1
|
Alonso-Alonso ML, Sampedro-Viana A, Fernandez-Bueno I, Pérez-Mato M, Correa-Paz C, Pérez-Gayol L, Romaus-Sanjurjo D, Ouro A, Blanco-Santero J, Oblitas CM, Castillo J, Hervella P, Iglesias-Rey R. Blood-brain barrier leakage in renovascular hypertensive rats: a quantitative MRI analysis. Neuroimage 2025:121283. [PMID: 40393574 DOI: 10.1016/j.neuroimage.2025.121283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 05/05/2025] [Accepted: 05/18/2025] [Indexed: 05/22/2025] Open
Abstract
Hypertension is a modifiable risk factor for cerebral small vessel disease (cSVD) which leads to blood-brain barrier (BBB) dysfunction. This study used gadolinium-enhanced MRI T1 and T2 in stroke-prone renovascular hypertensive rats (RHRSP) to quantify BBB leakage and brain lesions. Serum inflammation and endothelial disruption biomarkers were assessed. Brain aquaporin 4 (AQ4) and retinal morphology were evaluated by histology. RHRSP showed higher systolic blood pressure from the first post-surgical week (134.1 ± 16.6 vs 113.5 ± 11.4 mmHg; p=0.041). Gadolinium extravasation was increased in the whole brain of RHRSP (p<0.05), observing cerebral lesions in 50% of them. Regarding biomarkers, TNF-α was increased (13.5 ± 2.2 vs 11.1 ± 3.5 pg/ml, p=0.031) while soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) was lower (14.5 ± 2.2 vs 18.9 ± 2.5 pg/ml, p<0.001) in the acute hypertension phase. AQ4 was increased in the RHRSP brain (1.6 ± 0.7 vs 1.0 ± 0.2 NFU; p=0.001). Both retinal nuclear layers were reduced in RHRSP (p<0.05). Gadolinium-enhanced MRI allows the detection of BBB leakage during the establishment of RHRSP. BBB dysfunction is consistent with increased AQ4 and decreased sTWEAK release. Therefore, this animal model is useful for testing potential treatments at cSVD initial stages.
Collapse
Affiliation(s)
- María Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain.
| | - Ana Sampedro-Viana
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Ivan Fernandez-Bueno
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, Valladolid, Spain; Departamento de Cirugía, Oftalmología, Otorrinolaringología y Fisioterapia, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain.
| | - María Pérez-Mato
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Clara Correa-Paz
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lara Pérez-Gayol
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain; Centro de investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Ouro
- NeuroAging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain; Centro de investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Blanco-Santero
- Instituto Universitario de Oftalmobiología Aplicada (IOBA), Retina Group, Universidad de Valladolid, Valladolid, Spain
| | - Crhistian-Mario Oblitas
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain.
| |
Collapse
|
2
|
Schulte F, Reiter JT, Bauer T, Taube J, Bitzer F, Witt J, Piper R, Thanabalasingam A, von Wrede R, Racz A, Baumgartner T, Borger V, Specht‐Riemenschneider L, Vatter H, Hattingen E, Deichmann R, Helmstaedter C, Radbruch A, Friedman A, Surges R, Rüber T. Interictal blood-brain barrier dysfunction in piriform cortex of people with epilepsy. Ann Clin Transl Neurol 2024; 11:2623-2632. [PMID: 39190772 PMCID: PMC11514923 DOI: 10.1002/acn3.52176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
OBJECTIVE The piriform cortex is considered to be highly epileptogenic. Its resection during epilepsy surgery is a predictor for postoperative seizure freedom in temporal lobe epilepsy. Epilepsy is associated with a dysfunction of the blood-brain barrier. We investigated blood-brain barrier dysfunction in the piriform cortex of people with temporal lobe epilepsy using quantitative T1-relaxometry. METHODS Gadolinium-based contrast agent was administered ictally and interictally in 37 individuals before undergoing quantitative T1-relaxometry. Postictal and interictal images were co-registered, and subtraction maps were created as biomarkers for peri-ictal (∆qT1interictal-postictal) and interictal (∆qT1noncontrast-interictal) blood-brain barrier dysfunction. Values were extracted for the piriform cortex, hippocampus, amygdala, and the whole cortex. RESULTS In temporal lobe epilepsy (n = 14), ∆qT1noncontrast-interictal was significantly higher in the piriform cortex than in the whole cortex (p = 0.02). In extratemporal lobe epilepsy (n = 23), ∆qT1noncontrast-interictal was higher in the hippocampus than in the whole cortex (p = 0.05). Across all individuals (n = 37), duration of epilepsy was correlated with ∆qT1noncontrast-interictal (ß = 0.001, p < 0.001) in all regions, while the association was strongest in the piriform cortex. Impaired verbal memory was associated with ∆qT1noncontrast-interictal only in the piriform cortex (p = 0.04). ∆qT1interictal-postictal did not show differences in any region. INTERPRETATION Interictal blood-brain barrier dysfunction occurs in the piriform cortex in temporal lobe epilepsy. This dysfunction is linked to longer disease duration and worse cognitive deficits, emphasizing the central role of the piriform cortex in the epileptogenic network of temporal lobe epilepsy.
Collapse
Affiliation(s)
- Freya Schulte
- Department of NeuroradiologyUniversity Hospital BonnBonnGermany
- Department of EpileptologyUniversity Hospital BonnBonnGermany
| | - Johannes T. Reiter
- Department of NeuroradiologyUniversity Hospital BonnBonnGermany
- Department of EpileptologyUniversity Hospital BonnBonnGermany
| | - Tobias Bauer
- Department of NeuroradiologyUniversity Hospital BonnBonnGermany
- Department of EpileptologyUniversity Hospital BonnBonnGermany
| | - Julia Taube
- Department of EpileptologyUniversity Hospital BonnBonnGermany
| | - Felix Bitzer
- Department of NeuroradiologyUniversity Hospital BonnBonnGermany
- Department of EpileptologyUniversity Hospital BonnBonnGermany
| | | | - Rory Piper
- Developmental NeurosciencesUCL Great Ormond Street Institute of Child HealthLondonUK
| | | | - Randi von Wrede
- Department of EpileptologyUniversity Hospital BonnBonnGermany
| | - Attila Racz
- Department of EpileptologyUniversity Hospital BonnBonnGermany
| | | | - Valeri Borger
- Department of NeurosurgeryUniversity Hospital BonnBonnGermany
| | | | - Hartmut Vatter
- Department of NeurosurgeryUniversity Hospital BonnBonnGermany
| | - Elke Hattingen
- Department of NeuroradiologyClinics of Johann Wolfgang‐Goethe UniversityFrankfurt am MainGermany
- Brain Imaging CenterGoethe‐Universität FrankfurtFrankfurt am MainGermany
| | - Ralf Deichmann
- Brain Imaging CenterGoethe‐Universität FrankfurtFrankfurt am MainGermany
| | | | - Alexander Radbruch
- Department of NeuroradiologyUniversity Hospital BonnBonnGermany
- Center for Medical Data Usability and TranslationBonnGermany
- German Center for Neurodegenerative DiseasesBonnGermany
| | - Alon Friedman
- Department of Brain and Cognitive SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
- Department of Medical NeuroscienceDalhousie UniversityHalifaxCanada
| | - Rainer Surges
- Department of EpileptologyUniversity Hospital BonnBonnGermany
| | - Theodor Rüber
- Department of NeuroradiologyUniversity Hospital BonnBonnGermany
- Department of EpileptologyUniversity Hospital BonnBonnGermany
- Center for Medical Data Usability and TranslationBonnGermany
| |
Collapse
|
3
|
Sampedro-Viana A, Fernández-Rodicio S, Castillo J, Hervella P, Alonso-Alonso ML, Iglesias-Rey R. Assessment of Mannitol-Induced Chronic Blood-Brain Barrier Dysfunction In Vivo Using Magnetic Resonance. Int J Mol Sci 2024; 25:9792. [PMID: 39337280 PMCID: PMC11431755 DOI: 10.3390/ijms25189792] [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: 08/02/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
The blood-brain barrier (BBB) is essential for protection and plays a crucial role in chronic neurological disorders like small-vessel disease and Alzheimer's disease. Its complexity poses significant challenges for effective diagnostics and treatments, highlighting the need for novel animal models and comprehensive BBB dysfunction studies. This study investigates chronic BBB dysfunction induction using osmotic disruption via mannitol in healthy adult male Sprague Dawley rats over 12 weeks. Group 1 received 1 bolus/week (2.0 g/kg), Group 2 received 3 boluses/week (1.5 g/kg), and Group 3 received 3 boluses/week (2.5 g/kg). BBB dysfunction was assessed using gadolinium (Gd) infusion and MRI to evaluate location, severity, evolution, and persistence. MR spectroscopy (MRS) examined the brain metabolism changes due to intravenous mannitol, with T2-weighted MRI assessing brain lesions. Biomarkers of neuroinflammation were analyzed in the highest mannitol dose group. Our data show chronic BBB dysfunction primarily in the cortex, hippocampus, and striatum, but not in the corpus callosum of rats under periodic mannitol dosing in groups 1 and 2. MRS identified a distinctive metabolite signature, including changes in alanine, choline, and N-acetyl aspartate in the striatum of Group 1. No significant differences were found in the serum levels of all pro- and anti-inflammatory cytokines analyzed in the high-dose Group 3. This study underscores the feasibility and implications of using osmotic disruption to model chronic BBB dysfunction, offering insights for future neuroprotection and therapeutic strategies research.
Collapse
Affiliation(s)
- Ana Sampedro-Viana
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Sabela Fernández-Rodicio
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - María Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario, Rúa Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain
| |
Collapse
|
4
|
Meijer WC, Gorter JA. Role of blood-brain barrier dysfunction in the development of poststroke epilepsy. Epilepsia 2024; 65:2519-2536. [PMID: 39101543 DOI: 10.1111/epi.18072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 08/06/2024]
Abstract
Stroke is a major contributor to mortality and morbidity worldwide and the most common cause of epilepsy in the elderly in high income nations. In recent years, it has become increasingly evident that both ischemic and hemorrhagic strokes induce dysfunction of the blood-brain barrier (BBB), and that this impairment can contribute to epileptogenesis. Nevertheless, studies directly comparing BBB dysfunction and poststroke epilepsy (PSE) are largely absent. Therefore, this review summarizes the role of BBB dysfunction in the development of PSE in animal models and clinical studies. There are multiple mechanisms whereby stroke induces BBB dysfunction, including increased transcytosis, tight junction dysfunction, spreading depolarizations, astrocyte and pericyte loss, reactive astrocytosis, angiogenesis, matrix metalloproteinase activation, neuroinflammation, adenosine triphosphate depletion, oxidative stress, and finally cell death. The degree to which these effects occur is dependent on the severity of the ischemia, whereby cell death is a more prominent mechanism of BBB disruption in regions of critical ischemia. BBB dysfunction can contribute to epileptogenesis by increasing the risk of hemorrhagic transformation, increasing stroke size and the amount of cerebral vasogenic edema, extravasation of excitatory compounds, and increasing neuroinflammation. Furthermore, albumin extravasation after BBB dysfunction contributes to epileptogenesis primarily via increased transforming growth factor β signaling. Finally, seizures themselves induce BBB dysfunction, thereby contributing to epileptogenesis in a cyclical manner. In repairing this BBB dysfunction, pericyte migration via platelet-derived growth factor β signaling is indispensable and required for reconstruction of the BBB, whereby astrocytes also play a role. Although animal stroke models have their limitations, they provide valuable insights into the development of potential therapeutics designed to restore the BBB after stroke, with the ultimate goal of improving outcomes and minimizing the occurrence of PSE. In pursuit of this goal, rapamycin, statins, losartan, semaglutide, and metformin show promise, whereby modulation of pericyte migration could also be beneficial.
