1
|
Iorgu AM, Vasilescu AN, Pfeiffer N, Spanagel R, Mallien AS, Inta D, Gass P. Psilocybin does not induce the vulnerability marker HSP70 in neurons susceptible to Olney's lesions. Eur Arch Psychiatry Clin Neurosci 2024; 274:1013-1019. [PMID: 37934233 PMCID: PMC11127870 DOI: 10.1007/s00406-023-01699-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/02/2023] [Indexed: 11/08/2023]
Abstract
S-ketamine, a N-methyl-D-aspartate receptor (NMDAR) antagonist, and psilocybin, a 5-hydroxy-tryptamine (serotonin) 2A receptor (5-HT2AR) agonist, are reported as effective rapid-acting antidepressants. Both compounds increase glutamate signalling and evoke cortical hyperexcitation. S-ketamine induces neurotoxicity especially in the retrosplenial cortex (Olney's lesions). Whether psilocybin produces similar neurotoxic effects has so far not been investigated. We performed an immunohistochemical whole-brain mapping for heat shock protein 70 (HSP70) in rats treated with psilocybin, S-ketamine, and MK-801. In contrast to S-ketamine- and MK-801-treated animals, we did not detect any HSP70-positive neurons in retrosplenial cortex of rats treated with psilocybin. Our results suggest that psilocybin might be safer for clinical use compared to S-ketamine regarding neuronal damage.
Collapse
Affiliation(s)
- Ana-Maria Iorgu
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany.
| | - Andrei-Nicolae Vasilescu
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Natascha Pfeiffer
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anne Stephanie Mallien
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| | - Dragos Inta
- Department for Community Health, Faculty of Natural Sciences and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Research Group Animal Models in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, J5, 68159, Mannheim, Germany
| |
Collapse
|
2
|
Davletshin AI, Matveeva AA, Poletaeva II, Evgen'ev MB, Garbuz DG. The role of molecular chaperones in the mechanisms of epileptogenesis. Cell Stress Chaperones 2023; 28:599-619. [PMID: 37755620 PMCID: PMC10746656 DOI: 10.1007/s12192-023-01378-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/28/2023] Open
Abstract
Epilepsy is a group of neurological diseases which requires significant economic costs for the treatment and care of patients. The central point of epileptogenesis stems from the failure of synaptic signal transmission mechanisms, leading to excessive synchronous excitation of neurons and characteristic epileptic electroencephalogram activity, in typical cases being manifested as seizures and loss of consciousness. The causes of epilepsy are extremely diverse, which is one of the reasons for the complexity of selecting a treatment regimen for each individual case and the high frequency of pharmacoresistant cases. Therefore, the search for new drugs and methods of epilepsy treatment requires an advanced study of the molecular mechanisms of epileptogenesis. In this regard, the investigation of molecular chaperones as potential mediators of epileptogenesis seems promising because the chaperones are involved in the processing and regulation of the activity of many key proteins directly responsible for the generation of abnormal neuronal excitation in epilepsy. In this review, we try to systematize current data on the role of molecular chaperones in epileptogenesis and discuss the prospects for the use of chemical modulators of various chaperone groups' activity as promising antiepileptic drugs.
Collapse
Affiliation(s)
| | - Anna A Matveeva
- Engelhardt Institute of Molecular Biology RAS, 119991, Moscow, Russia
- Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Moscow Region, Russia
| | - Inga I Poletaeva
- Biology Department, Lomonosov Moscow State University, 119991, Moscow, Russia
| | | | - David G Garbuz
- Engelhardt Institute of Molecular Biology RAS, 119991, Moscow, Russia
| |
Collapse
|
3
|
Moyano P, Sola E, Naval MV, Guerra-Menéndez L, Fernández MDLC, del Pino J. Neurodegenerative Proteinopathies Induced by Environmental Pollutants: Heat Shock Proteins and Proteasome as Promising Therapeutic Tools. Pharmaceutics 2023; 15:2048. [PMID: 37631262 PMCID: PMC10458078 DOI: 10.3390/pharmaceutics15082048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Environmental pollutants' (EPs) amount and diversity have increased in recent years due to anthropogenic activity. Several neurodegenerative diseases (NDs) are theorized to be related to EPs, as their incidence has increased in a similar way to human EPs exposure and they reproduce the main ND hallmarks. EPs induce several neurotoxic effects, including accumulation and gradual deposition of misfolded toxic proteins, producing neuronal malfunction and cell death. Cells possess different mechanisms to eliminate these toxic proteins, including heat shock proteins (HSPs) and the proteasome system. The accumulation and deleterious effects of toxic proteins are induced through HSPs and disruption of proteasome proteins' homeostatic function by exposure to EPs. A therapeutic approach has been proposed to reduce accumulation of toxic proteins through treatment with recombinant HSPs/proteasome or the use of compounds that increase their expression or activity. Our aim is to review the current literature on NDs related to EP exposure and their relationship with the disruption of the proteasome system and HSPs, as well as to discuss the toxic effects of dysfunction of HSPs and proteasome and the contradictory effects described in the literature. Lastly, we cover the therapeutic use of developed drugs and recombinant proteasome/HSPs to eliminate toxic proteins and prevent/treat EP-induced neurodegeneration.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Emma Sola
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - María Victoria Naval
- Department of Pharmacology, Pharmacognosy and Bothanic, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Lucia Guerra-Menéndez
- Department of Physiology, Medicine School, San Pablo CEU University, 28003 Madrid, Spain
| | - Maria De la Cabeza Fernández
- Department of Chemistry and Pharmaceutical Sciences, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Javier del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
4
|
Rapastinel alleviates the neurotoxic effect induced by NMDA receptor blockade in the early postnatal mouse brain. Eur Arch Psychiatry Clin Neurosci 2021; 271:1587-1591. [PMID: 32789675 DOI: 10.1007/s00406-020-01180-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/31/2020] [Indexed: 10/23/2022]
Abstract
Rapastinel is a novel psychoactive substance that acts as an N-methyl-D-aspartate-receptor (NMDAR) agonist and triggers antidepressant- and antipsychotic-like effects in animal models. However, it is unknown if rapastinel possesses a better side-effect profile than fast-acting glutamatergic antidepressants, like ketamine, which trigger neurotoxicity in the perinatal rodent cortex and protracted schizophrenia-like alterations. Here we found a remarkable neuroprotective effect of rapastinel against apoptosis induced by the NMDAR antagonist MK-801 in comparison to that elicited by clozapine and the mGlu2/3 agonist LY354740. These results suggest the potential therapeutic/prophylactic effect of rapastinel in ameliorating deleterious effects induced by NMDAR blockade during neurodevelopment.
Collapse
|
5
|
Zummo L, Vitale AM, Caruso Bavisotto C, De Curtis M, Garbelli R, Giallonardo AT, Di Bonaventura C, Fanella M, Conway de Macario E, Cappello F, Macario AJL, Marino Gammazza A. Molecular Chaperones and miRNAs in Epilepsy: Pathogenic Implications and Therapeutic Prospects. Int J Mol Sci 2021; 22:ijms22168601. [PMID: 34445306 PMCID: PMC8395327 DOI: 10.3390/ijms22168601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a pathologic condition with high prevalence and devastating consequences for the patient and its entourage. Means for accurate diagnosis of type, patient monitoring for predicting seizures and follow up, and efficacious treatment are desperately needed. To improve this adverse outcome, miRNAs and the chaperone system (CS) are promising targets to understand pathogenic mechanisms and for developing theranostics applications. miRNAs implicated in conditions known or suspected to favor seizures such as neuroinflammation, to promote epileptic tolerance and neuronal survival, to regulate seizures, and others showing variations in expression levels related to seizures are promising candidates as useful biomarkers for diagnosis and patient monitoring, and as targets for developing novel therapies. Components of the CS are also promising as biomarkers and as therapeutic targets, since they participate in epileptogenic pathways and in cytoprotective mechanisms in various epileptogenic brain areas, even if what they do and how is not yet clear. The data in this review should help in the identification of molecular targets among the discussed miRNAs and CS components for research aiming at understanding epileptogenic mechanisms and, subsequently, develop means for predicting/preventing seizures and treating the disease.
Collapse
Affiliation(s)
- Leila Zummo
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy; (L.Z.); (A.M.V.); (C.C.B.); (F.C.)
- Department of Neurology and Stroke Unit, A.R.N.A.S. Ospedale Civico—Di Cristina Benfratelli, 90127 Palermo, Italy
| | - Alessandra Maria Vitale
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy; (L.Z.); (A.M.V.); (C.C.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy; (L.Z.); (A.M.V.); (C.C.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
| | - Marco De Curtis
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.D.C.); (R.G.)
| | - Rita Garbelli
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (M.D.C.); (R.G.)
| | - Anna Teresa Giallonardo
- Department of Human Neurosciences “Sapienza”, University of Rome, 00185 Rome, Italy; (A.T.G.); (C.D.B.); (M.F.)
- Policlinico Umberto I, 00161 Rome, Italy
| | - Carlo Di Bonaventura
- Department of Human Neurosciences “Sapienza”, University of Rome, 00185 Rome, Italy; (A.T.G.); (C.D.B.); (M.F.)
- Policlinico Umberto I, 00161 Rome, Italy
| | - Martina Fanella
- Department of Human Neurosciences “Sapienza”, University of Rome, 00185 Rome, Italy; (A.T.G.); (C.D.B.); (M.F.)
- Policlinico Umberto I, 00161 Rome, Italy
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA;
| | - Francesco Cappello
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy; (L.Z.); (A.M.V.); (C.C.B.); (F.C.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA;
| | - Antonella Marino Gammazza
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, Section of Human Anatomy, University of Palermo, 90127 Palermo, Italy; (L.Z.); (A.M.V.); (C.C.B.); (F.C.)
- Correspondence:
| |
Collapse
|
6
|
Advances in the Development of Biomarkers for Poststroke Epilepsy. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5567046. [PMID: 33959658 PMCID: PMC8075663 DOI: 10.1155/2021/5567046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 12/23/2022]
Abstract
Stroke is the main cause of acquired epilepsy in elderly people. Poststroke epilepsy (PSE) not only affects functional recovery after stroke but also brings considerable social consequences. While some factors such as cortical involvement, hemorrhagic transformation, and stroke severity are associated with increased seizure risk, so far that remains controversial. In recent years, there are an increasing number of studies on potential biomarkers of PSE as tools for diagnosing and predicting epileptic seizures. Biomarkers such as interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), glutamate, and S100 calcium-binding protein B (S100B) in blood are associated with the occurrence of PSE. This review is aimed at summarizing the progress on potential biomarkers of PSE.
