médecine Paris--Ouest--Université Paris, Garches, France
| | | | | | | | | | | |
Collapse
|
44
|
Differences in the Extent of Primary Ischemic Damage Between Middle Cerebral Artery Coagulation and Intraluminal Occlusion Models. J Cereb Blood Flow Metab 2002. [DOI: 10.1097/00004647-200210000-00007] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Zhao F, Kuroiwa T, Miyasaka N, Nagaoka T, Nakane M, Tamura A, Mizusawa H. Ultrastructural and MRI study of the substantia nigra evolving exofocal post-ischemic neuronal death in the rat. Neuropathology 2002; 22:91-105. [PMID: 12416550 DOI: 10.1046/j.1440-1789.2002.00437.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To clarify the morphological characteristics of exofocal post-ischemic neuronal death (EPND) in the substantia nigra (SN), we investigated the course of light- and electron-microscopic changes of the SN of rats subjected to occlusion of the left middle cerebral artery (MCA) for 1, 2, 4, 7 and 12 days. To assess cellular edema, sequential magnetic resonance (MR) mapping of the apparent diffusion coefficient (ADC) and the T2 value test was performed. Histological and electron-microscopic examination on day 1 showed dotted chromatin clumps in the nuclei of some neurons and mild swelling of the perivascular endfeet of astrocytes in the ipsilateral SN. On day 2, a few cells of the ipsilateral SN pars reticulata (SNr) revealed key morphological signs of apoptosis--apoptotic body-like condensation and segregation of the chromatin and DNA fragmentation-like nuclear remnants. On day 4, 38% of neurons became swollen (pale neurons) with cytoplasmic microvacuoles, which appeared to originate from rough endoplasmic reticulum (rER), mitochondria and Golgi apparatus. Twenty percent of neurons showed massive proliferation of the cisternae of the rER, some of which were fragmented or had lost their normal parallel arrangement. In addition, MR mapping revealed a transient ADC decrease with a T2 increase (signifying a phase of cellular edema), which coordinated with the phase of ultrastructural cellular swelling. Further, the total number of neurons started to decrease gradually, the perivascular endfeet of astrocytes were markedly swollen, and the neuropil became loose on day 4. On day 7, reactive astrocytes and dark neurons occurred most frequently. These results suggest that the EPND in the SN after occlusion of the MCA in adult rats is due to both apoptosis and necrosis, although necrosis seems to be the dominant mechanism of the EPND. However, the morphologic resemblances of EPND to delayed neuronal death suggest these processes have a common pathomechanism.
Collapse
Affiliation(s)
- Fengyu Zhao
- Department of Neurology and Neurological Science, Medical Research Institute, Tokyo Medical and Dental University, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Romero J, Lastres-Becker I, de Miguel R, Berrendero F, Ramos JA, Fernández-Ruiz J. The endogenous cannabinoid system and the basal ganglia. biochemical, pharmacological, and therapeutic aspects. Pharmacol Ther 2002; 95:137-52. [PMID: 12182961 DOI: 10.1016/s0163-7258(02)00253-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
New data strengthen the idea of a prominent role for endocannabinoids in the modulation of a wide variety of neurobiological functions. Among these, one of the most important is the control of movement. This finding is supported by 3 lines of evidence: (1) the demonstration of a powerful action, mostly inhibitory in nature, of synthetic and plant-derived cannabinoids and, more recently, of endocannabinoids on motor activity; (2) the presence of the cannabinoid CB(1) receptor subtype and the recent description of endocannabinoids in the basal ganglia and the cerebellum, the areas that control movement; and (3) the fact that CB(1) receptor binding was altered in the basal ganglia of humans affected by several neurological diseases and also of rodents with experimentally induced motor disorders. Based on this evidence, it has been suggested that new synthetic compounds that act at key steps of endocannabinoid activity (i.e., more-stable analogs of endocannabinoids, inhibitors of endocannabinoid reuptake or metabolism, antagonists of CB(1) receptors) might be of interest for their potential use as therapeutic agents in a variety of pathologies affecting extrapyramidal structures, such as Parkinson's and Huntington's diseases. Currently, only a few data exist in the literature studying such relationships in humans, but an increasing number of journal articles are revealing the importance of this new neuromodulatory system and arguing in favour of the funding of more extensive research in this field. The present article will review the current knowledge of this neuromodulatory system, trying to establish the future lines for research on the therapeutic potential of the endocannabinoid system in motor disorders.