Collapse
Affiliation(s)
- Wouter C Meijer
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
5
|
Santos AB, Carona A, Ettcheto M, Camins A, Falcão A, Fortuna A, Bicker J. Krüppel-like factors: potential roles in blood-brain barrier dysfunction and epileptogenesis. Acta Pharmacol Sin 2024; 45:1765-1776. [PMID: 38684799 PMCID: PMC11335766 DOI: 10.1038/s41401-024-01285-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/07/2024] [Indexed: 05/02/2024]
Abstract
Epilepsy is a chronic and debilitating neurological disorder, known for the occurrence of spontaneous and recurrent seizures. Despite the availability of antiseizure drugs, 30% of people with epilepsy experience uncontrolled seizures and drug resistance, evidencing that new therapeutic options are required. The process of epileptogenesis involves the development and expansion of tissue capable of generating spontaneous recurrent seizures, during which numerous events take place, namely blood-brain barrier (BBB) dysfunction, and neuroinflammation. The consequent cerebrovascular dysfunction results in a lower seizure threshold, seizure recurrence, and chronic epilepsy. This suggests that improving cerebrovascular health may interrupt the pathological cycle responsible for disease development and progression. Krüppel-like factors (KLFs) are a family of zinc-finger transcription factors, encountered in brain endothelial cells, glial cells, and neurons. KLFs are known to regulate vascular function and changes in their expression are associated with neuroinflammation and human diseases, including epilepsy. Hence, KLFs have demonstrated various roles in cerebrovascular dysfunction and epileptogenesis. This review critically discusses the purpose of KLFs in epileptogenic mechanisms and BBB dysfunction, as well as the potential of their pharmacological modulation as therapeutic approach for epilepsy treatment.
Collapse
Affiliation(s)
| | - Andreia Carona
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Miren Ettcheto
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Antoni Camins
- Biomedical Research Networking Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Amílcar Falcão
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal.
- University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| |
Collapse
|
6
|
Hanael E, Baruch S, Altman RK, Chai O, Rapoport K, Peery D, Friedman A, Shamir MH. Blood-brain barrier dysfunction and decreased transcription of tight junction proteins in epileptic dogs. J Vet Intern Med 2024; 38:2237-2248. [PMID: 38842297 PMCID: PMC11256172 DOI: 10.1111/jvim.17099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/25/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Epilepsy in dogs and humans is associated with blood-brain barrier (BBB) dysfunction (BBBD), which may involve dysfunction of tight junction (TJ) proteins, matrix metalloproteases, and astrocytes. Imaging techniques to assess BBB integrity, to identify potential treatment strategies, have not yet been evaluated in veterinary medicine. HYPOTHESIS Some dogs with idiopathic epilepsy (IE) will exhibit BBBD. Identifying BBBD may improve antiepileptic treatment in the future. ANIMALS Twenty-seven dogs with IE and 10 healthy controls. METHODS Retrospective, prospective cohort study. Blood-brain barrier permeability (BBBP) scores were calculated for the whole brain and piriform lobe of all dogs by using dynamic contrast enhancement (DCE) magnetic resonance imaging (MRI) and subtraction enhancement analysis (SEA). Matrix metalloproteinase-9 (MMP9) activity in serum and cerebrospinal fluid (CSF) was measured and its expression in the piriform lobe was examined using immunofluorescent staining. Gene expression of TJ proteins and astrocytic transporters was analyzed in the piriform lobe. RESULTS The DCE-MRI analysis of the piriform lobe identified higher BBBP score in the IE group when compared with controls (34.5% vs 26.5%; P = .02). Activity and expression of MMP9 were increased in the serum, CSF, and piriform lobe of IE dogs as compared with controls. Gene expression of Kir4.1 and claudin-5 in the piriform lobe of IE dogs was significantly lower than in control dogs. CONCLUSIONS AND CLINICAL IMPORTANCE Our findings demonstrate BBBD in dogs with IE and were supported by increased MMP9 activity and downregulation of astrocytic potassium channels and some TJ proteins. Blood brain barrier dysfunction may be a novel antiepileptic therapy target.
Collapse
Affiliation(s)
- Erez Hanael
- Koret School of Veterinary Medicine, Neurology and NeurosurgeryThe Hebrew University of JerusalemRehovotIsrael
| | - Shelly Baruch
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, Neurology and NeurosurgeryThe Hebrew University of JerusalemRehovotIsrael
| | - Rotem Kalev Altman
- Koret School of Veterinary MedicineThe Hebrew University of JerusalemRehovotIsrael
| | - Orit Chai
- Koret School of Veterinary MedicineThe Hebrew University of JerusalemRehovotIsrael
| | - Kira Rapoport
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, Neurology and NeurosurgeryThe Hebrew University of JerusalemRehovotIsrael
| | - Dana Peery
- Koret School of Veterinary MedicineThe Hebrew University of JerusalemRehovotIsrael
| | | | - Merav H. Shamir
- Koret School of Veterinary Medicine, The Robert H. Smith Faculty of Agriculture, Food and Environment, Neurology and NeurosurgeryThe Hebrew University of JerusalemRehovotIsrael
| |
Collapse
|
7
|
Hobson BA, Rowland DJ, Dou Y, Saito N, Harmany ZT, Bruun DA, Harvey DJ, Chaudhari AJ, Garbow JR, Lein PJ. A longitudinal MRI and TSPO PET-based investigation of brain region-specific neuroprotection by diazepam versus midazolam following organophosphate-induced seizures. Neuropharmacology 2024; 251:109918. [PMID: 38527652 PMCID: PMC11250911 DOI: 10.1016/j.neuropharm.2024.109918] [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: 11/29/2023] [Revised: 03/01/2024] [Accepted: 03/13/2024] [Indexed: 03/27/2024]
Abstract
Acute poisoning with organophosphorus cholinesterase inhibitors (OPs), such as OP nerve agents and pesticides, can cause life threatening cholinergic crisis and status epilepticus (SE). Survivors often experience significant morbidity, including brain injury, acquired epilepsy, and cognitive deficits. Current medical countermeasures for acute OP poisoning include a benzodiazepine to mitigate seizures. Diazepam was long the benzodiazepine included in autoinjectors used to treat OP-induced seizures, but it is now being replaced in many guidelines by midazolam, which terminates seizures more quickly, particularly when administered intramuscularly. While a direct correlation between seizure duration and the extent of brain injury has been widely reported, there are limited data comparing the neuroprotective efficacy of diazepam versus midazolam following acute OP intoxication. To address this data gap, we used non-invasive imaging techniques to longitudinally quantify neuropathology in a rat model of acute intoxication with the OP diisopropylfluorophosphate (DFP) with and without post-exposure intervention with diazepam or midazolam. Magnetic resonance imaging (MRI) was used to monitor neuropathology and brain atrophy, while positron emission tomography (PET) with a radiotracer targeting translocator protein (TSPO) was utilized to assess neuroinflammation. Animals were scanned at 3, 7, 28, 65, 91, and 168 days post-DFP and imaging metrics were quantitated for the hippocampus, amygdala, piriform cortex, thalamus, cerebral cortex and lateral ventricles. In the DFP-intoxicated rat, neuroinflammation persisted for the duration of the study coincident with progressive atrophy and ongoing tissue remodeling. Benzodiazepines attenuated neuropathology in a region-dependent manner, but neither benzodiazepine was effective in attenuating long-term neuroinflammation as detected by TSPO PET. Diffusion MRI and TSPO PET metrics were highly correlated with seizure severity, and early MRI and PET metrics were positively correlated with long-term brain atrophy. Collectively, these results suggest that anti-seizure therapy alone is insufficient to prevent long-lasting neuroinflammation and tissue remodeling.
Collapse
Affiliation(s)
- Brad A Hobson
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA; Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA 95616, USA.
| | - Douglas J Rowland
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA 95616, USA.
| | - Yimeng Dou
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA.
| | - Naomi Saito
- Department of Public Health Sciences, University of California, Davis, School of Medicine, California 95616, USA.
| | - Zachary T Harmany
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA 95616, USA.
| | - Donald A Bruun
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA.
| | - Danielle J Harvey
- Department of Public Health Sciences, University of California, Davis, School of Medicine, California 95616, USA.
| | - Abhijit J Chaudhari
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA 95616, USA; Department of Radiology, University of California, Davis, School of Medicine, California 95817, USA.
| | - Joel R Garbow
- Biomedical Magnetic Resonance Center, Mallinckrodt Institute of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, 63110, USA.
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA 95616, USA.
| |
Collapse
|
8
|
Ravizza T, Scheper M, Di Sapia R, Gorter J, Aronica E, Vezzani A. mTOR and neuroinflammation in epilepsy: implications for disease progression and treatment. Nat Rev Neurosci 2024; 25:334-350. [PMID: 38531962 DOI: 10.1038/s41583-024-00805-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 03/28/2024]
Abstract
Epilepsy remains a major health concern as anti-seizure medications frequently fail, and there is currently no treatment to stop or prevent epileptogenesis, the process underlying the onset and progression of epilepsy. The identification of the pathological processes underlying epileptogenesis is instrumental to the development of drugs that may prevent the generation of seizures or control pharmaco-resistant seizures, which affect about 30% of patients. mTOR signalling and neuroinflammation have been recognized as critical pathways that are activated in brain cells in epilepsy. They represent a potential node of biological convergence in structural epilepsies with either a genetic or an acquired aetiology. Interventional studies in animal models and clinical studies give strong support to the involvement of each pathway in epilepsy. In this Review, we focus on available knowledge about the pathophysiological features of mTOR signalling and the neuroinflammatory brain response, and their interactions, in epilepsy. We discuss mitigation strategies for each pathway that display therapeutic effects in experimental and clinical epilepsy. A deeper understanding of these interconnected molecular cascades could enhance our strategies for managing epilepsy. This could pave the way for new treatments to fill the gaps in the development of preventative or disease-modifying drugs, thus overcoming the limitations of current symptomatic medications.