Collapse
|
7
|
Mallien AS, Pfeiffer N, Vogt MA, Chourbaji S, Sprengel R, Gass P, Inta D. Cre-Activation in ErbB4-Positive Neurons of Floxed Grin1/NMDA Receptor Mice Is Not Associated With Major Behavioral Impairment. Front Psychiatry 2021; 12:750106. [PMID: 34899420 PMCID: PMC8660629 DOI: 10.3389/fpsyt.2021.750106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Extensive evidence suggests a dysfunction of the glutamate NMDA receptor (NMDAR) in schizophrenia, a severe psychiatric disorder with putative early neurodevelopmental origins, but clinical onset mainly during late adolescence. On the other hand, pharmacological models using NMDAR antagonists and the clinical manifestation of anti-NMDAR encephalitis indicate that NMDAR blockade/hypofunction can trigger psychosis also at adult stages, without any early developmental dysfunction. Previous genetic models of NMDAR hypofunction restricted to parvalbumin-positive interneurons indicate the necessity of an early postnatal impairment to trigger schizophrenia-like abnormalities, whereas the cellular substrates of NMDAR-mediated psychosis at adolescent/adult stages are unknown. Neuregulin 1 (NRG1) and its receptor ErbB4 represent schizophrenia-associated susceptibility factors that closely interact with NMDAR. To determine the neuronal populations implicated in "late" NMDAR-driven psychosis, we analyzed the effect of the inducible ablation of NMDARs in ErbB4-expressing cells in mice during late adolescence using a pharmacogenetic approach. Interestingly, the tamoxifen-inducible NMDAR deletion during this late developmental stage did not induce behavioral alterations resembling depression, schizophrenia or anxiety. Our data indicate that post-adolescent NMDAR deletion, even in a wider cell population than parvalbumin-positive interneurons, is also not sufficient to generate behavioral abnormalities resembling psychiatric disorders. Other neuronal substrates that have to be revealed by future studies, may underlie post-adolescent NMDAR-driven psychosis.
Collapse
Affiliation(s)
- Anne S Mallien
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, RG Animal Models in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Natascha Pfeiffer
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, RG Animal Models in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Miriam A Vogt
- Interfaculty Biomedical Research Facility (IBF), Heidelberg University, Heidelberg, Germany
| | - Sabine Chourbaji
- Interfaculty Biomedical Research Facility (IBF), Heidelberg University, Heidelberg, Germany
| | - Rolf Sprengel
- Research Group of the Max Planck Institute for Medical Research at the Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.,Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, RG Animal Models in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Dragos Inta
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, RG Animal Models in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| |
Collapse
|
8
|
Effects of GLP-1 Receptor Activation on a Pentylenetetrazole-Kindling Rat Model. Brain Sci 2019; 9:brainsci9050108. [PMID: 31091715 PMCID: PMC6562858 DOI: 10.3390/brainsci9050108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives: To study the possible anti-seizure and neuroprotective effect of glucagon like peptide 1 (GLP1) analogue (liraglutide) in a pentylenetetrazole (PTZ) induced kindled rat model and its underlying mechanisms. Methods: Thirty Sprague Dawley rats were allocated into 3 equal groups; i) Normal group: normal rats received normal saline, ii) PTZ (kindling) group: received PTZ (50 mg/Kg intraperitoneally (i.p.)) every other day for 2 weeks and iii) PTZ + GLP1 group: same as the PTZ group but rats received liraglutide (75 µg/kg i.p. daily) for 2 weeks before PTZ injection. Seizure severity score, seizure latency and duration were assessed. Also, the expression of caspase-3 (apoptotic marker) and β-catenin (Wnt pathway) by western blotting, markers of oxidative stress (GSH, CAT and MDA) by biochemical assay and the expression of LC3 (marker of autophagy) and heat shock protein 70 (Hsp70) by immunostaining were assessed in hippocampal regions of brain tissues. Results: PTZ caused a significant increase in Racine score and seizure duration with a significant decrease in seizure latency. These effects were associated with a significant increase in MDA, β-catenin, caspase-3, Hsp70 and LC3 in brain tissues (p < 0.05). Meanwhile, liraglutide treatment caused significant attenuation in PTZ-induced seizures, which were associated with significant improvement in markers of oxidative stress, reduction in LC3, caspase-3 and β-catenin and marked increase in Hsp70 in hippocampal regions (p < 0.05). Conclusion: Activation of GLP1R might have anticonvulsant and neuroprotective effects against PTZ-induced epilepsy. These effects could be due to suppression of oxidative stress, apoptosis and autophagy and upregulation of Hsp70.
Collapse
|
9
|
Hussein AM, Eldosoky M, El-Shafey M, El-Mesery M, Ali AN, Abbas KM, Abulseoud OA. Effects of metformin on apoptosis and α-synuclein in a rat model of pentylenetetrazole-induced epilepsy. Can J Physiol Pharmacol 2018; 97:37-46. [PMID: 30308130 DOI: 10.1139/cjpp-2018-0266] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The present study was designed to examine the possible neuroprotective and antiepileptic effects of metformin (Metf) in a rat model of pentylenetetrazole (PTZ)-induced epilepsy and its possible underlying mechanisms. Forty male albino rats were assigned to 4 groups of equal size: (1) normal control (NC) group, (2) Metf group: daily treatment with Metf (200 mg/kg, i.p.) for 2 weeks, (3) PTZ group: treatment with PTZ (50 mg/kg, i.p.) every other day for 2 weeks, and (4) Metf + PTZ group: daily treatment with PTZ and metformin (200 mg/kg, i.p.) for 2 weeks. Administration of PTZ caused a significant increase in seizure score and duration, induced a state of oxidative stress (high malondialdehyde, low reduced glutathione and catalase activity), and led to the upregulation of β-catenin, caspase-3, and its cleavage products, Hsp70 and α-synuclein, in hippocampal regions as well as a significant reduction in seizure latency. While Metf treatment significantly ameliorated PTZ-induced seizures, attenuated oxidative stress, and upregulated α-synuclein and β-catenin expression, it also inhibited caspase-3 activation and the release of the cleavage product and caused more upregulation in Hsp70 expression in hippocampal regions (p < 0.05). In conclusion, the antiepileptic and neuroprotective effects of Metf in PTZ-induced epilepsy might be due to the inhibition of apoptosis, attenuation of oxidative stress and α-synuclein expression, and upregulation of Hsp70.
Collapse
Affiliation(s)
- Abdelaziz M Hussein
- a Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Eldosoky
- a Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Shafey
- b Department of Human Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Mesery
- c Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Amr N Ali
- d Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Khaled M Abbas
- d Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Osama A Abulseoud
- e Chemistry and Drug Metabolism, IRP, National Institute on Drug Abuse, National Institutes of Health, Biomedical Research Center, Baltimore, MD, USA
| |
Collapse
|
10
|
Hussein AM, Adel M, El-Mesery M, Abbas KM, Ali AN, Abulseoud OA. l-Carnitine Modulates Epileptic Seizures in Pentylenetetrazole-Kindled Rats via Suppression of Apoptosis and Autophagy and Upregulation of Hsp70. Brain Sci 2018; 8:45. [PMID: 29538301 PMCID: PMC5870363 DOI: 10.3390/brainsci8030045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/03/2018] [Accepted: 03/09/2018] [Indexed: 12/30/2022] Open
Abstract
l-Carnitine is a unique nutritional supplement for athletes that has been recently studied as a potential treatment for certain neuropsychiatric disorders. However, its efficacy in seizure control has not been investigated. Sprague Dawley rats were randomly assigned to receive either saline (Sal) (negative control) or pentylenetetrazole (PTZ) 40 mg/kg i.p. × 3 times/week × 3 weeks. The PTZ group was further subdivided into two groups, the first received oral l-carnitine (l-Car) (100 mg/kg/day × 4 weeks) (PTZ + l-Car), while the second group received saline (PTZ + Sal). Daily identification and quantification of seizure scores, time to the first seizure and the duration of seizures were performed in each animal. Molecular oxidative markers were examined in the animal brains. l-Car treatment was associated with marked reduction in seizure score (p = 0.0002) that was indicated as early as Day 2 of treatment and continued throughout treatment duration. Furthermore, l-Car significantly prolonged the time to the first seizure (p < 0.0001) and shortened seizure duration (p = 0.028). In addition, l-Car administration for four weeks attenuated PTZ-induced increase in the level of oxidative stress marker malondialdehyde (MDA) (p < 0.0001) and reduced the activity of catalase enzyme (p = 0.0006) and increased antioxidant GSH activity (p < 0.0001). Moreover, l-Car significantly reduced PTZ-induced elevation in protein expression of caspase-3 (p < 0.0001) and β-catenin (p < 0.0001). Overall, our results suggest a potential therapeutic role of l-Car in seizure control and call for testing these preclinical results in a proof of concept pilot clinical study.
Collapse
Affiliation(s)
- Abdelaziz M Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Mohamed Adel
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Khaled M Abbas
- Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Amr N Ali
- Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Osama A Abulseoud
- Neuroimaging Research Branch, IRP, National Institute on Drug Abuse, National Institutes of Health, Biomedical Research Center, 251 Bayview Blvd, Baltimore, MD 21224, USA.
| |
Collapse
|
11
|
Biedermann SV, Auer MK, Bindila L, Ende G, Lutz B, Weber-Fahr W, Gass P, Fuss J. Restricted vs. unrestricted wheel running in mice: Effects on brain, behavior and endocannabinoids. Horm Behav 2016; 86:45-54. [PMID: 27664019 DOI: 10.1016/j.yhbeh.2016.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 09/16/2016] [Accepted: 09/18/2016] [Indexed: 11/26/2022]
Abstract
Beneficial effects of voluntary wheel running on hippocampal neurogenesis, morphology and hippocampal-dependent behavior have widely been studied in rodents, but also serious side effects and similarities to stereotypy have been reported. Some mouse strains run excessively when equipped with running wheels, complicating the comparability to human exercise regimes. Here, we investigated how exercise restriction to 6h/day affects hippocampal morphology and metabolism, stereotypic and basal behaviors, as well as the endocannabinoid system in wheel running C57BL/6 mice; the strain most commonly used for behavioral analyses and psychiatric disease models. Restricted and unrestricted wheel running had similar effects on immature hippocampal neuron numbers, thermoregulatory nest building and basal home-cage behaviors. Surprisingly, hippocampal gray matter volume, assessed with magnetic resonance (MR) imaging at 9.4 Tesla, was only increased in unrestricted but not in restricted runners. Moreover, unrestricted runners showed less stereotypic behavior than restricted runners did. However, after blockage of running wheels for 24h stereotypic behavior also increased in unrestricted runners, arguing against a long-term effect of wheel running on stereotypic behavior. Stereotypic behaviors correlated with frontal glutamate and glucose levels assessed by 1H-MR spectroscopy. While acute running increased plasma levels of the endocannabinoid anandamide in former studies in mice and humans, we found an inverse correlation of anandamide with the daily running distance after long-term running. In conclusion, although there are some diverging effects of restricted and unrestricted running on brain and behavior, restricted running does not per se seem to be a better animal model for aerobic exercise in mice.