Collapse
Affiliation(s)
- Julián Romero
- Laboratorio de Apoyo a la Investigación, Fundación Hospital Alcorcón, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
47
|
Kilpatrick IC, Neal JW, Pearson RC, Powell TP. Lesions of the Cerebral Cortex and Caudate-Putamen Enhance GABA Function in the Rat Superior Colliculus. Eur J Neurosci 2002; 3:971-80. [PMID: 12106255 DOI: 10.1111/j.1460-9568.1991.tb00033.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Unilateral lesions of the rat frontal cortex were made either alone or in combination with the caudate-putamen in order to examine (a) their morphological influence on the substantia nigra and (b) their neurochemical influence on GABA function in the superior colliculus. One to two months following the combined lesion, neuronal somata in the ipsilateral pars reticulata of the substantia nigra were clearly hypertrophied (+ 30%). Morphological changes in the substantia nigra were not evident contralaterally or in animals bearing only cortical lesions. One to two months following cortex-only lesions, no significant alterations in tectal GABA concentration were observed. However, the combined lesion induced elevations of GABA within both the medial and lateral sectors of the intermediate and deep layers of the superior colliculus. This effect was restricted to the ipsilateral side and was most pronounced in lateral sectors. The vast majority of GABA released from superfused control tectal slices by a depolarizing stimulus (35 mM KCl) was calcium-dependent. Such evoked GABA release from ipsilateral tectal slices was significantly reduced (- 25%) by unilateral lesions of the substantia nigra, a structure that is known to provide GABA-containing inputs to the tectum. In contrast, cortical lesions alone significantly enhanced the evoked tectal GABA release (+ 66%), although their influence was again confined to the ipsilateral side. Combined lesions of the cerebral cortex and caudate-putamen significantly enhanced the evoked GABA release from tectal slices in both hemispheres but the changes were most marked ipsilaterally (+ 147%). It is suggested that the hypertrophy of GABA-containing nigrotectal somata seen after removal of corticostriatal, corticotectal and in particular GABA-containing striatonigral fibres may reflect concomitant increases in GABA synthesis within and/or sprouting of nigrotectal terminals.
Collapse
Affiliation(s)
- I C Kilpatrick
- Department of Pharmacology, School of Medical Sciences, University Walk, Bristol BS8 1TD, UK and
| | | | | | | |
Collapse
|
48
|
Hutchinson PJ, O'Connell MT, Coles JP, Chatfield DA, Coleman MR, Al-Rawi PG, Kett-White CR, Gupta AK, Menon DK, Boniface SJ, Heazell M, Kirkpatrick PJ, Pickard JD. Investigation of the effect of chlormethiazole on cerebral chemistry in neurosurgical patients: a combined study of microdialysis and a neuroprotective agent. Br J Clin Pharmacol 2002; 53:275-83. [PMID: 11874391 PMCID: PMC1874304 DOI: 10.1046/j.0306-5251.2001.01552.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2001] [Accepted: 10/18/2001] [Indexed: 11/20/2022] Open
Abstract
AIMS Promising pre-clinical results from laboratory studies of neuro-protective drugs for the treatment of patients with stroke and head injury have not been translated into benefit during clinical trials. The objective of the study was to assess the feasibility of administrating a potential neuro-protective drug (chlormethiazole) in conjunction with multimodality monitoring (including microdialysis) to patients with severe head injury in order to determine the effect of the agent on surrogate endpoints and penetration into the brain. METHODS Multimodality monitoring including cerebral and peripheral microdialysis was applied to five head-injured patients on the neuro-intensive care unit. Chlormethiazole (0.8%) was administered as a rapid (10 ml min(-1)) intravenous loading infusion for 5 min followed by a slow (1 ml min(-1)) continuous infusion for 60 min. The following parameters were monitored: heart rate, mean arterial blood pressure, intracranial pressure, cerebral perfusion pressure, peripheral oxygen saturation, continuous arterial oxygen partial pressure, arterial carbon dioxide partial pressure, arterial pH, arterial temperature, cerebral tissue oxygen pressure, cerebral tissue carbon dioxide pressure, cerebral pH, cerebral temperature, electroencephalograph (EEG), bi-spectral index, plasma glucose, plasma chlormethiazole, and cerebral and peripheral microdialysis assay for chlormethiazole, glucose, lactate, pyruvate and amino acids. RESULTS Despite achieving adequate plasma concentrations, chlormethiazole was not detected in the peripheral or cerebral microdialysis samples. The drug was well tolerated and did not induce hypotension, hyperglycaemia or withdrawal seizures. The drug did not change the values of the physiological or chemical parameters including levels of GABA, lactate/pyruvate ratio and glutamate. The drug did, however, induce EEG changes, including burst suppression in two patients. CONCLUSIONS Chlormethiazole can be safely given to ventilated patients with severe head injury. There was no evidence of hypotension or withdrawal seizures. Combining a pilot clinical study of a neuro-protective agent with multimodality monitoring is feasible and, despite the lack of effect on physiological and chemical parameters in this study, may be a useful adjunct to the development of neuro-protective drugs in the future. Further investigation of the capability of microdialysis in this setting is required. By investigating the effect of a drug on surrogate end-points, it may be possible to identify promising agents from small pilot clinical studies before embarking on large phase III clinical trials.