Collapse
Affiliation(s)
- Teresa Ravizza
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy
| | - Mirte Scheper
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rossella Di Sapia
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy
| | - Jan Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands.
| | - Annamaria Vezzani
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy.
| |
Collapse
|
9
|
Shen J, Zhang Y, Wu X. Rapamycin promotes hematoma resorption and enhances endothelial cell function by suppressing the mTOR/STAT3 signaling in chronic subdural hematoma. Exp Cell Res 2023; 433:113829. [PMID: 37879548 DOI: 10.1016/j.yexcr.2023.113829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
Chronic subdural hematoma (CSDH) remains a neurosurgical condition and a healthy burden especially in elderly patients. This study focuses on the functions of rapamycin and its related molecular mechanisms in CSDH management. A rat model of CSDH was induced, which developed significant hematoma on day 5 after operation. The rats were treated with rapamycin or atorvastatin, a drug with known effect on hematoma alleviation, or treated with rapamycin and atorvastatin in combination. The atorvastatin or rapamycin treatment reduced the hematoma development, blood-brain barrier permeability, neurological dysfunction in CSDH rats, and the combination treatment showed more pronounced effects. Human brain microvascular endothelial cells hCMEC/D3 were stimulated by hematoma samples to mimic a CSDH condition in vitro. The drug treatments elevated the cell junction-related factors and reduced the pro-inflammatory cytokines both in rat hematoma tissues and in hCMEC/D3 cells. Rapamycin suppressed the mTOR and STAT3 signaling pathways. Overexpression of mTOR or the STAT3 agonist suppressed the alleviating effects of rapamycin on CSDH. In summary, this study demonstrates that rapamycin promotes hematoma resorption and enhances endothelial cell function by suppressing the mTOR/STAT3 signaling.
Collapse
Affiliation(s)
- Jie Shen
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, PR China.
| | - Yile Zhang
- Department of Neurosurgery, Xijing Hospital, Xi'an, 710000, Shaanxi, PR China
| | - Xiaoqiang Wu
- Department of Neurosurgery, The People's Hospital of Sixian County, Suzhou, 234399, Anhui, PR China
| |
Collapse
|
10
|
Gaspard N. Magnetic Resonance Imaging in Status Epilepticus: Useful Scrying Board or Expensive Stopwatch? Epilepsy Curr 2023; 23:162-165. [PMID: 37334407 PMCID: PMC10273823 DOI: 10.1177/15357597231160608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
Association of Peri-Ictal MRI Abnormalities With Mortality, Antiseizure Medications Refractoriness, and Morbidity in Status Epilepticus Bonduelle T, Ollivier M, Trin K, Thomas B, Daubigney A, Michel V, De Montaudouin M, Marchal C, Aupy J. Neurology . 2022. doi:10.1212/WNL.0000000000201599 . Online ahead of print. Background and objectives: Status epilepticus (SE) is a life-threatening emergency requiring a prompt assessment of patient prognosis to guide management. Magnetic resonance imaging (MRI) allows the identification of peri-ictal MRI abnormalities (PMA) and provides insight into brain structural modifications induced by SE. However, little is known about the significance of PMA in SE prognosis. The aim of this study was to determine whether PMA are associated with an increased mortality in SE, and to establish the association between PMA and refractoriness to antiseizure medications, complications encountered and induced morbidity. Methods: We conducted a retrospective observational cohort study including all eligible consecutive patients over 15 years-old and hospitalized with SE at Bordeaux University Hospital (France), between January 2015 and December 2019. The primary endpoint was in-hospital mortality. Together with a dedicated neuroradiological reassessment, baseline characteristics, in-hospital death, SE characterization, drug refractoriness and following outcome in survivors were assessed by comprehensive medical review. Results: Of 307 patients included, 79 (26%) showed PMA related to SE. Demographic, functional status at baseline and median delay between SE onset and MRI exam were similar in PMA-positive and PMA-negative group. In-hospital death occurred in 15% (45/307) patients and was significantly higher in the PMA-positive group (27%, 21/79 vs 11%, 24/228; p<0.001). In multivariate analysis, the presence of PMA (odds ratio [OR] 2.86, 95% confidence interval [CI] 1.02-8.18; p=0.045), together with SE duration ([OR] 1.01, 95% CI 1.01-1.02; p=0.007), older age at SE onset ([OR] 1.05, 95% CI 1.01-1.09; p=0.013), preexisting ultimately fatal comorbidity ([OR] 4.01, 95% CI 1.56-10.6; p=0.004) and acute lesional SE etiology ([OR] 3.74, 95% CI 1.45-10.2; p=0.007) were independent predictors associated with in-hospital death. Patients with PMA had a higher risk of refractory SE (71 vs 33%, p<0.001). Among survivors, delayed onset epilepsy (40% vs 21%, p=0.009) occurred more frequently in the PMA-positive group. Discussion: PMA-positive cases had a higher mortality rate in the largest cohort so far to assess the prognosis value of PMA in SE. As a non-invasive and easily available tool, PMA represents a promising structural biomarker for developing a personalized approach to prognostication in SE patients receiving MRI. Association of Ictal Imaging Changes in Status Epilepticus and Neurological Deterioration Cornwall CD, Dahl SM, Nguyen N, Roberg LE, Monsson O, Krøigård T, Beier CP. Epilepsia . 2022;63(11):2970-2980. doi:10.1111/epi.17404 Objective: In patients with status epilepticus (SE), the clinical significance of ictal changes on magnetic resonance imaging (MRI) is insufficiently understood. We here studied whether the presence of ictal MRI changes was associated with neurological deterioration at discharge. Methods: The retrospective cohort comprised all identifiable patients treated at Odense University Hospital in the period 2008-2017. All amenable MRIs were systemically screened for ictal changes. Patient demographics, electroencephalography, seizure characteristics, treatment, and SE duration were assessed. Neurological status was estimated before and after SE. The predefined endpoint was the association of neurological deterioration and ictal MRI changes. Results: Of 261 eligible patients, 101 received at least one MRI during SE or within 7 days after cessation; 43.6% (44/101) had SE due to non- or less brain-damaging etiologies. Patients who received MRI had a longer duration of SE, less frequently had a history of epilepsy, and were more likely to have SE due to unknown causes. Basic characteristics (including electroencephalographic features defined by the Salzburg criteria) did not differ between patients with (n = 20) and without (n = 81) ictal MRI changes. Timing of MRI was important; postictal changes were rare within the first 24 h and hardly seen >5 days after cessation of SE. Ictal MRI changes were associated with a higher risk of neurological deterioration at discharge irrespective of etiology. Furthermore, they were associated with a longer duration of SE and higher long-term mortality that reached statistical significance in patients with non- or less brain-damaging etiologies. Significance: In this retrospective cohort, ictal changes on MRI were associated with a higher risk of neurological deterioration at discharge and, possibly, with a longer duration of SE and poorer survival.
Collapse
Affiliation(s)
- Nicolas Gaspard
- Professor of Neurology Hôpital Universitaire de Bruxelles - Université Libre de Bruxelles
| |
Collapse
|
11
|
Hanael E, Baruch S, Chai O, Lishitsky L, Blum T, Rapoport K, Ruggeri M, Aizenberg Z, Peery D, Meyerhoff N, Volk HA, De Decker S, Tipold A, Baumgaertner W, Friedman A, Shamir M. Quantitative analysis of magnetic resonance images for characterization of blood-brain barrier dysfunction in dogs with brain tumors. J Vet Intern Med 2023; 37:606-617. [PMID: 36847997 DOI: 10.1111/jvim.16654] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) permeability can be assessed quantitatively using advanced imaging analysis. HYPOTHESIS/OBJECTIVES Quantification and characterization of blood-brain barrier dysfunction (BBBD) patterns in dogs with brain tumors can provide useful information about tumor biology and assist in distinguishing between gliomas and meningiomas. ANIMALS Seventy-eight hospitalized dogs with brain tumors and 12 control dogs without brain tumors. METHODS In a 2-arm study, images from a prospective dynamic contrast-enhanced (DCE; n = 15) and a retrospective archived magnetic resonance imaging study (n = 63) were analyzed by DCE and subtraction enhancement analysis (SEA) to quantify BBB permeability in affected dogs relative to control dogs (n = 6 in each arm). For the SEA method, 2 ranges of postcontrast intensity differences, that is, high (HR) and low (LR), were evaluated as possible representations of 2 classes of BBB leakage. BBB score was calculated for each dog and was associated with clinical characteristics and tumor location and class. Permeability maps were generated, using the slope values (DCE) or intensity difference (SEA) of each voxel, and analyzed. RESULTS Distinctive patterns and distributions of BBBD were identified for intra- and extra-axial tumors. At a cutoff of 0.1, LR/HR BBB score ratio yielded a sensitivity of 80% and specificity of 100% in differentiating gliomas from meningiomas. CONCLUSIONS AND CLINICAL IMPORTANCE Blood-brain barrier dysfunction quantification using advanced imaging analyses has the potential to be used for assessment of brain tumor characteristics and behavior and, particularly, to help differentiating gliomas from meningiomas.