Collapse
Affiliation(s)
- Sarah V Biedermann
- Department of Neuroimaging, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany; Department of Psychiatry and Psychotherapy, Center of Psychosocial Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Matthias K Auer
- Department of Clinical Neuroendocrinology, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Laura Bindila
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55099 Mainz, Germany
| | - Gabriele Ende
- Department of Neuroimaging, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55099 Mainz, Germany
| | - Wolfgang Weber-Fahr
- Department of Neuroimaging, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany; Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Peter Gass
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany
| | - Johannes Fuss
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, University Medicine Mannheim, University of Heidelberg, 68159 Mannheim, Germany; Institute for Sex Research and Forensic Psychiatry, Center of Psychosocial Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
12
|
Ebrahimzadeh Bideskan AR, Lale Ataei M, Mansouri S, Hosseini M. The effects of tamoxifen and soy on dark neuron production in hippocampal formation after pentylenetetrazole-induced repeated seizures in rats. PATHOPHYSIOLOGY 2015; 22:125-135. [DOI: 10.1016/j.pathophys.2015.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 04/10/2015] [Accepted: 04/30/2015] [Indexed: 11/25/2022] Open
|
13
|
Gammazza AM, Colangeli R, Orban G, Pierucci M, Di Gennaro G, Bello ML, D'Aniello A, Bucchieri F, Pomara C, Valentino M, Muscat R, Benigno A, Zummo G, de Macario EC, Cappello F, Di Giovanni G, Macario AJL. Hsp60 response in experimental and human temporal lobe epilepsy. Sci Rep 2015; 5:9434. [PMID: 25801186 PMCID: PMC4371150 DOI: 10.1038/srep09434] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/20/2015] [Indexed: 01/01/2023] Open
Abstract
The mitochondrial chaperonin Hsp60 is a ubiquitous molecule with multiple roles, constitutively expressed and inducible by oxidative stress. In the brain, Hsp60 is widely distributed and has been implicated in neurological disorders, including epilepsy. A role for mitochondria and oxidative stress has been proposed in epileptogenesis of temporal lobe epilepsy (TLE). Here, we investigated the involvement of Hsp60 in TLE using animal and human samples. Hsp60 immunoreactivity in the hippocampus, measured by Western blotting and immunohistochemistry, was increased in a rat model of TLE. Hsp60 was also increased in the hippocampal dentate gyrus neurons somata and neuropil and hippocampus proper (CA3, CA1) of the epileptic rats. We also determined the circulating levels of Hsp60 in epileptic animals and TLE patients using ELISA. The epileptic rats showed circulating levels of Hsp60 higher than controls. Likewise, plasma post-seizure Hsp60 levels in patients were higher than before the seizure and those of controls. These results demonstrate that Hsp60 is increased in both animals and patients with TLE in affected tissues, and in plasma in response to epileptic seizures, and point to it as biomarker of hippocampal stress potentially useful for diagnosis and patient management.
Collapse
Affiliation(s)
- Antonella Marino Gammazza
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Roberto Colangeli
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Gergely Orban
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Massimo Pierucci
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | | | - Margherita Lo Bello
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | | | - Fabio Bucchieri
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
| | - Cristoforo Pomara
- Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Department of Forensic Pathology, University of Foggia, Foggia, Italy
| | - Mario Valentino
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Richard Muscat
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Arcangelo Benigno
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Giovanni Zummo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore; and IMET, Columbus Center, Baltimore, MD, USA
| | - Francesco Cappello
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Institute “Paolo Sotgiu” for Research in Quantitative and Quantum Psychiatry and Cardiology, University of Human Sciences and Technology (LUDES), Lugano, Switzerland
| | - Giuseppe Di Giovanni
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Neuroscience Division, School of Bioscience, Cardiff University, Cardiff, UK
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology, Palermo, Italy
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore; and IMET, Columbus Center, Baltimore, MD, USA
| |
Collapse
|
14
|
Kandratavicius L, Hallak JE, Carlotti CG, Assirati JA, Leite JP. Hippocampal expression of heat shock proteins in mesial temporal lobe epilepsy with psychiatric comorbidities and their relation to seizure outcome. Epilepsia 2014; 55:1834-43. [DOI: 10.1111/epi.12787] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Ludmyla Kandratavicius
- Department of Neurosciences and Behavior; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); University of Sao Paulo (USP); Ribeirao Preto Brazil
| | - Jaime E. Hallak
- Department of Neurosciences and Behavior; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); University of Sao Paulo (USP); Ribeirao Preto Brazil
- National Institute of Science and Technology in Translational Medicine (INCT-TM - CNPq); Ribeirao Preto Brazil
| | - Carlos G. Carlotti
- Department of Surgery; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
| | - Joao A. Assirati
- Department of Surgery; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
| | - Joao P. Leite
- Department of Neurosciences and Behavior; Ribeirao Preto School of Medicine; University of Sao Paulo (USP); Ribeirao Preto Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); University of Sao Paulo (USP); Ribeirao Preto Brazil
| |
Collapse
|
15
|
Otabe H, Nibuya M, Shimazaki K, Toda H, Suzuki G, Nomura S, Shimizu K. Electroconvulsive seizures enhance autophagy signaling in rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry 2014; 50:37-43. [PMID: 24316174 DOI: 10.1016/j.pnpbp.2013.11.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 11/20/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022]
Abstract
The putative antidepressive mechanisms of a series of electroconvulsive seizures (ECS) are the following: 1) downregulation of monoaminergic receptor expression in several brain regions, 2) upregulation of the expression of brain-derived neurotrophic factor (BDNF), and 3) increased neurogenesis in the hippocampus. In this study, we used Western blot techniques to present another mechanism in which ECS enhances the autophagy signaling that is involved in the machinery related to synaptic and neural plasticity. Antibodies for conjugated Atg5-Atg12 (58kD) and cleaved light chain protein 3-II (LC3-II; 14 kD) were used to detect autophagy signals. An antibody for cleaved caspase-3 (17 kD) was used to detect alterations in apoptotic signals. Mature BDNF (14kD) expression in the hippocampus was evaluated in order to qualify the effectiveness of the ECS or stress-loading treatment. While significantly increased autophagy signals and no increases in apoptotic signals were detected in the ECS-treated rat hippocampus, the reverse (increased apoptotic signals and no altered autophagy signals) was observed in stressed rat hippocampus. No neuronal cell loss but new mossy fiber sprouting has been reported to accompany multiple ECS treatments, and recent studies have revealed that autophagy processes regulate the number of specific neurotransmitter receptors and the plasticity of synaptic components. The present study illustrated the neuroplastic and neurotrophic profiles of ECS and the neurotoxic impact of severe stress loading on hippocampal regions. This is the first report to demonstrate increased autophagy signals in ECS-treated rat hippocampus and no alterations in autophagy signals in stress-loaded rat hippocampus.
Collapse
Affiliation(s)
- Hiroyuki Otabe
- Department of Psychiatry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Masashi Nibuya
- Department of Psychiatry, National Defense Medical College, Tokorozawa, Saitama, Japan.
| | - Kuniko Shimazaki
- Department of Neurosurgery, Jichi Medical School, Shimotsuke, Tochigi, Japan
| | - Hiroyuki Toda
- Department of Psychiatry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Go Suzuki
- Division of Behavioral Sciences, National Defense Medical College, Tokorozawa, Saitama, Japan; Military Medicine Research Unit, Test and Evaluation Command, Japan Ground Self Defense Force, Setagaya, Tokyo, Japan
| | - Soichiro Nomura
- Department of Psychiatry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Kunio Shimizu
- Division of Behavioral Sciences, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
16
|
Lima-Ojeda JM, Vogt MA, Pfeiffer N, Dormann C, Köhr G, Sprengel R, Gass P, Inta D. Pharmacological blockade of GluN2B-containing NMDA receptors induces antidepressant-like effects lacking psychotomimetic action and neurotoxicity in the perinatal and adult rodent brain. Prog Neuropsychopharmacol Biol Psychiatry 2013; 45:28-33. [PMID: 23643674 DOI: 10.1016/j.pnpbp.2013.04.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 04/22/2013] [Accepted: 04/23/2013] [Indexed: 12/01/2022]
Abstract
NMDA receptor (NMDAR) antagonists like ketamine and MK-801 possess remarkable antidepressant effects with fast onset. However, they over-stimulate the retrosplenial cortex, evoking psychosis-like effects and neuronal injury, revealed by de novo induction of the heat shock protein 70 (Hsp70). Moreover, early in the development MK-801 triggers widespread cortical apoptosis, inducing extensive caspase-3 expression. Altogether these data raise strong concerns on the clinical applicability of NMDAR antagonist therapies. Therefore, the development of novel therapeutics targeting more specifically NMDAR to avoid psychotomimetic effects is necessary. Here we investigated a GluN2B (NR2B) antagonist in behavioral and neurotoxicity paradigms in rats to assess its potential as possible alternative to unspecific NMDA receptor antagonists. We found that treatment with the GluN2B specific antagonist Ro 25-6981 evoked robust antidepressant-like effects. Moreover, Ro 25-6981 did not cause hyperactivity as displayed after treatment with unspecific NMDAR antagonists, a correlate of psychosis-like effects in rodents. Additionally, Ro 25-6981, unlike MK-801, did not induce caspase-3 and HSP70 expression, markers of neurotoxicity in the perinatal and adult brain, respectively. Moreover, unexpectedly, in the adult retrosplenial cortex Ro 25-6981 pretreatment significantly reduced MK-801-triggered neurotoxicity. Our results suggest that GluN2B antagonists may represent valuable alternatives to unspecific NMDAR antagonists with robust antidepressant efficacy and a more favorable side-effect profile.