Collapse
Affiliation(s)
- P J Hutchinson
- Department of Neurosurgery, Addenbrooke's Hospital, University of Cambridge, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hutchinson PJ, O'Connell MT, Al-Rawi PG, Kett-White CR, Gupta AK, Maskell LB, Pickard JD, Kirkpatrick PJ. Increases in GABA concentrations during cerebral ischaemia: a microdialysis study of extracellular amino acids. J Neurol Neurosurg Psychiatry 2002; 72:99-105. [PMID: 11784833 PMCID: PMC1737714 DOI: 10.1136/jnnp.72.1.99] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Increases in the extracellular concentration of the excitatory amino acids glutamate and aspartate during cerebral ischaemia in patients are well recognised. Less emphasis has been placed on the concentrations of the inhibitory amino acid neurotransmitters, notably gamma-amino-butyric acid (GABA), despite evidence from animal studies that GABA may act as a neuroprotectant in models of ischaemia. The objective of this study was to investigate the concentrations of various excitatory, inhibitory and non-transmitter amino acids under basal conditions and during periods of cerebral ischaemia in patients with head injury or a subarachnoid haemorrhage. METHODS Cerebral microdialysis was established in 12 patients with head injury (n=7) or subarachnoid haemorrhage (n=5). Analysis was performed using high performance liquid chromatography for a total of 19 (excitatory, inhibitory and non-transmitter) amino acids. Patients were monitored in neurointensive care or during aneurysm clipping. RESULTS During stable periods of monitoring the concentrations of amino acids were relatively constant enabling basal values to be established. In six patients, cerebral ischaemia was associated with increases (up to 1350 fold) in the concentration of GABA, in addition to the glutamate and aspartate. Parallel increases in the concentration of glutamate and GABA were found (r=0.71, p<0.005). CONCLUSIONS The results suggest that, in the human brain, acute cerebral ischaemia is not accompanied by an imbalance between excitatory and inhibitory amino acids, but by an increase in all neurotransmitter amino acids. These findings concur with the animal models of ischaemia and raise the possibility of an endogenous GABA mediated neuroprotective mechanism in humans.
Collapse
Affiliation(s)
- P J Hutchinson
- Academic Department of Neurosurgery, University of Cambridge, Box 167, Addenbrooke's Hospital, Cambridge CB2 2QQ, UK.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Cervantes M, González-Vidal MD, Ruelas R, Escobar A, Moralí G. Neuroprotective effects of progesterone on damage elicited by acute global cerebral ischemia in neurons of the caudate nucleus. Arch Med Res 2002; 33:6-14. [PMID: 11825624 DOI: 10.1016/s0188-4409(01)00347-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND In addition to the hippocampus, the dorsolateral caudate nucleus (CN) and the pars reticularis of the substantia nigra (SNr) are among the most vulnerable brain areas to ischemia. A possible association of the neuronal injury in these two subcortical nuclei has been proposed, the primary damage affecting the CN GABAergic neurons innervating the SNr, and secondarily the SNr neurons as a result of an imbalance of GABAergic and glutamatergic input to the SNr. Progesterone (P(4)) exerts a GABAergic action on the central nervous system (CNS) and is known to protect neurons in the cat hippocampus from the damaging effect of acute global cerebral ischemia (AGCI). The effects of AGCI on the neuronal populations of the CN and SNr, in addition to the possible neuroprotective effects of P(4), were assessed in cats in the present study. METHODS Ovariectomized adult cats were treated subcutaneously (s.c.) with either P(4) (10 mg/kg/day) or corn oil during the 7 days before and 7 days after being subjected to a period of AGCI by 15 min of cardiorespiratory arrest followed by 4 min of reanimation. After 14 days of survival, animals were sacrificed and their brains perfused in situ with phosphate-buffered 10% formaldehyde for histologic examination. RESULTS ACGI resulted in an intense glial reaction in the CN and a significant loss (43%) of medium-sized neurons of the CN, but no difference was found in the densities of SNr neurons between controls and ischemic oil- and P(4)-treated cats. Progesterone treatment completely prevented CN neuronal loss. CONCLUSIONS The overall results point to the higher vulnerability of CN neurons to ischemia as compared to neurons in the SNr and show the protective effects of P(4) upon CN neuronal damage after ischemia.
Collapse
Affiliation(s)
- Miguel Cervantes
- Laboratorio de Neurofarmacología, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social (IMSS), Morelia, Michoacán, Mexico
| | | | | | | | | |
Collapse
|