Collapse
Affiliation(s)
- Erez Hanael
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Shelly Baruch
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Orit Chai
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Liron Lishitsky
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Tal Blum
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Kira Rapoport
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Marco Ruggeri
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Zahi Aizenberg
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Dana Peery
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| | - Nina Meyerhoff
- School of Veterinary Medicine Hannover, Small Animal Medicine and Surgery, Hannover, Germany
| | - Holger Andreas Volk
- School of Veterinary Medicine Hannover, Small Animal Medicine and Surgery, Hannover, Germany
| | - Steven De Decker
- Department of Clinical Sciences, Royal Veterinary College, University of London, Hertfordshire, UK
| | - Andrea Tipold
- School of Veterinary Medicine Hannover, Small Animal Medicine and Surgery, Hannover, Germany
| | - Wolfgang Baumgaertner
- School of Veterinary Medicine Hannover, Small Animal Medicine and Surgery, Hannover, Germany
| | - Alon Friedman
- Faculty of Medicine, Department of Medical Neuroscience Halifax, Dalhousie University, Nova Scotia, Canada.,Departments of Physiology and Cell Biology, Brain, and Cognitive Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Merav Shamir
- The Koret School of Veterinary Medicine, Neurology and Neurosurgery, Hebrew University of Jerusalem, Reehovot, Israel
| |
Collapse
|
12
|
Torres CL, Scalco FB, de Oliveira MLC, Peake RWA, Garrett R. Untargeted LC-HRMS metabolomics reveals candidate biomarkers for mucopolysaccharidoses. Clin Chim Acta 2023; 541:117250. [PMID: 36764508 DOI: 10.1016/j.cca.2023.117250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND Mucopolysaccharidoses (MPSs) are inherited genetic diseases caused by an absence or deficiency of lysosomal enzymes responsible for catabolizing glycosaminoglycans (GAGs). Undiagnosed patients, or those without adequate treatment in early life, can be severely and irreversibly affected by the disease. In this study, we applied liquid chromatography-high resolution mass spectrometry (LC-HRMS)-based untargeted metabolomics to identify potential biomarkers for MPS disorders to better understand how MPS may affect the metabolome of patients. METHODS Urine samples from 37 MPS patients (types I, II, III, IV, and VI; untreated and treated with enzyme replacement therapy (ERT)) and 38 controls were analyzed by LC-HRMS. Data were processed by an untargeted metabolomics workflow and submitted to multivariate statistical analyses to reveal significant differences between the MPS and control groups. RESULTS A total of 12 increased metabolites common to all MPS types were identified. Dipeptides, amino acids and derivatives were increased in the MPS group compared to controls. N-acetylgalactosamines 4- or 6-sulfate, important constituents of GAGs, were also elevated in MPS patients, most prominently in those with MPS VI. Notably, treated patients exhibited lower levels of the aforementioned acylaminosugars than untreated patients in all MPS types. CONCLUSIONS Untargeted metabolomics has enabled the detection of metabolites that could improve our understanding of MPS physiopathology. These potential biomarkers can be utilized in screening methods to support diagnosis and ERT monitoring.
Collapse
Affiliation(s)
- Clarisse L Torres
- Metabolomics Laboratory, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernanda B Scalco
- Inborn Error of Metabolism Laboratory, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Maria Lúcia C de Oliveira
- Inborn Error of Metabolism Laboratory, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Roy W A Peake
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rafael Garrett
- Metabolomics Laboratory, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Hamed R, Eyal AD, Berman E, Eyal S. In silico screening for clinical efficacy of antiseizure medications: Not all central nervous system drugs are alike. Epilepsia 2023; 64:311-319. [PMID: 36478573 PMCID: PMC10107105 DOI: 10.1111/epi.17479] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Roaa Hamed
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amit David Eyal
- Computational Medicine Program, School of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Erez Berman
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sara Eyal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
14
|
Dusing M, LaSarge CL, White A, Jerow LG, Gross C, Danzer SC. Neurovascular Development in Pten and Tsc2 Mouse Mutants. eNeuro 2023; 10:ENEURO.0340-22.2023. [PMID: 36759189 PMCID: PMC9953070 DOI: 10.1523/eneuro.0340-22.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Hyperactivation of the mechanistic target of rapamycin (mTOR) signaling pathway is linked to more than a dozen neurologic diseases, causing a range of pathologies, including excess neuronal growth, disrupted neuronal migration, cortical dysplasia, epilepsy and autism. The mTOR pathway also regulates angiogenesis. For the present study, therefore, we queried whether loss of Pten or Tsc2, both mTOR negative regulators, alters brain vasculature in three mouse models: one with Pten loss restricted to hippocampal dentate granule cells [DGC-Pten knock-outs (KOs)], a second with widespread Pten loss from excitatory forebrain neurons (FB-Pten KOs) and a third with focal loss of Tsc2 from cortical excitatory neurons (f-Tsc2 KOs). Total hippocampal vessel length and volume per dentate gyrus were dramatically increased in DGC-Pten knock-outs. DGC-Pten knock-outs had larger dentate gyri overall, however, and when normalized to these larger structures, vessel density was preserved. In addition, tests of blood-brain barrier integrity did not reveal increased permeability. FB-Pten KOs recapitulated the findings in the more restricted DGC-Pten KOs, with increased vessel area, but preserved vessel density. FB-Pten KOs did, however, exhibit elevated levels of the angiogenic factor VegfA. In contrast to findings with Pten, focal loss of Tsc2 from cortical excitatory neurons produced a localized increase in vessel density. Together, these studies demonstrate that hypervascularization is not a consistent feature of mTOR hyperactivation models and suggest that loss of different mTOR pathway regulatory genes exert distinct effects on angiogenesis.
Collapse
Affiliation(s)
- Mary Dusing
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Candi L LaSarge
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
- Departments of Anesthesia and Pediatrics, University of Cincinnati, Cincinnati, OH 45229
- Center for Pediatric Neuroscience, Cincinnati Children's Hospital, Cincinnati, OH 45229
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45219
| | - Angela White
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Lilian G Jerow
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45219
| | - Christina Gross
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
- Departments of Anesthesia and Pediatrics, University of Cincinnati, Cincinnati, OH 45229
- Center for Pediatric Neuroscience, Cincinnati Children's Hospital, Cincinnati, OH 45229
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45219
| | - Steve C Danzer
- Department of Anesthesia, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
- Departments of Anesthesia and Pediatrics, University of Cincinnati, Cincinnati, OH 45229
- Center for Pediatric Neuroscience, Cincinnati Children's Hospital, Cincinnati, OH 45229
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45219
| |
Collapse
|
15
|
Burek M, Kaupp V, Blecharz-Lang K, Dilling C, Meybohm P. Protocadherin gamma C3: a new player in regulating vascular barrier function. Neural Regen Res 2023. [PMID: 35799511 PMCID: PMC9241426 DOI: 10.4103/1673-5374.343896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Defects in the endothelial cell barrier accompany diverse malfunctions of the central nervous system such as neurodegenerative diseases, stroke, traumatic brain injury, and systemic diseases such as sepsis, viral and bacterial infections, and cancer. Compromised endothelial sealing leads to leaking blood vessels, followed by vasogenic edema. Brain edema as the most common complication caused by stroke and traumatic brain injury is the leading cause of death. Brain microvascular endothelial cells, together with astrocytes, pericytes, microglia, and neurons form a selective barrier, the so-called blood-brain barrier, which regulates the movement of molecules inside and outside of the brain. Mechanisms that regulate blood-brain barrier permeability in health and disease are complex and not fully understood. Several newly discovered molecules that are involved in the regulation of cellular processes in brain microvascular endothelial cells have been described in the literature in recent years. One of these molecules that are highly expressed in brain microvascular endothelial cells is protocadherin gamma C3. In this review, we discuss recent evidence that protocadherin gamma C3 is a newly identified key player involved in the regulation of vascular barrier function.
Collapse
|
16
|
Zhao X, Cheng P, Xu R, Meng K, Liao S, Jia P, Zheng X, Xiao C. Insights into the development of pentylenetetrazole-induced epileptic seizures from dynamic metabolomic changes. Metab Brain Dis 2022; 37:2441-2455. [PMID: 35838870 DOI: 10.1007/s11011-022-01018-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/26/2022] [Indexed: 10/17/2022]
Abstract
Epilepsy is often considered to be a progressive neurological disease, and the nature of this progression remains unclear. Understanding the overall and common metabolic changes of epileptic seizures can provide novel clues for their control and prevention. Herein, a chronic kindling animal model was established to obtain generalized tonic-clonic seizures via the repeated injections of pentylenetetrazole (PTZ) at subconvulsive dose. Dynamic metabolomic changes in plasma and urine from PTZ-kindled rats at the different kindling phases were explored using NMR-based metabolomics, in combination with behavioral assessment, brain neurotransmitter measurement, electroencephalography and histopathology. The increased levels of glucose, lactate, glutamate, creatine and creatinine, together with the decreased levels of pyruvate, citrate and succinate, ketone bodies, asparagine, alanine, leucine, valine and isoleucine in plasma and/or urine were involved in the development and progression of seizures. These altered metabolites reflected the pathophysiological processes including the compromised energy metabolism, the disturbed amino acid metabolism, the peripheral inflammation and changes in gut microbiota functions. NMR-based metabolomics could provide brain disease information by the dynamic plasma and urinary metabolic changes during chronic epileptic seizures, yielding classification of seizure stages and profound insights into controlling epilepsy via targeting deficient energy metabolism.
Collapse
Affiliation(s)
- Xue Zhao
- The College of Life Sciences, Northwest University, 710069, Xi'an, PR China
| | - Peixuan Cheng
- The College of Life Sciences, Northwest University, 710069, Xi'an, PR China
| | - Ru Xu
- The College of Life Sciences, Northwest University, 710069, Xi'an, PR China
| | - Kaili Meng
- The College of Life Sciences, Northwest University, 710069, Xi'an, PR China
| | - Sha Liao
- The College of Life Sciences, Northwest University, 710069, Xi'an, PR China
| | - Pu Jia
- The College of Life Sciences, Northwest University, 710069, Xi'an, PR China
| | - Xiaohui Zheng
- The College of Life Sciences, Northwest University, 710069, Xi'an, PR China
| | - Chaoni Xiao
- The College of Life Sciences, Northwest University, 710069, Xi'an, PR China.
| |
Collapse
|
17
|
Broekaart DWM, Zimmer TS, Cohen ST, Tessers R, Anink JJ, de Vries HE, Gorter JA, Prades R, Aronica E, van Vliet EA. The Gelatinase Inhibitor ACT-03 Reduces Gliosis in the Rapid Kindling Rat Model of Epilepsy, and Attenuates Inflammation and Loss of Barrier Integrity In Vitro. Biomedicines 2022; 10:biomedicines10092117. [PMID: 36140216 PMCID: PMC9495904 DOI: 10.3390/biomedicines10092117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/12/2022] [Accepted: 08/20/2022] [Indexed: 11/25/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are endopeptidases responsible for the cleavage of intra- and extracellular proteins. Several brain MMPs have been implicated in neurological disorders including epilepsy. We recently showed that the novel gelatinase inhibitor ACT-03 has disease-modifying effects in models of epilepsy. Here, we studied its effects on neuroinflammation and blood–brain barrier (BBB) integrity. Using the rapid kindling rat model of epilepsy, we examined whether ACT-03 affected astro- and microgliosis in the brain using immunohistochemistry. Cellular and molecular alterations were further studied in vitro using human fetal astrocyte and brain endothelial cell (hCMEC/D3) cultures, with a focus on neuroinflammatory markers as well as on barrier permeability using an endothelial and astrocyte co-culture model. We observed less astro- and microgliosis in the brains of kindled animals treated with ACT-03 compared to control vehicle-treated animals. In vitro, ACT-03 treatment attenuated stimulation-induced mRNA expression of several pro-inflammatory factors in human fetal astrocytes and brain endothelial cells, as well as a loss of barrier integrity in endothelial and astrocyte co-cultures. Since ACT-03 has disease-modifying effects in epilepsy models, possibly via limiting gliosis, inflammation, and barrier integrity loss, it is of interest to further evaluate its effects in a clinical trial.