Collapse
Affiliation(s)
- Juan M Lima-Ojeda
- RG Animal Models in Psychiatry, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Postweaning social isolation exacerbates neurotoxic effects of the NMDA receptor antagonist MK-801 in rats. J Neural Transm (Vienna) 2013; 120:1605-9. [DOI: 10.1007/s00702-013-1049-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 05/23/2013] [Indexed: 01/23/2023]
|
18
|
The differential effects of acute vs. chronic stress and their combination on hippocampal parvalbumin and inducible heat shock protein 70 expression. Neuroscience 2013; 236:47-54. [DOI: 10.1016/j.neuroscience.2013.01.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 01/09/2013] [Accepted: 01/10/2013] [Indexed: 11/20/2022]
|
19
|
Yang XS, He GL, Hao YT, Xiao Y, Chen CH, Zhang GB, Yu ZP. Exposure to 2.45GHz electromagnetic fields elicits an HSP-related stress response in rat hippocampus. Brain Res Bull 2012; 88:371-8. [DOI: 10.1016/j.brainresbull.2012.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 03/08/2012] [Accepted: 04/02/2012] [Indexed: 11/28/2022]
|
20
|
Rejdak K, Kuhle J, Rüegg S, Lindberg RLP, Petzold A, Sulejczak D, Papuc E, Rejdak R, Stelmasiak Z, Grieb P. Neurofilament heavy chain and heat shock protein 70 as markers of seizure-related brain injury. Epilepsia 2012; 53:922-7. [PMID: 22509781 DOI: 10.1111/j.1528-1167.2012.03459.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE Status epilepticus (SE) has deleterious effects on brain tissue, but whether brief recurrent seizures may also damage neurons represents a matter of controversy. Therefore, it remains a central area of epilepsy research to identify individuals at risk where disease progression can be potentially prevented. Biomarkers may serve as tools for such identification. Thus the present study aimed at analyzing the levels of heat shock protein 70 (HSP-70, also designated as HSPA1A) and neurofilament heavy chain protein (NfH(SMI35) ) in cerebrospinal fluid (CSF) of patients with seizures of different severity. METHODS Forty-one patients were included, of whom 20 patients had a single generalized tonic-clonic seizure (GTCS) episode (SS), 11 had repetitive GTCS (RS), and 10 experienced convulsive SE. The control group consisted of 18 subjects. HSP-70 levels were measured using a conventional enzyme-linked immunosorbent assay (ELISA), whereas the NfH(SMI35) protein levels were detected by an electrochemiluminescence (ECL) immunoassay. KEY FINDINGS Patients with SE (p < 0.001) and RS (p < 0.05) had significantly higher NfH(SMI35) levels than controls, and SE was associated with increased concentrations when compared with SS (p < 0.001). NfH(SMI35) levels in SS did not differ from controls. Patients with SE had significantly raised HSP-70 levels compared to RS (p < 0.05), SS (p < 0.05), and controls (p < 0.001). SS and RS did not differ from each or from controls. Levels of NfH(SMI35) and HSP-70 showed a significant correlation (r = 0.34; p = 0.007) in the group of all study subjects, which was not apparent when controls and patients with seizures were considered separately. The correlation between NfH(SMI35) and HSP-70 tended to be inverse in patients with SE, but it did not reach statistical significance (r = -0.3; p > 0.05). SIGNIFICANCE Studying biochemical markers as additional quantitative tools for the measurement of neuronal damage (especially subclinical), complementary to available techniques of imaging, and clinical assessment might prove useful for identifying patients at risk of accumulating neuronal injury resulting from uncontrolled seizures. NfH(SMI35) and HSP-70 are of potential value as sensitive and specific biomarkers of seizure-related pathologic events. Future longitudinal studies are needed to monitor such patients by correlating biochemical, neuroimaging, and clinical methods of assessment.
Collapse
Affiliation(s)
- Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Jaczewskiego 8, Lublin, Poland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Karlócai MR, Tóth K, Watanabe M, Ledent C, Juhász G, Freund TF, Maglóczky Z. Redistribution of CB1 cannabinoid receptors in the acute and chronic phases of pilocarpine-induced epilepsy. PLoS One 2011; 6:e27196. [PMID: 22076136 PMCID: PMC3208595 DOI: 10.1371/journal.pone.0027196] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 10/12/2011] [Indexed: 01/29/2023] Open
Abstract
The endocannabinoid system plays a central role in retrograde synaptic communication and may control the spread of activity in an epileptic network. Using the pilocarpine model of temporal lobe epilepsy we examined the expression pattern of the Type 1 cannabinoid receptor (CB1-R) in the hippocampi of CD1 mice at survival times of 2 hours, 1 day, 3 days and 2 months (acute, latent and chronic phases). Based on the behavioral signs of the acute seizures, animals were classified as "weakly" or "strongly" epileptic using the modified Racine scale. Mice of the weak group had mild seizures, whereas seizures in the strong group were frequent with intense motor symptoms and the majority of these animals developed sclerosis in the chronic phase. In control samples the most intense staining of CB1-R-positive fibers was found in the molecular layer of the dentate gyrus and in str. pyramidale of the cornu Ammonis. In weak animals no significant changes were seen at any survival time compared to controls. In strong animals, however, in the acute phase, a massive reduction in CB1-R-stained terminals occurred in the hippocampus. In the latent phase CB1-R immunoreactivity gradually recovered. In the chronic phase, CB1-immunostaining in sclerotic samples was stronger throughout the hippocampus. Quantitative electron microscopic analysis showed an increase in the number of CB1-R-positive terminals in the dentate gyrus. Moreover, the number of immunogold particles significantly increased in GABAergic terminals. Our results suggest a proconvulsive downregulation of CB1 receptors in the acute phase most probably due to receptor internalization, followed by compensatory upregulation and sprouting in the chronic phase of epilepsy. In conclusion, the changes in CB1 receptor expression pattern revealed in this study are associated with the severity of hippocampal injury initiated by acute seizures that ultimately leads to sclerosis in the vulnerable regions in the chronic phase.
Collapse
Affiliation(s)
- Mária R Karlócai
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Studies of epilepsy have mainly focused on the membrane proteins that control neuronal excitability. Recently, attention has been shifting to intracellular proteins and their interactions, signaling cascades and feedback regulation as they relate to epilepsy. The mTOR (mammalian target of rapamycin) signal transduction pathway, especially, has been suggested to play an important role in this regard. These pathways are involved in major physiological processes as well as in numerous pathological conditions. Here, involvement of the mTOR pathway in epilepsy will be reviewed by presenting; an overview of the pathway, a brief description of key signaling molecules, a summary of independent reports and possible implications of abnormalities of those molecules in epilepsy, a discussion of the lack of experimental data, and questions raised for the understanding its epileptogenic mechanism.
Collapse
Affiliation(s)
- Chang Hoon Cho
- Epilepsy Research Laboratory Department of Pediatrics Children's Hospital of Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
23
|
Kharlamov EA, Lepsveridze E, Meparishvili M, Solomonia RO, Lu B, Miller ER, Kelly KM, Mtchedlishvili Z. Alterations of GABA(A) and glutamate receptor subunits and heat shock protein in rat hippocampus following traumatic brain injury and in posttraumatic epilepsy. Epilepsy Res 2011; 95:20-34. [PMID: 21439793 DOI: 10.1016/j.eplepsyres.2011.02.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/20/2010] [Accepted: 02/18/2011] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) can result in the development of posttraumatic epilepsy (PTE). Recently, we reported differential alterations in tonic and phasic GABA(A) receptor (GABA(A)R) currents in hippocampal dentate granule cells 90 days after controlled cortical impact (CCI) (Mtchedlishvili et al., 2010). In the present study, we investigated long-term changes in the protein expression of GABA(A)R α1, α4, γ2, and δ subunits, NMDA (NR2B) and AMPA (GluR1) receptor subunits, and heat shock proteins (HSP70 and HSP90) in the hippocampus of Sprague-Dawley rats evaluated by Western blotting in controls, CCI-injured animals without PTE (CCI group), and CCI-injured animals with PTE (PTE group). No differences were found among all three groups for α1 and α4 subunits. Significant reduction of γ2 protein was observed in the PTE group compared to control. CCI caused a 194% and 127% increase of δ protein in the CCI group compared to control (p<0.0001), and PTE (p<0.0001) groups, respectively. NR2B protein was increased in CCI and PTE groups compared to control (p=0.0001, and p=0.011, respectively). GluR1 protein was significantly decreased in CCI and PTE groups compared to control (p=0.003, and p=0.001, respectively), and in the PTE group compared to the CCI group (p=0.036). HSP70 was increased in CCI and PTE groups compared to control (p=0.014, and p=0.005, respectively); no changes were found in HSP90 expression. These results provide for the first time evidence of long-term alterations of GABA(A) and glutamate receptor subunits and a HSP following CCI.
Collapse
Affiliation(s)
- Elena A Kharlamov
- Center for Neuroscience Research, Allegheny-Singer Research Institute, Allegheny General Hospital, 320 East North Avenue, Pittsburgh, PA 15212-4772, United States.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Hsp70 and its molecular role in nervous system diseases. Biochem Res Int 2011; 2011:618127. [PMID: 21403864 PMCID: PMC3049350 DOI: 10.1155/2011/618127] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 10/19/2010] [Accepted: 01/05/2011] [Indexed: 02/07/2023] Open
Abstract
Heat shock proteins (HSPs) are induced in response to many injuries including stroke, neurodegenerative disease, epilepsy, and trauma. The overexpression of one HSP in particular, Hsp70, serves a protective role in several different models of nervous system injury, but has also been linked to a deleterious role in some diseases. Hsp70 functions as a chaperone and protects neurons from protein aggregation and toxicity (Parkinson disease, Alzheimer disease, polyglutamine diseases, and amyotrophic lateral sclerosis), protects cells from apoptosis (Parkinson disease), is a stress marker (temporal lobe epilepsy), protects cells from inflammation (cerebral ischemic injury), has an adjuvant role in antigen presentation and is involved in the immune response in autoimmune disease (multiple sclerosis). The worldwide incidence of neurodegenerative diseases is high. As neurodegenerative diseases disproportionately affect older individuals, disease-related morbidity has increased along with the general increase in longevity. An understanding of the underlying mechanisms that lead to neurodegeneration is key to identifying methods of prevention and treatment. Investigators have observed protective effects of HSPs induced by preconditioning, overexpression, or drugs in a variety of models of brain disease. Experimental data suggest that manipulation of the cellular stress response may offer strategies to protect the brain during progression of neurodegenerative disease.
Collapse
|
25
|
Ryufuku M, Toyoshima Y, Kitaura H, Zheng Y, Fu YJ, Miyahara H, Murakami H, Masuda H, Kameyama S, Takahashi H, Kakita A. Hypertrophy of hippocampal end folium neurons in patients with mesial temporal lobe epilepsy. Neuropathology 2011; 31:476-85. [DOI: 10.1111/j.1440-1789.2010.01191.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
26
|
Dag E, Aydin S, Ozkan Y, Erman F, Dagli AF, Gurger M. Alteration in chromogranin A, obestatin and total ghrelin levels of saliva and serum in epilepsy cases. Peptides 2010; 31:932-7. [PMID: 20172008 DOI: 10.1016/j.peptides.2010.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 02/11/2010] [Accepted: 02/11/2010] [Indexed: 11/23/2022]
Abstract
This study was designed to measure the levels of chromogranin A (CgA), ghrelin and obestatin in serum and saliva (including CgA expression in healthy tissue) in epileptic patients to determine any significant differences between these patients and healthy controls. Samples were obtained from a total of 91 subjects: 10 newly-diagnosed primary generalized epilepsy (PGE) patients who had started treatment with valproic acid and phenytoin for seizure control; 18 PGE patients who were previously and currently receiving treatment with valproic acid and phenytoin for seizure control; 37 patients with partial epilepsy (PE) (simple, n=17 or complex, n=20) who had been and were still being treated with carbazebime for seizures; and 26 healthy controls. CgA immunoreactivity in healthy salivary gland was analyzed by immunohistochemistry and ELISA. The levels of CgA, total ghrelin and obestatin in serum and saliva were measured by ELISA. The results revealed that normal salivary gland produces its own CgA. Before treatment, CgA levels in saliva and serum were significantly greater in patients newly-diagnosed with PGE than controls. Ghrelin and CgA concentrations were also greater in PGE patients previously or currently treated with drugs, and in patients with simple or complex partial epilepsy (PE) previously or currently treated with drugs, than in healthy normal controls. In conclusion, salivary concentrations of CgA, ghrelin and obestatin were similar to their serum levels, so saliva might be a desirable alternative to serum for measuring these hormones because it is easy and painless to collect.
Collapse
Affiliation(s)
- Ersel Dag
- Department of Neurology, Elazig Research and Education Hospital, Elazig 23119, Turkey
| | | | | | | | | | | |
Collapse
|
27
|
Myo-inositol treatment prevents biochemical changes triggered by kainate-induced status epilepticus. Neurosci Lett 2010; 468:277-81. [DOI: 10.1016/j.neulet.2009.11.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 11/03/2009] [Accepted: 11/04/2009] [Indexed: 11/21/2022]
|
28
|
Kalesnykas G, Tuulos T, Uusitalo H, Jolkkonen J. Neurodegeneration and cellular stress in the retina and optic nerve in rat cerebral ischemia and hypoperfusion models. Neuroscience 2008; 155:937-47. [DOI: 10.1016/j.neuroscience.2008.06.038] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 06/12/2008] [Accepted: 06/12/2008] [Indexed: 11/30/2022]
|
29
|
Mori F, Tanji K, Wakabayashi K. Convulsive effects of thiophene, a heterocyclic hydrocarbon: Behavioral, electrographic and c‐Fos immunocytochemical studies. Neuropathology 2008. [DOI: 10.1111/j.1440-1789.2000.00351.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Fumiaki Mori
- Department of Neuropathology, Institute of Brain Science, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Kunikazu Tanji
- Department of Neuropathology, Institute of Brain Science, Hirosaki University School of Medicine, Hirosaki, Japan
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University School of Medicine, Hirosaki, Japan
| |
Collapse
|
30
|
Liu XY, Yang JL, Chen LJ, Zhang Y, Yang ML, Wu YY, Li FQ, Tang MH, Liang SF, Wei YQ. Comparative proteomics and correlated signaling network of rat hippocampus in the pilocarpine model of temporal lobe epilepsy. Proteomics 2008; 8:582-603. [DOI: 10.1002/pmic.200700514] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
31
|
Sumanont Y, Murakami Y, Tohda M, Vajragupta O, Watanabe H, Matsumoto K. Effects of Manganese Complexes of Curcumin and Diacetylcurcumin on Kainic Acid-Induced Neurotoxic Responses in the Rat Hippocampus. Biol Pharm Bull 2007; 30:1732-9. [PMID: 17827730 DOI: 10.1248/bpb.30.1732] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study aimed to investigate the mechanism underlying the protective effects of manganese complexes of curcumin (Cp-Mn) and diacetylcurcumin (DiAc-Cp-Mn) on kainic acid (KA)-induced excitotoxicity in the rat hippocampus. Systemic injection of KA (10 mg/kg, i.p.) caused seizures and increased the expression of neurotoxic markers, immediate early genes [c-jun, cyclooxygenase 2 (COX-2), brain-derived neurotrophic factor (BDNF), and heat shock protein 70 (hsp70)] and a delayed response gene [inducible nitric oxide synthase (iNOS)], which were measured at 6 and 72 h after KA injection, respectively, in the hippocampus. Pretreatment with Cp-Mn (50 mg/kg, i.p.) and DiAc-Cp-Mn (50 mg/kg, i.p.) but not with curcumin (50 mg/kg, i.p.) delayed the onset of KA-induced seizure without affecting the seizure score. KA injection induced c-Fos immunoreactivity in DG, CA1, and CA3 hippocampal regions, the expression of which peaked at 6 h after injection. Cp-Mn and DiAc-Cp-Mn treatment significantly decreased c-Fos expression elicited by KA. Moreover, Cp-Mn and DiAc-Cp-Mn administration suppressed the KA-induced expression of c-jun, COX-2, BDNF, and iNOS mRNA, whereas curcumin attenuated only iNOS mRNA expression. No compounds tested had an effect on KA-induced hsp70 expression. It is therefore likely that in addition to radical scavenging and SOD-like activities, the suppression of potential neuronal injury marker expression by Cp-Mn and DiAc-Cp-Mn, contributes to the neuroprotective activities of these compounds, which are superior to those of curcumin, on KA-induced excitotoxicity in the hippocampus. These results suggest the beneficial effects of Cp-Mn, and DiAc-Cp-Mn on the treatment of excitotoxicity-induced neurodegenerative diseases.
Collapse
Affiliation(s)
- Yaowared Sumanont
- Division of Medicinal Pharmacology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Xi ZQ, Sun JJ, Wang XF, Li MW, Liu XZ, Wang LY, Zhu X, Xiao F, Li JM, Gong Y, Guan LF. HSPBAP1 is found extensively in the anterior temporal neocortex of patients with intractable epilepsy. Synapse 2007; 61:741-7. [PMID: 17568411 DOI: 10.1002/syn.20417] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Heat Shock Protein BAP1 (heat shock 27-kDa-associated protein 1, HSPBAP1) inhibits the function of heat shock protein 27, which has a neuroprotective effect during experimentally induced epileptic neuropathology. In our study, fluorescence quantitative polymerase chain reaction, immunohistochemistry, immunofluorescence, western blot were used to test the levels of HSPBAP1 mRNA and protein in surgical samples of the anterior temporal neocortex of patients with intractable epilepsy (IE) and normal controls samples. HSPBAP1 mRNA was abnormally expressed in the anterior temporal neocortex of patients with IE. Moreover, HSPBAP1 was found extensively in the cytoplasm of neurons and glial cells in all epilepsy specimens. Western blot showed a clear immunoreactive band of HSPBAP1 in IE specimens whereas it was absent in control specimens. The expression of HSPBAP1 mRNA and protein in the anterior temporal neocortex from patients with IE may play a role in the development of epileptic seizures in patients with cell loss in this brain region. Additional studies will be required to elucidate the mechanism by which HSPBAP1 affects brain function in IE.
Collapse
Affiliation(s)
- Zhi-Qin Xi
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Domínguez MI, Blasco-Ibáñez JM, Crespo C, Nacher J, Marqués-Marí AI, Martínez-Guijarro FJ. Neural Overexcitation and Implication of NMDA and AMPA Receptors in a Mouse Model of Temporal Lobe Epilepsy Implying Zinc Chelation. Epilepsia 2006; 47:887-99. [PMID: 16686654 DOI: 10.1111/j.1528-1167.2006.00501.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE Zinc chelation with diethyldithiocarbamate (DEDTC) during nondamaging kainic acid administration enhances excitotoxicity to the level of cell damage. The objective of this work was to study the developing of the lesion in this model of temporal lobe epilepsy and the implications of the different types of glutamate receptors. METHODS The antagonist of the N-methyl-D-aspartate (NMDA) receptor MK-801, and the antagonist of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptor GYKI52466, were used concomitantly with intraperitoneal administration of kainic acid (15 mg/kg) followed by DEDTC (150 mg/kg) in mouse. The animals were killed at different times from 4 h to 7 days. Fos proteins were used as markers of cell overexcitation; heat-shock protein 72 (HSP72) as marker of cell stress. RESULTS Neither kainic acid nor DEDTC alone, at the doses used, led to cell loss, HSP72 expression, or permanent Fos protein induction. When combined, the hilus and cornu ammonis were damaged; principal cells in these areas coexpressed c-Fos and HSP72, with the exception of CA2; interneurons did not express HSP72 in any area. MK-801 completely abolished damage and HSP72 expression from the hippocampus. GYKI52466 blocked CA1 damage and HSP72 expression in the CA1 but not in the CA3. CONCLUSIONS Synaptic zinc increases the tolerance of hippocampus to overexcitation. All the areas that are fated to die are determined simultaneously; the damage in the CA1 is not an extension of the damage in the CA3. Damage of the CA3 is dependent on kainate and NMDA receptors, whereas the damage of the CA1 depends on AMPA and NMDA receptors.