Collapse
Affiliation(s)
- Diede W. M. Broekaart
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Till S. Zimmer
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sophie T. Cohen
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Rianne Tessers
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jasper J. Anink
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Helga E. de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Jan A. Gorter
- Swammerdam Institute for Life Sciences Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Roger Prades
- Accure Therapeutics S.L., 08028 Barcelona, Spain
| | - Eleonora Aronica
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), 2103 SW Heemstede, The Netherlands
- Correspondence: (E.A.); (E.A.v.V.)
| | - Erwin A. van Vliet
- Amsterdam UMC, Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
- Correspondence: (E.A.); (E.A.v.V.)
| |
Collapse
|
18
|
van Vliet EA, Immonen R, Prager O, Friedman A, Bankstahl JP, Wright DK, O'Brien TJ, Potschka H, Gröhn O, Harris NG. A companion to the preclinical common data elements and case report forms for in vivo rodent neuroimaging: A report of the TASK3-WG3 Neuroimaging Working Group of the ILAE/AES Joint Translational Task Force. Epilepsia Open 2022. [PMID: 35962745 DOI: 10.1002/epi4.12643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/01/2022] [Indexed: 11/10/2022] Open
Abstract
The International League Against Epilepsy/American Epilepsy Society (ILAE/AES) Joint Translational Task Force established the TASK3 working groups to create common data elements (CDEs) for various aspects of preclinical epilepsy research studies, which could help improve the standardization of experimental designs. In this article, we discuss CDEs for neuroimaging data that are collected in rodent models of epilepsy, with a focus on adult rats and mice. We provide detailed CDE tables and case report forms (CRFs), and with this companion manuscript, we discuss the methodologies for several imaging modalities and the parameters that can be collected.
Collapse
Affiliation(s)
- Erwin A van Vliet
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam UMC Location University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Riikka Immonen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Ofer Prager
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Department of Medical Neuroscience and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- The Royal Melbourne Hospital, The University of Melbourne, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Olli Gröhn
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Neil G Harris
- Department of Neurosurgery UCLA, UCLA Brain Injury Research Center, Los Angeles, California, USA
- Intellectual and Developmental Disabilities Research Center, UCLA, Los Angeles, California, USA
| |
Collapse
|
19
|
Hanael E, Baruch S, Chai O, Nir Z, Rapoport K, Ruggeri M, Eizenberg I, Peery D, Friedman A, Shamir MH. Detection of blood‐brain barrier dysfunction using advanced imaging methods to predict seizures in dogs with meningoencephalitis of unknown origin. J Vet Intern Med 2022; 36:702-712. [PMID: 35285550 PMCID: PMC8965229 DOI: 10.1111/jvim.16396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 12/01/2022] Open
Abstract
Background The blood‐brain barrier (BBB), which separates the intravascular and neuropil compartments, characterizes the vascular bed of the brain and is essential for its proper function. Recent advances in imaging techniques have driven the development of methods for quantitative assessment of BBB permeability. Hypothesis/Objectives Permeability of the BBB can be assessed quantitatively in dogs with meningoencephalitis of unknown origin (MUO) and its status is associated with the occurrence of seizures. Animals Forty dogs with MUO and 12 dogs without MUO. Methods Retrospective, prospective cohort study. Both dynamic contrast enhancement (DCE) and subtraction enhancement analysis (SEA) methods were used to evaluate of BBB permeability in affected (DCE, n = 8; SEA, n = 32) and control dogs (DCE, n = 6; SEA, n = 6). Association between BBB dysfunction (BBBD) score and clinical characteristics was examined. In brain regions where BBBD was identified by DCE or SEA magnetic resonance imaging (MRI) analysis, immunofluorescent staining for albumin, glial fibrillary acidic protein, ionized calcium binding adaptor molecule, and phosphorylated mothers against decapentaplegic homolog 2 were performed to detect albumin extravasation, reactive astrocytes, activated microglia, and transforming growth factor beta signaling, respectively. Results Dogs with BBBD had significantly higher seizure prevalence (72% vs 19%; P = .01) when compared to MUO dogs with no BBBD. The addition of SEA to routine MRI evaluation increased the identification rate of brain pathology in dogs with MUO from 50% to 72%. Conclusions and Clinical Importance Imaging‐based assessment of BBB integrity has the potential to predict risk of seizures in dogs with MUO.
Collapse
Affiliation(s)
- Erez Hanael
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Shelly Baruch
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Orit Chai
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Zohar Nir
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Kira Rapoport
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Marco Ruggeri
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Itzhak Eizenberg
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Dana Peery
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Brain, and Cognitive Sciences, Zlotowski Center for Neuroscience Ben‐Gurion University of the Negev Beer Sheva Israel
- Department of Medical Neuroscience, Faculty of Medicine Dalhousie University Halifax NS Canada
| | - Merav H. Shamir
- Hebrew University Koret School of Veterinary Medicine‐Veterinary Teaching Hospital Rehovot Israel
| |
Collapse
|
20
|
van Vliet EA, Marchi N. Neurovascular unit dysfunction as a mechanism of seizures and epilepsy during aging. Epilepsia 2022; 63:1297-1313. [PMID: 35218208 PMCID: PMC9321014 DOI: 10.1111/epi.17210] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022]
Abstract
The term neurovascular unit (NVU) describes the structural and functional liaison between specialized brain endothelium, glial and mural cells, and neurons. Within the NVU, the blood‐brain barrier (BBB) is the microvascular structure regulating neuronal physiology and immune cross‐talk, and its properties adapt to brain aging. Here, we analyze a research framework where NVU dysfunction, caused by acute insults or disease progression in the aging brain, represents a converging mechanism underlying late‐onset seizures or epilepsy and neurological or neurodegenerative sequelae. Furthermore, seizure activity may accelerate brain aging by sustaining regional NVU dysfunction, and a cerebrovascular pathology may link seizures to comorbidities. Next, we focus on NVU diagnostic approaches that could be tailored to seizure conditions in the elderly. We also examine the impending disease‐modifying strategies based on the restoration of the NVU and, more in general, the homeostatic control of anti‐ and pro‐inflammatory players. We conclude with an outlook on current pre‐clinical knowledge gaps and clinical challenges pertinent to seizure onset and conditions in an aging population.
Collapse
Affiliation(s)
- Erwin A van Vliet
- Amsterdam UMC, University of Amsterdam, dept. of (Neuro)pathology, Amsterdam, the Netherlands.,University of Amsterdam, Swammerdam Institute for Life Sciences, Center for Neuroscience, Amsterdam, the Netherlands
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
21
|
Hanael E, Chai O, Konstanitin L, Gibeon L, Rapaport K, Ruggeri M, Friedman A, Shamir MH. Telmisartan as an add-on treatment for dogs with refractory idiopathic epilepsy: a nonrandomized, uncontrolled, open-label clinical trial. J Am Vet Med Assoc 2022; 260:735-740. [PMID: 35201995 DOI: 10.2460/javma.20.12.0683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To evaluate the effect on seizure frequency of add-on telmisartan treatment in dogs with refractory idiopathic epilepsy. ANIMALS 11 client-owned dogs with idiopathic epilepsy and ≥ 2 generalized seizures/mon that were currently being treated with ≥ 2 antiepileptic drugs. PROCEDURES Telmisartan was administered at a dosage of 0.25 to 1 mg/kg, PO, every 12 hours for 4 to 16 months. Seizure frequencies before and during telmisartan treatment were recorded. RESULTS 10 dogs completed the 4-month treatment protocol. One dog was excluded owing to a transient increase in serum creatinine concentration; no adverse effects of telmisartan were observed in the remaining 10 dogs. A reduction in seizure frequency greater than an estimated expected placebo effect of 30% was evident in 7 of the 10 dogs. Long-term (12 to 16 months) follow-up information was available for 6 dogs, of which 4 had a further reduction in seizure frequency. Differences in seizure frequency were not statistically significant. No significant difference was found in serum phenobarbital concentration throughout the treatment period in the 7 dogs that were tested. CLINICAL RELEVANCE Telmisartan has the potential to reduce seizure frequency when administered as an add-on antiepileptic drug in dogs with refractory idiopathic epilepsy. A randomized, double-blind, placebo-controlled trial is needed to determine the true efficacy of telmisartan. On the basis of our results, a sample size of 54 dogs with refractory idiopathic epilepsy would be needed.
Collapse
Affiliation(s)
- Erez Hanael
- Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Orit Chai
- Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Lilach Konstanitin
- Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | | | - Kira Rapaport
- Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Marco Ruggeri
- Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Alon Friedman
- Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Merav H Shamir
- Veterinary Teaching Hospital, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
22
|
Advances regarding Neuroinflammation Biomarkers with Noninvasive Techniques in Epilepsy. Behav Neurol 2022; 2021:7946252. [PMID: 34976232 PMCID: PMC8716206 DOI: 10.1155/2021/7946252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/23/2022] Open
Abstract
A rapidly growing body of evidence supports that neuroinflammation plays a major role in epileptogenesis and disease progression. The capacity to identify pathological neuroinflammation in individuals with epilepsy is a crucial step on the timing of anti-inflammatory intervention and patient selection, which will be challenging aspects in future clinical studies. The discovery of noninvasive biomarkers that are accessible in the blood or molecular neuroimaging would facilitate clinical translation of experimental findings into humans. These innovative and noninvasive approaches have the advantage of monitoring the dynamic changes of neuroinflammation in epilepsy. Here, we will review the available evidence for the measurement of neuroinflammation in patients with epilepsy using noninvasive techniques and critically analyze the major scientific challenges of noninvasive methods. Finally, we propose the potential for use in clinical applications.
Collapse
|
23
|
Stegmayr C, Surges R, Choi CH, Burda N, Stoffels G, Filß C, Willuweit A, Neumaier B, Heinzel A, Shah NJ, Mottaghy FM, Langen KJ. Investigation of Cerebral O-(2-[ 18F]Fluoroethyl)-L-Tyrosine Uptake in Rat Epilepsy Models. Mol Imaging Biol 2021; 22:1255-1265. [PMID: 32409931 PMCID: PMC7497431 DOI: 10.1007/s11307-020-01503-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE A recent study reported on high, longer lasting and finally reversible cerebral uptake of O-(2-[18F]fluoroethyl)-L-tyrosine ([18F]FET) induced by epileptic activity. Therefore, we examined cerebral [18F]FET uptake in two chemically induced rat epilepsy models and in patients with focal epilepsy to further investigate whether this phenomenon represents a major pitfall in brain tumor diagnostics and whether [18F]FET may be a potential marker to localize epileptic foci. PROCEDURES Five rats underwent kainic acid titration to exhibit 3 to 3.5 h of class IV-V motor seizures (status epilepticus, SE). Rats underwent 4× [18F]FET PET and 4× MRI on the following 25 days. Six rats underwent kindling with pentylenetetrazol (PTZ) 3 to 8×/week over 10 weeks, and hence, seizures increased from class I to class IV. [18F]FET PET and MRI were performed regularly on days with and without seizures. Four rats served as healthy controls. Additionally, five patients with focal epilepsy underwent [18F]FET PET within 12 days after the last documented seizure. RESULTS No abnormalities in [18F]FET PET or MRI were detected in the kindling model. The SE model showed significantly decreased [18F]FET uptake 3 days after SE in all examined brain regions, and especially in the amygdala region, which normalized within 2 weeks. Corresponding signal alterations in T2-weighted MRI were noted in the amygdala and hippocampus, which recovered 24 days post-SE. No abnormality of cerebral [18F]FET uptake was noted in the epilepsy patients. CONCLUSIONS There was no evidence for increased cerebral [18F]FET uptake after epileptic seizures neither in the rat models nor in patients. The SE model even showed decreased [18F]FET uptake throughout the brain. We conclude that epileptic seizures per se do not cause a longer lasting increased [18F]FET accumulation and are unlikely to be a major cause of pitfall for brain tumor diagnostics.
Collapse
Affiliation(s)
- Carina Stegmayr
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.
| | - Rainer Surges
- Department of Neurology, RWTH University Aachen, Aachen, Germany.,Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Chang-Hoon Choi
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Nicole Burda
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Christian Filß
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Alexander Heinzel
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany
| | - N Jon Shah
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Neurology, RWTH University Aachen, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany.,Centre of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-4; INM-5; INM-11), Forschungszentrum Jülich, 52425, Jülich, Germany.,Department of Nuclear Medicine, RWTH University Hospital Aachen, Aachen, Germany.,JARA - BRAIN - Translational Medicine, Aachen, Germany.,Centre of Integrated Oncology (CIO), University of Aachen, Bonn, Cologne and Düsseldorf, Germany
| |
Collapse
|
24
|
Tilelli CQ, Flôres LR, Cota VR, Castro OWD, Garcia-Cairasco N. Amygdaloid complex anatomopathological findings in animal models of status epilepticus. Epilepsy Behav 2021; 121:106831. [PMID: 31864944 DOI: 10.1016/j.yebeh.2019.106831] [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: 07/11/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022]
Abstract
Temporal lobe epileptic seizures are one of the most common and well-characterized types of epilepsies. The current knowledge on the pathology of temporal lobe epilepsy relies strongly on studies of epileptogenesis caused by experimentally induced status epilepticus (SE). Although several temporal lobe structures have been implicated in the epileptogenic process, the hippocampal formation is the temporal lobe structure studied in the greatest amount and detail. However, studies in human patients and animal models of temporal lobe epilepsy indicate that the amygdaloid complex can be also an important seizure generator, and several pathological processes have been shown in the amygdala during epileptogenesis. Therefore, in the present review, we systematically selected, organized, described, and analyzed the current knowledge on anatomopathological data associated with the amygdaloid complex during SE-induced epileptogenesis. Amygdaloid complex participation in the epileptogenic process is evidenced, among others, by alterations in energy metabolism, circulatory, and fluid regulation, neurotransmission, immediate early genes expression, tissue damage, cell suffering, inflammation, and neuroprotection. We conclude that major efforts should be made in order to include the amygdaloid complex as an important target area for evaluation in future research on SE-induced epileptogenesis. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Cristiane Queixa Tilelli
- Laboratory of Physiology, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Bairro Belvedere, Divinópolis, MG 35.501-296, Brazil.
| | - Larissa Ribeiro Flôres
- Laboratory of Physiology, Campus Centro-Oeste Dona Lindu, Universidade Federal de São João del-Rei, Av. Sebastião Gonçalves Coelho, 400, Bairro Belvedere, Divinópolis, MG 35.501-296, Brazil
| | - Vinicius Rosa Cota
- Laboratory of Neuroengineering and Neuroscience (LINNce), Department of Electrical Engineering, Campus Santo Antônio, Universidade Federal de São João del-Rei, Praça Frei Orlando, 170, Centro, São João Del Rei, MG 36307-352, Brazil
| | - Olagide Wagner de Castro
- Institute of Biological Sciences and Health, Campus A. C. Simões, Universidade Federal de Alagoas, Av. Lourival Melo Mota, s/n, Tabuleiro do Martins, Maceió, AL 57072-970, Brazil
| | - Norberto Garcia-Cairasco
- Neurophysiology and Experimental Neuroethology Laboratory (LNNE), Department of Physiology, School of Medicine, Universidade de São Paulo, Av. Bandeirantes, 3900, Monte Alegre, Ribeirão Preto, SP 14049-900, Brazil.
| |
Collapse
|
25
|
Endothelial-specific insulin receptor substrate-1 overexpression worsens neonatal hypoxic-ischemic brain injury via mTOR-mediated tight junction disassembly. Cell Death Discov 2021; 7:150. [PMID: 34226528 PMCID: PMC8257791 DOI: 10.1038/s41420-021-00548-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/09/2021] [Accepted: 06/09/2021] [Indexed: 12/20/2022] Open
Abstract
Hypoxic-ischemic (HI) encephalopathy is the major cause of mortality and disability in newborns. The neurovascular unit is a major target of acute and chronic brain injury, and therapies that protect simultaneously both neurons and vascular endothelial cells from neonatal HI injury are in demand. Insulin receptors and its key downstream molecule-insulin receptor substrate −1 (IRS-1) are potential neuroprotective targets and expressed both in neuron and endothelial cells. To investigate whether IRS-1 can act similarly in neurons and vascular endothelial cells in protecting neurovascular units and brain form HI injury, we found that neuron-specific IRS-1 transgenic rats showed reduced neurovascular injury and infarct volumes, whereas endothelial-specific IRS-1 transgenic rats showed increased blood-brain barrier (BBB) disruption and exaggerated neurovascular injury after neonatal HI brain injury. Endothelial-specific IRS-1 overexpression increased vascular permeability and disassembled the tight junction protein (zonula occludens-1) complex. Inhibition of mammalian target of rapamycin (mTOR) by rapamycin preserved tight junction proteins and attenuated BBB leakage and neuronal apoptosis after HI in the endothelial-specific IRS-1 transgenic pups. Together, our findings suggested that neuronal and endothelial IRS-1 had opposite effects on the neurovascular integrity and damage after neonatal HI brain injury and that endothelial IRS-1 worsens neurovascular integrity after HI via mTOR-mediated tight junction protein disassembly.
Collapse
|
26
|
Boux F, Forbes F, Collomb N, Zub E, Mazière L, de Bock F, Blaquiere M, Stupar V, Depaulis A, Marchi N, Barbier EL. Neurovascular multiparametric MRI defines epileptogenic and seizure propagation regions in experimental mesiotemporal lobe epilepsy. Epilepsia 2021; 62:1244-1255. [PMID: 33818790 DOI: 10.1111/epi.16886] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Improving the identification of the epileptogenic zone and associated seizure-spreading regions represents a significant challenge. Innovative brain-imaging modalities tracking neurovascular dynamics during seizures may provide new disease biomarkers. METHODS With use of a multi-parametric magnetic resonance imaging (MRI) analysis at 9.4 Tesla, we examined, elaborated, and combined multiple cellular and cerebrovascular MRI read-outs as imaging biomarkers of the epileptogenic and seizure-propagating regions. Analyses were performed in an experimental model of mesial temporal lobe epilepsy (MTLE) generated by unilateral intra-hippocampal injection of kainic acid (KA). RESULTS In the ipsilateral epileptogenic hippocampi, tissue T1 and blood-brain barrier (BBB) permeability to gadolinium were increased 48-72 hours post-KA, as compared to sham and contralateral hippocampi. BBB permeability endured during spontaneous focal seizures (4-6 weeks), along with a significant increase of apparent diffusion coefficient (ADC) and blood volume fraction (BVf). Simultaneously, ADC and BVf were augmented in the contralateral hippocampus, a region characterized by electroencephalographic seizure spreading, discrete histological neurovascular cell modifications, and no tissue sclerosis. We next asked whether combining all the acquired MRI parameters could deliver criteria to classify the epileptogenic from the seizure-spreading and sham hippocampi in these experimental conditions and over time. To differentiate sham from epileptogenic areas, the automatic multi-parametric classification provided a maximum accuracy of 97.5% (32 regions) 48-72 hours post-KA and of 100% (60 regions) at spontaneous seizures stage. To differentiate sham, epileptogenic, and seizure-spreading areas, the accuracies of the automatic classification were 93.1% (42 regions) 48-72 hours post-KA and 95% (80 regions) at spontaneous seizure stage. SIGNIFICANCE Combining multi-parametric MRI acquisition and machine-learning analyses delivers specific imaging identifiers to segregate the epileptogenic from the contralateral seizure-spreading hippocampi in experimental MTLE. The potential clinical value of our findings is critically discussed.