Collapse
MESH Headings
- Animals
- Benzodiazepines/pharmacology
- Cell Death/drug effects
- Chelating Agents/metabolism
- Disease Models, Animal
- Ditiocarb/analogs & derivatives
- Ditiocarb/pharmacology
- Dizocilpine Maleate/pharmacology
- Epilepsy, Temporal Lobe/chemically induced
- Epilepsy, Temporal Lobe/metabolism
- Epilepsy, Temporal Lobe/physiopathology
- HSP72 Heat-Shock Proteins/metabolism
- Hippocampus/drug effects
- Hippocampus/pathology
- Hippocampus/physiopathology
- Kainic Acid/pharmacology
- Male
- Mice
- Neuroprotective Agents/pharmacology
- Proto-Oncogene Proteins c-fos/metabolism
- Receptors, AMPA/drug effects
- Receptors, AMPA/physiology
- Receptors, Kainic Acid/drug effects
- Receptors, Kainic Acid/physiology
- Receptors, N-Methyl-D-Aspartate/drug effects
- Receptors, N-Methyl-D-Aspartate/physiology
- Synaptic Transmission/drug effects
- Synaptic Transmission/physiology
- Zinc/metabolism
- Zinc/physiology
Collapse
Affiliation(s)
- María-Isabel Domínguez
- Universidad de Valencia, Facultad de Ciencias Biológicas, Departamento de Biología Celular, Unidad de Neurobiología, Burjasot, Spain
| | | | | | | | | | | |
Collapse
|
34
|
Ayala GX, Tapia R. LateN-methyl-d-aspartate receptor blockade rescues hippocampal neurons from excitotoxic stress and death after 4-aminopyridine-induced epilepsy. Eur J Neurosci 2005; 22:3067-76. [PMID: 16367773 DOI: 10.1111/j.1460-9568.2005.04509.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The intrahippocampal perfusion of 4-aminopyridine (4-AP) in the rat produces immediate seizures and delayed neuronal death, due to the overactivation of N-methyl-D-aspartate (NMDA) receptors by endogenous glutamate released from nerve endings. With the same time course, 4-AP also induces the expression of the cell stress marker heat shock protein 70 (HSP70) in the contralateral non-damaged hippocampus. We have used this experimental model to study the mechanisms of the delayed neuronal stress and death. The NMDA receptor antagonist (+)-5-methyl-10,11-dihydro-5H-dibenzo(a,d)cyclohepten-5,10-imine maleate (MK-801), administered intraperitoneally 30 or 60 but not 120 min after 4-AP perfusion, when animals show intense electroencephalography epileptiform activity, prevented the delayed neurodegeneration whereas the seizures continued for about 3 h as in the control animals. With an identical time window, MK-801 treatment also modified the pattern of HSP70 expression; the protein was expressed in the protected perfused hippocampus but no longer in the undamaged contralateral hippocampus. The possible role of Ca2+ in the delayed cell death and HSP70 expression was also studied by coperfusing the intracellular Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis(acetoxymethyl ester) with 4-AP. This treatment resulted in protective and HSP70 effects very similar to those of MK-801. These results suggest that the seizures are not linked to neurodegeneration and that NMDA receptors need to be continuously overactivated by endogenous glutamate for at least 60 min in order to induce delayed neuronal stress and death, which are dependent on Ca2+ entry through the NMDA receptor channel.
Collapse
Affiliation(s)
- Gabriela X Ayala
- Departamento de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, AP 70-253, 04510-México, DF, México
| | | |
Collapse
|
35
|
Mori F, Okada M, Tomiyama M, Kaneko S, Wakabayashi K. Effects of ryanodine receptor activation on neurotransmitter release and neuronal cell death following kainic acid-induced status epilepticus. Epilepsy Res 2005; 65:59-70. [PMID: 15979854 DOI: 10.1016/j.eplepsyres.2005.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2004] [Revised: 02/24/2005] [Accepted: 04/23/2005] [Indexed: 10/25/2022]
Abstract
Dynamic changes in intracellular free Ca(2+) concentration play a crucial role in various neural functions. The inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) and the ryanodine (Ry) receptor (RyR) are involved in Ca(2+)-induced Ca(2+)-release (CICR). Recent studies have shown that type 3 IP3R is highly expressed in rat hippocampal neurons after kainic acid (KA)-induced seizures and that dantrolene, a RyR antagonist, reduces KA-induced neuronal cell death. We investigated the RyR-associated effects of CICR agents on basal and K(+)-evoked releases of GABA and glutamate in rat hippocampus and the changes in expression of mRNA for RyRs in mouse brain after KA-induced seizures. The stimulatory effect of Ry on releases of GABA and glutamate was concentration-dependent in a biphasic manner. The inflection point in concentration-response curves for Ry on GABA release was lower than that for glutamate in both basal and K(+)-evoked conditions, suggesting that hyperactivation of RyR-associated CICR produces the imbalance between GABAergic and glutamatergic transmission. Following KA-induced seizures, transient up-regulation of brain-type RyR mRNA was observed in the hippocampal CA3 region and striatum, and signals for c-Fos mRNA increased transiently in the hippocampus, dentate gyrus and deeper layers of the neocortex. Thereafter, some dead neurons with single-stranded DNA (ssDNA) immunoreactive fragmented nuclei appeared in these areas. These findings suggest that intracellular Ca(2+) release via the RyR might be one of the mechanisms involved in KA-induced neuronal cell death.
Collapse
Affiliation(s)
- Fumiaki Mori
- Department of Neuropathology, Institute of Brain Science, Hirosaki University School of Medicine, Hirosaki 036-8562, Japan.
| | | | | | | | | |
Collapse
|
36
|
Filipović D, Gavrilović L, Dronjak S, Radojcić MB. Brain glucocorticoid receptor and heat shock protein 70 levels in rats exposed to acute, chronic or combined stress. Neuropsychobiology 2005; 51:107-14. [PMID: 15741752 DOI: 10.1159/000084168] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The pattern and intensity of glucocorticoid receptor (GR) and heat shock 70 protein (Hsp 70) changes in the hippocampus and brain cortex of adult Wistar rat males exposed to acute (immobilization, cold) and chronic (social isolation, crowding, daily swimming) stress or their combinations were followed by Western immunoblotting. Plasma ACTH and CORT were measured by chemiluminescent method and RIA. A significant decrease in cytosol GR and Hsp 70 was observed after acute stress, while chronic stresses led to negligible changes in both these proteins and caused a reduced responsiveness to a novel acute stress. This was valid irrespective of the type of chronic or acute stress combinations for both hippocampal and cortical GR and Hsp 70. The results support the hypothesis that chronic stress-induced deregulation of the LHPA axis may be caused, at least in part, by partial disruption of intracelullar negative feedback control in the higher centers of the brain.
Collapse
Affiliation(s)
- Dragana Filipović
- Laboratory of Molecular Biology and Endocrinology, VINCA Institute of Nuclear Sciences, Belgrade, Serbia and Montenegro
| | | | | | | |
Collapse
|
37
|
Jeon GS, Park SW, Kim DW, Seo JH, Cho J, Lim SY, Kim SD, Cho SS. Glial expression of the 90-kDa heat shock protein (HSP90) and the 94-kDa glucose-regulated protein (GRP94) following an excitotoxic lesion in the mouse hippocampus. Glia 2005; 48:250-8. [PMID: 15390117 DOI: 10.1002/glia.20075] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Heat shock proteins (HSPs) are immediately expressed in neuronal and glial cells under various stressful conditions and play a protective role through molecular chaperones. Although several studies have been focused on the expression of HSPs, little is known about HSP90s expression in glial cells under neuropathological conditions. In this study, we evaluated the expression pattern of the glial cell-related HSP90 and GRP94 proteins, following the induction of an excitotoxic lesion in the mouse brain. Adult mice received an intracerebroventricular injection of kainic acid; the brain tissue was then analyzed immunohistochemically for HSPs and double labeling using glial markers. HSPs expression was quantified by Western blot analysis. Excitotoxic damage was found to cause pyramidal cell degeneration in the CA3 region of the hippocampus. In the injured hippocampus, reactive microglia/macrophages expressed HSP90 from 12 h until 7 days postlesion (PL), showing maximal levels at day 1. In parallel, hippocampal reactive astrocytes showed the expression of GRP94 from 12 h until 7 days PL. In general, HSPs expression was transient, peaked at 1-3 days PL and reached basal levels by day 7. For the first time, our data demonstrate the injury-induced expression of HSP90 and GRP94 in glial cells, which may contribute to the mechanism of glial cell protection and adaptation in response to damage, thereby playing an important role in the evolution of the glial response and the excitotoxic lesion outcome. HSP90 may provide antioxidant protective mechanisms against microglia/macrophages, whereas GRP94 may stabilize the astroglial cytoskeleton and participate in astroglial antioxidant mechanisms.
Collapse
Affiliation(s)
- Gye Sun Jeon
- Department of Anatomy, College of Medicine, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Bidmon HJ, Görg B, Palomero-Gallagher N, Behne F, Lahl R, Pannek HW, Speckmann EJ, Zilles K. Heat Shock Protein-27 Is Upregulated in the Temporal Cortex of Patients with Epilepsy. Epilepsia 2004; 45:1549-59. [PMID: 15571513 DOI: 10.1111/j.0013-9580.2004.14904.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE Heat shock protein-27 (HSP-27) belongs to the group of small heat shock proteins that become induced in response to various pathologic conditions. HSP-27 has been shown to protect cells and subcellular structures, particularly mitochondria, and serves as a carrier for estradiol. It is a reliable marker for tissues affected by oxidative stress. Oxidative stress and related cellular defence mechanisms are currently thought to play a major role during experimentally induced epileptic neuropathology. We addressed the question whether HSP-27 becomes induced in the neocortex resected from patients with pharmacoresistant epilepsy. METHODS Human epileptic temporal neocortex was obtained during neurosurgery, and control tissue was obtained at autopsy from subjects without known neurologic diseases. The tissues were either frozen for Western blot analysis or fixed in Zamboni's fixative for the topographic detection of HSP-27 at the cellular level by means of immunohistochemistry. RESULTS HSP-27 was highly expressed in all epilepsy specimens and in the cortex of a patient who died in the final stage of multiple sclerosis (positive control), whereas only low amounts of HSP-27 were detectable in control brains. In epilepsy patients, HSP-27 was present in astrocytes and in the walls of blood vessels. The intracortical distribution patterns varied strongly among the epilepsy specimens. CONCLUSIONS These results demonstrate that HSP-27 becomes induced in response to epileptic pathology. Although the functional aspects of HSP-27 induction during human epilepsy have yet to be elucidated, it can be concluded that HSP-27 is a marker for cortical regions in which a stress response has been caused by seizures.