Collapse
Affiliation(s)
- Fabien Boux
- Univ. Grenoble Alpes, Grenoble Institut Neurosciences, Inserm, U1216, Grenoble 38000, France.,Inria, CNRS, G-INP, University of Grenoble Alpes, Grenoble, France
| | - Florence Forbes
- Inria, CNRS, G-INP, University of Grenoble Alpes, Grenoble, France
| | - Nora Collomb
- Univ. Grenoble Alpes, Grenoble Institut Neurosciences, Inserm, U1216, Grenoble 38000, France
| | - Emma Zub
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (University of Montpellier, UMR 5203 CNRS, U 1191 INSERM), Montpellier, France
| | - Lucile Mazière
- Univ. Grenoble Alpes, Grenoble Institut Neurosciences, Inserm, U1216, Grenoble 38000, France
| | - Fréderic de Bock
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (University of Montpellier, UMR 5203 CNRS, U 1191 INSERM), Montpellier, France
| | - Marine Blaquiere
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (University of Montpellier, UMR 5203 CNRS, U 1191 INSERM), Montpellier, France
| | - Vasile Stupar
- Univ. Grenoble Alpes, Grenoble Institut Neurosciences, Inserm, U1216, Grenoble 38000, France
| | - Antoine Depaulis
- Univ. Grenoble Alpes, Grenoble Institut Neurosciences, Inserm, U1216, Grenoble 38000, France
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (University of Montpellier, UMR 5203 CNRS, U 1191 INSERM), Montpellier, France
| | - Emmanuel L Barbier
- Univ. Grenoble Alpes, Grenoble Institut Neurosciences, Inserm, U1216, Grenoble 38000, France
| |
Collapse
|
27
|
Towner RA, Gulej R, Zalles M, Saunders D, Smith N, Lerner M, Morton KA, Richardson A. Rapamycin restores brain vasculature, metabolism, and blood-brain barrier in an inflammaging model. GeroScience 2021; 43:563-578. [PMID: 33846885 PMCID: PMC8110648 DOI: 10.1007/s11357-021-00363-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/06/2021] [Indexed: 02/08/2023] Open
Abstract
Rapamycin (RAPA) is found to have neuro-protective properties in various neuroinflammatory pathologies, including brain aging. With magnetic resonance imaging (MRI) techniques, we investigated the effect of RAPA in a lipopolysaccharide (LPS)-induced inflammaging model in rat brains. Rats were exposed to saline (control), or LPS alone or LPS combined with RAPA treatment (via food over 6 weeks). Arterial spin labeling (ASL) perfusion imaging was used to measure relative cerebral blood flow (rCBF). MR spectroscopy (MRS) was used to measure brain metabolite levels. Contrast-enhanced MRI (CE-MRI) was used to assess blood-brain barrier (BBB) permeability. Immunohistochemistry (IHC) was used to confirm neuroinflammation. RAPA restored NF-κB and HIF-1α to normal levels. RAPA was able to significantly restore rCBF in the cerebral cortex post-LPS exposure (p < 0.05), but not in the hippocampus. In the hippocampus, RAPA was able to restore total creatine (Cr) acutely, and N-acetyl aspartate (NAA) at 6 weeks, post-LPS. Myo-inositol (Myo-Ins) levels were found to decrease with RAPA treatment acutely post-LPS. RAPA was also able to significantly restore the BBB acutely post-LPS in both the cortex and hippocampus (p < 0.05 for both). RAPA was found to increase the percent change in BOLD signal in the cortex at 3 weeks, and in the hippocampus at 6 weeks post-LPS, compared to LPS alone. RAPA treatment also restored the neuronal and macro-vascular marker, EphB2, back to normal levels. These results indicate that RAPA may play an important therapeutic role in inhibiting neuroinflammation by normalizing brain vascularity, BBB, and some brain metabolites, and has a high translational capability.
Collapse
Affiliation(s)
- Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA.
- Neuroscience Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Nathan Shock Center for Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Rafal Gulej
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Michelle Zalles
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
- Neuroscience Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Megan Lerner
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kathryn A Morton
- Department of Radiology and Imaging Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Arlan Richardson
- Oklahoma Nathan Shock Center for Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
28
|
Deficiency of Microglial Autophagy Increases the Density of Oligodendrocytes and Susceptibility to Severe Forms of Seizures. eNeuro 2021; 8:ENEURO.0183-20.2021. [PMID: 33472865 PMCID: PMC7890520 DOI: 10.1523/eneuro.0183-20.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/11/2020] [Accepted: 01/02/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive activation of mTOR in microglia impairs CNS homeostasis and causes severe epilepsy. Autophagy constitutes an important part of mTOR signaling. The contribution of microglial autophagy to CNS homeostasis and epilepsy remains to be determined. Here, we report that ATG7KO mice deficient for autophagy in microglia display a marked increase of myelination markers, a higher density of mature oligodendrocytes (ODCs), and altered lengths of the nodes of Ranvier. Moreover, we found that deficiency of microglial autophagy (ATG7KO) leads to increased seizure susceptibility in three seizure models (pilocarpine, kainic acid, and amygdala kindling). We demonstrated that ATG7KO mice develop severe generalized seizures and display nearly 100% mortality to convulsions induced by pilocarpine and kainic acid. In the amygdala kindling model, we observed significant facilitation of contralateral propagation of seizures, a process underlying the development of generalized seizures. Taken together, our results reveal impaired microglial autophagy as a novel mechanism underlying altered homeostasis of ODCs and increased susceptibility to severe and fatal generalized seizures.
Collapse
|
29
|
Vila Verde D, de Curtis M, Librizzi L. Seizure-Induced Acute Glial Activation in the in vitro Isolated Guinea Pig Brain. Front Neurol 2021; 12:607603. [PMID: 33574794 PMCID: PMC7870799 DOI: 10.3389/fneur.2021.607603] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/04/2021] [Indexed: 12/02/2022] Open
Abstract
Introduction: It has been proposed that seizures induce IL-1β biosynthesis in astrocytes and increase blood brain barrier (BBB) permeability, even without the presence of blood borne inflammatory molecules and leukocytes. In the present study we investigate if seizures induce morphological changes typically observed in activated glial cells. Moreover, we will test if serum albumin extravasation into the brain parenchyma exacerbates neuronal hyperexcitability by inducing astrocytic and microglial activation. Methods: Epileptiform seizure-like events (SLEs) were induced in limbic regions by arterial perfusion of bicuculline methiodide (BMI; 50 μM) in the in vitro isolated guinea pig brain preparation. Field potentials were recorded in both the hippocampal CA1 region and the medial entorhinal cortex. BBB permeability changes were assessed by analyzing extravasation of arterially perfused fluorescein isothiocyanate (FITC)–albumin. Morphological changes in astrocytes and microglia were evaluated with tridimensional reconstruction and Sholl analysis in the ventral CA1 area of the hippocampus following application of BMI with or without co-perfusion of human serum albumin. Results: BMI-induced SLE promoted morphological changes of both astrocytes and microglia cells into an activated phenotype, confirmed by the quantification of the number and length of their processes. Human-recombinant albumin extravasation, due to SLE-induced BBB impairment, worsened both SLE duration and the activated glia phenotype. Discussion: Our study provides the first direct evidence that SLE activity per se is able to promote the activation of astro- and microglial cells, as observed by their changes in phenotype, in brain regions involved in seizure generation; we also hypothesize that gliosis, significantly intensified by h-recombinant albumin extravasation from the bloodstream to the brain parenchyma due to SLE-induced BBB disruption, is responsible for seizure activity reinforcement.
Collapse
Affiliation(s)
- Diogo Vila Verde
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Marco de Curtis
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Laura Librizzi
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| |
Collapse
|
30
|
Tesfaye BA, Hailu HG, Zewdie KA, Ayza MA, Berhe DF. Montelukast: The New Therapeutic Option for the Treatment of Epilepsy. J Exp Pharmacol 2021; 13:23-31. [PMID: 33505173 PMCID: PMC7829127 DOI: 10.2147/jep.s277720] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
Currently, there is no definitive cure for epilepsy. The available medications relieve symptoms and reduce seizure attacks. The major challenge with the available antiepileptic medication is safety and affordability. The repurposing of montelukast for epilepsy can be an alternative medication with a better safety profile. Montelukast is a leukotriene receptor antagonist that binds to the cysteinyl leukotrienes (CysLT) receptors used in the treatment of bronchial asthma and seasonal allergies. Emerging evidence suggests that montelukast's anti-inflammatory effect can help to maintain BBB integrity. The drug has also neuroprotective and anti-oxidative activities to reduce seizure incidence and epilepsy. The present review summarizes the neuropharmacological actions of montelukast in epilepsy with an emphasis on the recent findings associated with CysLT and cell-specific effects.
Collapse
Affiliation(s)
- Bekalu Amare Tesfaye
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Haftom Gebregergs Hailu
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Kaleab Alemayehu Zewdie
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Muluken Altaye Ayza
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| | - Derbew Fikadu Berhe
- Department of Pharmacology and Toxicology, School of Pharmacy, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
31
|
Zimmer TS, Broekaart DWM, Gruber VE, van Vliet EA, Mühlebner A, Aronica E. Tuberous Sclerosis Complex as Disease Model for Investigating mTOR-Related Gliopathy During Epileptogenesis. Front Neurol 2020; 11:1028. [PMID: 33041976 PMCID: PMC7527496 DOI: 10.3389/fneur.2020.01028] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberous sclerosis complex (TSC) represents the prototypic monogenic disorder of the mammalian target of rapamycin (mTOR) pathway dysregulation. It provides the rational mechanistic basis of a direct link between gene mutation and brain pathology (structural and functional abnormalities) associated with a complex clinical phenotype including epilepsy, autism, and intellectual disability. So far, research conducted in TSC has been largely neuron-oriented. However, the neuropathological hallmarks of TSC and other malformations of cortical development also include major morphological and functional changes in glial cells involving astrocytes, oligodendrocytes, NG2 glia, and microglia. These cells and their interglial crosstalk may offer new insights into the common neurobiological mechanisms underlying epilepsy and the complex cognitive and behavioral comorbidities that are characteristic of the spectrum of mTOR-associated neurodevelopmental disorders. This review will focus on the role of glial dysfunction, the interaction between glia related to mTOR hyperactivity, and its contribution to epileptogenesis in TSC. Moreover, we will discuss how understanding glial abnormalities in TSC might give valuable insight into the pathophysiological mechanisms that could help to develop novel therapeutic approaches for TSC or other pathologies characterized by glial dysfunction and acquired mTOR hyperactivation.
Collapse
Affiliation(s)
- Till S Zimmer
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Diede W M Broekaart
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | | | - Erwin A van Vliet
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| |
Collapse
|
32
|
Milikovsky DZ, Ofer J, Senatorov VV, Friedman AR, Prager O, Sheintuch L, Elazari N, Veksler R, Zelig D, Weissberg I, Bar-Klein G, Swissa E, Hanael E, Ben-Arie G, Schefenbauer O, Kamintsky L, Saar-Ashkenazy R, Shelef I, Shamir MH, Goldberg I, Glik A, Benninger F, Kaufer D, Friedman A. Paroxysmal slow cortical activity in Alzheimer's disease and epilepsy is associated with blood-brain barrier dysfunction. Sci Transl Med 2020; 11:11/521/eaaw8954. [PMID: 31801888 DOI: 10.1126/scitranslmed.aaw8954] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/13/2019] [Accepted: 10/09/2019] [Indexed: 12/12/2022]
Abstract
A growing body of evidence shows that epileptic activity is frequent but often undiagnosed in patients with Alzheimer's disease (AD) and has major therapeutic implications. Here, we analyzed electroencephalogram (EEG) data from patients with AD and found an EEG signature of transient slowing of the cortical network that we termed paroxysmal slow wave events (PSWEs). The occurrence per minute of the PSWEs was correlated with level of cognitive impairment. Interictal (between seizures) PSWEs were also found in patients with epilepsy, localized to cortical regions displaying blood-brain barrier (BBB) dysfunction, and in three rodent models with BBB pathology: aged mice, young 5x familial AD model, and status epilepticus-induced epilepsy in young rats. To investigate the potential causative role of BBB dysfunction in network modifications underlying PSWEs, we infused the serum protein albumin directly into the cerebral ventricles of naïve young rats. Infusion of albumin, but not artificial cerebrospinal fluid control, resulted in high incidence of PSWEs. Our results identify PSWEs as an EEG manifestation of nonconvulsive seizures in patients with AD and suggest BBB pathology as an underlying mechanism and as a promising therapeutic target.