Collapse
Affiliation(s)
- Hans-J Bidmon
- C. & O. Vogt Institute for Brain Research, Dusseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Ayala GX, Tapia R. Expression of heat shock protein 70 induced by 4-aminopyridine through glutamate-mediated excitotoxic stress in rat hippocampus in vivo. Neuropharmacology 2003; 45:649-60. [PMID: 12941378 DOI: 10.1016/s0028-3908(03)00230-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The intrahippocampal administration of 4-aminopyridine (4-AP) induces epileptic seizures and neurodegeneration, due probably to stimulation of glutamate release from synaptic terminals. We have studied the time course of the neurodegenerative changes produced by 4-AP, perfused through microdialysis cannulas in rat hippocampus, and correlated them with the expression of the inducible heat shock protein 70 (HSP70), detected immunocytochemically. Electroencephalographic seizure activity appeared immediately after the beginning of 4-AP perfusion. The first signs of histological neuronal damage were observed in CA1 and CA3 subfields of the perfused hippocampus 3 h after treatment and progressed until reaching a maximal neuronal loss at 24 h. In 4-AP-treated rats HSP70 was expressed mainly in neurons of the contralateral hippocampus, with a time course and cellular distribution very similar to the neurodegeneration observed in the perfused hippocampus, but no neuronal damage was observed. The N-methyl-D-aspartate (NMDA) receptor antagonists MK-801 and (3-phosphonopropyl)-piperazine-2-carboxylic acid prevented the seizures, the neurodegeneration and the expression of HSP70. These data demonstrate that the 4-AP-induced release of endogenous glutamate overactivates NMDA receptors in the perfused hippocampus and that the resulting neuronal hyperexcitability propagates to the contralateral hippocampus, generating a glutamate-mediated neuronal stress sufficient to induce the expression of HSP70 but not to produce neurodegeneration. These findings provide a useful model for investigating the relationships between neuronal hyperexcitation, neurodegeneration and the role of HSP expression.
Collapse
Affiliation(s)
- Gabriela X Ayala
- Departamento de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, AP 70-253, C.P. 04510 México D.F., Mexico
| | | |
Collapse
|
40
|
Abstract
In response to many metabolic disturbances and injuries including stroke, neurodegenerative disease, epilepsy and trauma, the cell mounts a stress response with induction of a variety of proteins, most notably the 70 kD heat shock protein (Hsp70). The possibility that stress proteins might be neuroprotective was suspected because Hsp70, in particular, was induced to high levels in brain regions that were relatively resistant to injury. Hsp70 expression was also correlated with the phenomenon of induced tolerance. With the availability of transgenic animals and gene transfer, has it become increasingly clear that such heat shock proteins do indeed protect cells from injury. Several reports have now shown that selective overexpression of Hsp70 leads to protection in several different models of nervous system injury. This review will cover these studies, along with potential mechanisms by which Hsp70 might mediate cellular protection.
Collapse
Affiliation(s)
- Midori A Yenari
- Department of Neurosurgery, Stanford University, 1201 Welch Road, MSLS Building P304, Stanford, CA 94305-3487, USA
| |
Collapse
|
41
|
Thom M, Seetah S, Sisodiya S, Koepp M, Scaravilli F. Sudden and unexpected death in epilepsy (SUDEP): evidence of acute neuronal injury using HSP-70 and c-Jun immunohistochemistry. Neuropathol Appl Neurobiol 2003; 29:132-43. [PMID: 12662321 DOI: 10.1046/j.1365-2990.2003.00452.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Post-mortem and neuropathological examination in sudden and unexpected death in epilepsy (SUDEP) shows no specific lesions and the exact cause and mechanism of death in these cases remains undetermined. There is clinical evidence to support the fact that SUDEP is a seizure-mediated event, and patients with poorly controlled seizures are at higher risk. We aimed to identify any evidence of acute neuronal injury in SUDEP cases at post-mortem to support that a recent seizure had occurred. We analysed the distribution and frequency of heat shock protein (HSP)-70 and c-Jun immunopositive neurones in the hippocampus in 18 SUDEP cases and 22 control cases, both markers being nonspecific but early and reliable indicators of acute neuronal injury. Post-mortem control groups included patients with epilepsy with cause of death other than SUDEP (including status epilepticus and accidental death), and patients with sudden cardiac death without an epilepsy history. An additional surgical control group included patients with refractory epilepsy and hippocampal sclerosis who had undergone temporal lobectomy. Semiquantitative analysis of the distribution of HSP-70 staining showed significantly more SUDEP cases with positively labelled neurones in hippocampal subfields compared to epilepsy and cardiac post-mortem controls (P < 0.001) but not compared to the epilepsy surgical controls (P = 0.4). No significant difference in immunostaining patterns between groups was seen in the parahippocampal gyrus with HSP-70 or with c-Jun in either the hippocampus or parahippocampal gyrus regions. The detection of HSP-70 positive neurones in the hippocampus in SUDEP is supportive of ante-mortem neuronal injury including a recent seizure prior to death.
Collapse
Affiliation(s)
- M Thom
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, London.
| | | | | | | | | |
Collapse
|
42
|
Canudas AM, Pubill D, Sureda FX, Verdaguer E, Camps P, Muñoz-Torrero D, Jiménez A, Camins A, Pallàs M. Neuroprotective effects of (+/-)-huprine Y on in vitro and in vivo models of excitoxicity damage. Exp Neurol 2003; 180:123-30. [PMID: 12684026 DOI: 10.1016/s0014-4886(02)00029-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have investigated the neuroprotective effects of (+/-)-huprine Y on excitotoxic lesions in rat cerebellar granule cells (CGCs). (+/-)-Huprine Y prevented cell death induced by 100 microM glutamate, as well as, 10 microM MK-801, a NMDA receptor antagonist, in a significant manner. On the other hand, intracellular calcium increase induced by NMDA (200 microM), measured by fura-2 fluorescence, was prevented by (+/-)-huprine Y with an EC(50) of 12.44 microM, which evidences the modulatory action of this compound on NMDA-induced calcium currents. In vivo, we have studied (+/-)-huprine Y neuroprotective effects on striatal lesions induced by the subacute administration of the mitochondrial toxin 3-nitropropionic acid (3-NP, 30 mg/kg, ip, for 10 days). We have assessed that both the behavioral and the morphological consequences of the lesion were prevented by pretreatment with (+/-)-huprine Y (2.5 mg/kg/twice a day, ip). Striatal gliosis induced by 3-NP treatment was prevented by (+/-)-huprine Y pretreatment, as demonstrated by the attenuation of both the increase in [(3)H]PK 11195 specific binding indicative of microgliosis and the expression of hsp27 kDa, a chaperone expressed mainly in astrocytes. In conclusion, (+/-)-huprine Y attenuated excitotoxic-induced lesions, both in vitro and in vivo, and further evidence is provided for the potential use of this compound in the prevention of neurodegenerative disorders.
Collapse
Affiliation(s)
- Anna M Canudas
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kalwy SA, Akbar MT, Coffin RS, de Belleroche J, Latchman DS. Heat shock protein 27 delivered via a herpes simplex virus vector can protect neurons of the hippocampus against kainic-acid-induced cell loss. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 111:91-103. [PMID: 12654509 DOI: 10.1016/s0169-328x(02)00692-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Heat shock proteins are expressed in response to cellular stress and can protect cells from further stress and facilitate recovery. Heat shock protein 27 is of particular interest because it has been implicated in a range of protective roles including protein chaperoning, stabilising elements of the cytoskeleton and as an active inhibitor of apoptosis. In the present study, we have examined the potential of administration of exogenous HSP27 to confer protection against KA-induced neuronal cell death in vivo. We aimed to exploit the neurotropic specificity of herpes simplex virus-1 based virus vectors, which have been rendered replication-incompetent, to infect neurons of the hippocampus. The systemic administration of kainic acid, an analogue of glutamate, causes seizures resulting in neuronal damage and is an established animal model of epilepsy. Neuron loss is particularly prominent in the hippocampus and the mode of death is at least partly apoptotic in nature. We show that the overexpression of HSP27 in these neurons can significantly augment their survival following kainic acid administration. In contrast, injection of a control virus expressing beta-galactosidase does not confer protection. This is the first time that protection by exogenously expressed HSP27 has been demonstrated in an in vivo model of neuronal cell death.
Collapse
Affiliation(s)
- Stephan A Kalwy
- Institute of Child Health, University College London, 30 Guilford St., London WC1 1EH, UK
| | | | | | | | | |
Collapse
|
44
|
Akbar MT, Wells DJ, Latchman DS, de Belleroche J. Heat shock protein 27 shows a distinctive widespread spatial and temporal pattern of induction in CNS glial and neuronal cells compared to heat shock protein 70 and caspase 3 following kainate administration. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 93:148-63. [PMID: 11589992 DOI: 10.1016/s0169-328x(01)00199-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Kainate-induced status epilepticus is associated with both apoptotic and necrotic cell death and induction of heat shock proteins (HSPs) in hippocampal and cortical regions of the rodent brain. In the present study we have examined the temporal, spatial and cellular expression patterns of mRNAs for the highly inducible HSPs, HSP70 and HSP27, together with the apoptotic marker, caspase 3 (CPP32) in rat brain after systemic administration of kainate. HSP70 mRNA was transiently induced in the forebrain by kainate, principally in the CA1, CA3 and hilar cells of the hippocampal formation, in piriform cortex and discrete thalamic nuclei. Maximal expression was seen at 8 h after kainate which then declined to background levels by 7 days. Labelling was predominantly neuronal. In contrast, HSP27 mRNA expression was more widespread. Intense labelling was observed in CA1, CA3 and the hilar region at 8 h after kainate but the expression profile for HSP27 mRNA expanded considerably with intense signals seen in corpus callosum, cortex and thalamus at 24 h post kainate. Emulsion autoradiographs indicated a predominantly glial localisation for HSP27 mRNA. In the hilus, a distinct subpopulation of interneurones were found to express HSP27 mRNA. CPP32 mRNA was upregulated in CA1, CA3 and hilus of the hippocampal formation and in piriform cortex. CPP32 mRNA expression was more restricted and similar in distribution to HSP70 mRNA being localised to neurones. The present study demonstrates the unique early expression of HSP27 mRNA by glial cells and distinct populations of neurones which extends beyond those in which HSP70 and CPP32 induction occurs with subsequent cell loss.
Collapse
Affiliation(s)
- M T Akbar
- Department of Neuromuscular Diseases, Division of Neuroscience and Psychological Medicine, Imperial College of Science, Technology and Medicine, Charing Cross Hospital, Fulham Palace Road, W6 8RF, London, UK
| | | | | | | |
Collapse
|
45
|
Candelario-Jalil E, Al-Dalain SM, Castillo R, Martínez G, Fernández OS. Selective vulnerability to kainate-induced oxidative damage in different rat brain regions. J Appl Toxicol 2001; 21:403-7. [PMID: 11746182 DOI: 10.1002/jat.768] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Some markers of oxidative injury were measured in different rat brain areas (hippocampus, cerebral cortex, striatum, hypothalamus, amygdala/piriform cortex and cerebellum) after the systemic administration of an excitotoxic dose of kainic acid (KA, 9 mg kg(-1) i.p.) at two different sampling times (24 and 48 h). Kainic acid was able to lower markedly (P < 0.05) the glutathione (GSH) levels in hippocampus, cerebellum and amygdala/piriform cortex (maximal reduction at 24 h). In a similar way, lipid peroxidation, as assessed by malonaldehyde and 4-hydroxyalkenal levels, significantly increased (P < 0.05) in hippocampus, cerebellum and amygdala/piriform cortex mainly at 24 h after KA. In addition, hippocampal superoxide dismutase (SOD) activity decreased significantly (P < 0.05) with respect to basal levels by 24 h after KA application. On the other hand, brain areas such as hypothalamus, striatum and cerebral cortex seem to be less susceptible to KA excitotoxicity. According to these findings, the pattern of oxidative injury induced by systemically administered KA seems to be highly region-specific. Further, our results have shown that a lower antioxidant status (GSH and SOD) seems not to play an important role in the selective vulnerability of certain brain regions because it correlates poorly with increases in markers of oxidative damage.
Collapse
Affiliation(s)
- E Candelario-Jalil
- Department of Pharmacology, University of Havana (CIEB-IFAL), Apartado Postal 6079, Havana 10600, Cuba.
| | | | | | | | | |
Collapse
|
46
|
Orendácová J, Cízková D, Kafka J, Lukácová N, Marsala M, Sulla I, Marsala J, Katsube N. Cauda equina syndrome. Prog Neurobiol 2001; 64:613-37. [PMID: 11311464 DOI: 10.1016/s0301-0082(00)00065-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Single or double-level compression of the lumbosacral nerve roots located in the dural sac results in a polyradicular symptomatology clinically diagnosed as cauda equina syndrome. The cauda equina nerve roots provide the sensory and motor innervation of most of the lower extremities, the pelvic floor and the sphincters. Therefore, in a fully developed cauda equina syndrome, multiple signs of sensory disorders may appear. These disorders include low-back pain, saddle anesthesia, bilateral sciatica, then motor weakness of the lower extremities or chronic paraplegia and, bladder dysfunction. Multiple etiologies can cause the cauda equina syndrome. Among them, non-neoplastic compressive etiologies such as herniated lumbosacral discs and spinal stenosis and spinal neoplasms play a significant role in the development of the cauda equina syndrome. Non-compressive etiologies of the cauda equina syndrome include ischemic insults, inflammatory conditions, spinal arachnoiditis and other infectious etiologies. The use of canine, porcine and rat models mimicking the cauda equina syndrome enabled discovery of the effects of the compression on nerve root neural and vascular anatomy, the impairment of impulse propagation and the changes of the neurotransmitters in the spinal cord after compression of cauda equina. The involvement of intrinsic spinal cord neurons in the compression-induced cauda equina syndrome includes anterograde, retrograde and transneuronal degeneration in the lumbosacral segments. Prominent changes of NADPH diaphorase exhibiting, Fos-like immunoreactive and heat shock protein HSP72 were detected in the lumbosacral segments in a short-and long-lasting compression of the cauda equina in the dog. Developments in the diagnosis and treatment of patients with back pain, sciatica and with a herniated lumbar disc are mentioned, including many treatment options available.
Collapse
Affiliation(s)
- J Orendácová
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Krapfenbauer K, Berger M, Friedlein A, Lubec G, Fountoulakis M. Changes in the levels of low-abundance brain proteins induced by kainic acid. EUROPEAN JOURNAL OF BIOCHEMISTRY 2001; 268:3532-7. [PMID: 11422383 DOI: 10.1046/j.1432-1327.2001.02256.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Low-abundance gene products are of interest in proteomic studies, because they are probably involved in disease-related changes and their altered levels or modifications may carry significant biological information. Detection of low-abundance proteins of a proteome is one of the major limitations of proteomics and a scientific challenge. We investigated the changes in the levels of low-abundance rat brain cytosolic proteins after administration of kainic acid, a potent neurotoxin and excitatory amino acid. The cytosolic proteins from controls and animals treated with kainic acid were fractionated on an ion-exchange column. The fractions collected were analyzed by 2D electrophoresis, and the proteins with altered levels were identified by matrix-assisted laser desorption ionization or ion-spray MS. We found a manifold decrease in annexin VII, heat-shock cofactor HOP/p60 and SP-22 and a manifold increase in heparin-binding protein p30. The results suggest, respectively, the involvement of an apoptotic pathway, recruitment of the heat-shock protein machinery, generation of an antioxidant response, and, probably, induction of repair mechanisms. Three of the four proteins with altered levels had not been previously detected in the cytosolic fraction, and detection of the altered levels was possible only after the protein-enriching step.
Collapse
Affiliation(s)
- K Krapfenbauer
- F. Hoffmann-La Roche Ltd, Pharmaceutical Research, Genomics Technologies, University of Vienna, Austria
| | | | | | | | | |
Collapse
|
48
|
Abstract
Kainic acid (KA), a potent neurotoxin and excitatory amino acid, leads to derangements and modulation of brain proteins. No global brain protein expression pattern induced by KA-treatment has been reported yet. We therefore studied the effect of systemic KA administration on the levels of brain proteins. Rats were injected placebo or KA intraperitoneally and brain was taken after one week. The mitochondrial and cytosolic fractions of the brain proteins were analyzed by proteomics technologies and the levels of selected proteins were quantified using specific software. Heat shock protein HSP 27 was exclusively detected in brains of animals treated with KA, whereas the glucose regulated protein GRP 78 was downregulated. The levels of neurofilaments and alpha-internexin were significantly decreased and a fragment of tubulin alpha-1 chain was manifold increased in KA-brains. The mitochondrial enzymes dihydrolipoamide dehydrogenase, ATP synthase beta chain and isocitrate dehydrogenase were reduced and pyruvate kinase M1 was increased following KA treatment. We conclude that the concomitant determination of the brain proteins indicates altered regulation of heat shock proteins, neuronal death, cytoskeletal disruption, and mitochondrial derangement by systemic KA administration. This report confirms and extends previous studies on the effect of KA on the expression of brain proteins and suggests that our analytical system can serve as a model for neurotoxicological, neurobiological, and neuropathological proteome studies.
Collapse
Affiliation(s)
- K Krapfenbauer
- F. Hoffman-La Roche Ltd, Pharmaceutical Research, Genomics Technologies, Basel, Switzerland
| | | | | | | |
Collapse
|
49
|
Zhou F, Xiang Z, Peiling L, Junjie J, Zhen LX. The expression and changes of heat shock protein 70, MDA and haemorheology in rat cortex after diffuse axonal injury with secondary insults. J Clin Neurosci 2001; 8:250-2. [PMID: 11386800 DOI: 10.1054/jocn.2000.0760] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In the present study the role of heat shock protein 70 (HSP70) expression, changes of malonyldialdehyde (MDA) in rat cortex and haemorheology with time after diffuse axonal injury (DAI) only and DAI with secondary insults (SI) were studied. The rat DAI and DAI with SI model were made according to our previous work and animals were divided into a control and another five injury groups with time after injury. Immunohistochemical assay was used to detect the neuronal expression of HSP70 at 0.5h, 3h, 12h, 24h, 72h after DAI or DAI with SI. In the meantime, the high (etah ) and low whole blood viscosity (etaL ), haematocrit (HCT) and RBC aggregation index (AI = etaL/etah ) were also detected and calculated. MDA in the homogenised brain tissue was assayed by thiobarbituric acid (TBA) reaction. The results showed that HSP70 positive neurons were not detected at 30 minutes, but the number of HSP70 positive neurons begin to increase obviously at 3 hours, reach a peak at 24 hours (P< 0.01), and decrease at 72 hours (P= 0.05) after brain injury. The trend of expression of HSP70 was alike for both DAI only or DAI with SI. Meanwhile, MDA, etah, etaL, HCT and AI changes showed the same tendency. Compared with DAI only group, MDA and blood viscosity indexes in DAI with SI were significantly higher at respective time points (P< 0.01). It is concluded that HSP70 expression, MDA and haemorheology indices increased after brain injury and brain injury with SI. Free radicals and haemorheological changes play an important role in the aggravation of brain damage and HSP70 expression upregulation.
Collapse
Affiliation(s)
- F Zhou
- Department of Neurosurgery, Xijing Hospital, Xi'an, PRChina.
| | | | | | | | | |
Collapse
|
50
|
Masada T, Hua Y, Xi G, Ennis SR, Keep RF. Attenuation of ischemic brain edema and cerebrovascular injury after ischemic preconditioning in the rat. J Cereb Blood Flow Metab 2001; 21:22-33. [PMID: 11149665 DOI: 10.1097/00004647-200101000-00004] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ischemic preconditioning (IPC) induces neuroprotection to subsequent severe ischemia, but its effect on the cerebrovasculature has not been studied extensively. This study evaluated the effects of IPC on brain edema formation and endothelial cell damage that follows subsequent permanent focal cerebral ischemia in the rat. Transient (15 minute) middle cerebral artery occlusion (MCAO) was used for IPC. Three days after IPC or a sham operation, permanent MCAO was induced. Twenty-four hours after permanent MCAO, neurologic deficit, infarction volume, and water and ion content were evaluated. Six hours post-ischemia, blood-brain barrier (BBB) permeability was examined using [3H]-inulin. Water, ion contents, and BBB permeability were assessed in three zones (core, intermediate, and outer) depending on their relation to the MCA territory. Heat shock protein 70 (HSP70) was also examined as a potential marker of vascular injury. The model of IPC significantly reduced brain infarction and neurologic deficit. Compared with a sham operation, IPC also significantly attenuated brain edema formation in the intermediate (sham and IPC water contents: 5.99+/-0.65 vs. 4.99+/-0.81 g/g dry weight; P < 0.01) and outer zones (5.02+/-0.48 vs. 4.37+/-0.42 g/g dry weight; P < 0.01) of the ipsilateral hemisphere but not in the core zone. Blood-brain barrier disruption assessed by [3H]-inulin was significantly attenuated in the IPC group and the number of blood vessels that displayed HSP70 immunoreactivity was also reduced. Thus, IPC significantly attenuates ischemic brain edema formation, BBB disruption, and, as assessed by HSP70, vascular injury. Understanding the mechanisms involved in IPC may provide insight into methods for preserving cerebrovascular function during ischemia.
Collapse
Affiliation(s)
- T Masada
- Department of Surgery, University of Michigan, Ann Arbor 48109-0532, USA
| | | | | | | | | |
Collapse
|