Collapse
Affiliation(s)
- Dan Z Milikovsky
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jonathan Ofer
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Vladimir V Senatorov
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA.,Berkeley Stem Cell Center, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Aaron R Friedman
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ofer Prager
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Liron Sheintuch
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Netta Elazari
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Ronel Veksler
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Daniel Zelig
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Itai Weissberg
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Guy Bar-Klein
- Howard Hughes Medical Institute and the Institute of Genetic Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Evyatar Swissa
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Erez Hanael
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, P.O. Box 12, Rehovot 76100, Israel
| | - Gal Ben-Arie
- Department of Medical Imaging, Soroka University Medical Center, Beer-Sheva 84105, Israel
| | - Osnat Schefenbauer
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Lyna Kamintsky
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS B3H4R2, Canada
| | - Rotem Saar-Ashkenazy
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.,Faculty of Social Work, Ashkelon Academic College, Ashkelon 78211, Israel
| | - Ilan Shelef
- Department of Medical Imaging, Soroka University Medical Center, Beer-Sheva 84105, Israel
| | - Merav H Shamir
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, P.O. Box 12, Rehovot 76100, Israel
| | - Ilan Goldberg
- Department of Neurology, Wolfson Medical Center, Holon 58100, Israel
| | - Amir Glik
- Department of Neurology, Rabin Medical Center, Beilinson Hospital, Petach Tikva 49100, Israel.,Cognitive Neurology Clinic, Rabin Medical Center, Beilinson Hospital, Petach Tikva 49100, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Felix Benninger
- Department of Neurology, Rabin Medical Center, Beilinson Hospital, Petach Tikva 49100, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.,Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel. .,Department of Medical Neuroscience, Dalhousie University, Halifax, NS B3H4R2, Canada
| |
Collapse
|
33
|
Hanael E, Veksler R, Friedman A, Bar-Klein G, Senatorov VV, Kaufer D, Konstantin L, Elkin M, Chai O, Peery D, Shamir MH. Blood-brain barrier dysfunction in canine epileptic seizures detected by dynamic contrast-enhanced magnetic resonance imaging. Epilepsia 2020; 60:1005-1016. [PMID: 31032909 DOI: 10.1111/epi.14739] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Dogs with spontaneous or acquired epilepsy exhibit resemblance in etiology and disease course to humans, potentially offering a translational model of the human disease. Blood-brain barrier dysfunction (BBBD) has been shown to partake in epileptogenesis in experimental models of epilepsy. To test the hypothesis that BBBD can be detected in dogs with naturally occurring seizures, we developed a linear dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) analysis algorithm that was validated in clinical cases of seizing dogs and experimental epileptic rats. METHODS Forty-six dogs with naturally occurring seizures of different etiologies and 12 induced epilepsy rats were imaged using DCE-MRI. Six healthy dogs and 12 naive rats served as control. DCE-MRI was analyzed by linear-dynamic method. BBBD scores were calculated in whole brain and in specific brain regions. Immunofluorescence analysis for transforming growth factor beta (TGF-β) pathway proteins was performed on the piriform cortex of epileptic dogs. RESULTS We found BBBD in 37% of dogs with seizures. A significantly higher cerebrospinal fluid to serum albumin ratio was found in dogs with BBBD relative to dogs with intact blood-brain barrier (BBB). A significant difference was found between epileptic and control rats when BBBD scores were calculated for the piriform cortex at 48 hours and 1 month after status epilepticus. Mean BBBD score of the piriform lobe in idiopathic epilepsy (IE) dogs was significantly higher compared to control. Immunohistochemistry results suggested active TGF-β signaling and neuroinflammation in the piriform cortex of dogs with IE, showing increased levels of serum albumin colocalized with glial acidic fibrillary protein and pSMAD2 in an area where BBBD had been detected by linear DCE-MRI. SIGNIFICANCE Detection of BBBD in dogs with naturally occurring epilepsy provides the ground for future studies for evaluation of novel treatment targeting the disrupted BBB. The involvement of the piriform lobe seen using our linear DCE-MRI protocol and algorithm emphasizes the possibility of using dogs as a translational model for the human disease.
Collapse
Affiliation(s)
- Erez Hanael
- Hebrew University Koret School of Veterinary Medicine-Veterinary Teaching Hospital, Rehovot, Israel
| | - Ronel Veksler
- Departments of Physiology and Cell Biology, Brain, and Cognitive Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Brain, and Cognitive Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Department of Medical Neuroscience and Brain Repair Center, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Guy Bar-Klein
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Vladimir V Senatorov
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California.,Department of Integrative Biology, University of California, Berkeley, Berkeley, California
| | - Lilach Konstantin
- Hebrew University Koret School of Veterinary Medicine-Veterinary Teaching Hospital, Rehovot, Israel
| | - Maria Elkin
- Hebrew University Koret School of Veterinary Medicine-Veterinary Teaching Hospital, Rehovot, Israel
| | - Orit Chai
- Hebrew University Koret School of Veterinary Medicine-Veterinary Teaching Hospital, Rehovot, Israel
| | - Dana Peery
- Hebrew University Koret School of Veterinary Medicine-Veterinary Teaching Hospital, Rehovot, Israel
| | - Merav H Shamir
- Hebrew University Koret School of Veterinary Medicine-Veterinary Teaching Hospital, Rehovot, Israel
| |
Collapse
|
34
|
Swissa E, Serlin Y, Vazana U, Prager O, Friedman A. Blood-brain barrier dysfunction in status epileptics: Mechanisms and role in epileptogenesis. Epilepsy Behav 2019; 101:106285. [PMID: 31711869 DOI: 10.1016/j.yebeh.2019.04.038] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB), a unique anatomical and physiological interface between the central nervous system (CNS) and the peripheral circulation, is essential for the function of neural circuits. Interactions between the BBB, cerebral blood vessels, neurons, astrocytes, microglia, and pericytes form a dynamic functional unit known as the neurovascular unit (NVU). The NVU-BBB crosstalk plays a key role in the regulation of blood flow, response to injury, neuronal firing, and synaptic plasticity. Blood-brain barrier dysfunction (BBBD), a hallmark of brain injury, is a prominent finding in status epilepticus. Blood-brain barrier dysfunction is observed within the first hour of status epilepticus, and in epileptogenic brain regions, may last for months. Blood-brain barrier dysfunction was shown to have a role in astroglial dysfunction, neuroinflammation, increasing neural excitability, reduction of seizure threshold, excitatory synaptogenesis, impaired plasticity, and epileptogenesis. A key signaling pathway associated with BBBD-induced neurovascular dysfunction is the transforming growth factor beta (TGF-β) proinflammatory pathway, activated by the extravasation of serum albumin into the brain when BBB functions are compromised. Specific small molecules blocking TGF-β, and the nonspecific, Food and Drug Administration (FDA) approved blocker and angiotensin antagonist losartan, were shown to reduce BBBD and block epileptogenesis. With these encouraging preclinical data, we have developed imaging approach to quantitatively assess BBBD as a diagnostic, predictive, and pharmacodynamic biomarker after brain injury. Clinical trials in the foreseen future are expected to test the feasibility of BBB-targeted diagnostic coupled therapy in status epileptics and seizure disorders. This article is part of the Special Issue "Proceedings of the 7th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures".
Collapse
Affiliation(s)
- Evyatar Swissa
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Inter-Faculty Brain Science School, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva Israel
| | - Yonatan Serlin
- Neurology Residency Training Program, McGill University, Montreal, QC, Canada
| | - Udi Vazana
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Inter-Faculty Brain Science School, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva Israel
| | - Ofer Prager
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Inter-Faculty Brain Science School, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva Israel
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, The Inter-Faculty Brain Science School, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva Israel; Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
35
|
Jiang Y, Liu DF, Zhang X, Liu HG, Zhang JG. Microstructure and functional connectivity-based evidence for memory-related regional impairments in the brains of pilocarpine-treated rats. Brain Res Bull 2019; 154:127-134. [PMID: 31756422 DOI: 10.1016/j.brainresbull.2019.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/09/2019] [Accepted: 11/17/2019] [Indexed: 12/21/2022]
Abstract
Patients with temporal lobe epilepsy (TLE) frequently suffer from memory disorders, and the pathological changes show widespread regional impairments in the brain. In lithium-pilocarpine (LIP)-treated rats with TLE, an abnormal hippocampal microstructure and functional connectivity have been observed. However, changes in other brain regions are still unclear. In the present study, diffusion tensor imaging and functional magnetic resonance imaging (MRI) signals were collected in LIP-TLE rats and controls using a 7.0 T MRI. Microstructural parameters and functional connectivity were calculated among regions of interest (ROIs), including the bilateral prefrontal cortex, amygdala, hippocampus and entorhinal cortex. A correlation analysis was further performed between the neuroimaging results and the behavioral performance in the novel object and novel location memory tests. In our results, TLE rats showed increased fractional anisotropy (FA) values in the hippocampus and decreased FA values in the amygdala and entorhinal cortex. In addition, decreased functional connectivity between the amygdala and the CA3, and increased connectivity between the prefrontal cortex and the CA1 were observed in the TLE rats compared to control rats. Moreover, FA values in the amygdala, the hippocampus and the entorhinal cortex, as well as the amygdala-CA3 and the prefrontal-CA1 connectivity correlated with the memory performance. Based on our results, both the microstructure and functional connections were impaired in memory-related brain regions of LIP-TLE rats. Furthermore, the abnormal changes in the microstructure and functional connectivity were related to behavioral deficits in object and location memory.
Collapse
Affiliation(s)
- Yin Jiang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Beijing Key Laboratory of Neurostimulation, Beijing, China.
| | - De-Feng Liu
- Department of Functional Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Huan-Guang Liu
- Department of Functional Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian-Guo Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Beijing Key Laboratory of Neurostimulation, Beijing, China; Department of Functional Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
36
|
Simões PSR, Zanelatto AO, Assis MC, Varella PPV, Yacubian EM, Carrete H, Centeno R, Araujo MS, Cavalheiro EA, Tersariol ILS, Motta G, Naffah-Mazzacoratti MDG. Plasma kallikrein-kinin system contributes to peripheral inflammation in temporal lobe epilepsy. J Neurochem 2019; 150:296-311. [PMID: 31206169 DOI: 10.1111/jnc.14793] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/10/2019] [Accepted: 06/07/2019] [Indexed: 01/15/2023]
Abstract
Temporal lobe epilepsy (TLE) is a chronic disease, characterized by severe and refractory seizures, triggered in the hippocampus and/or amygdala, disrupting the blood-brain barrier. This disruption can sustain, or aggravate, the epileptic condition. The aim of this study was to evaluate the activation of the kallikrein-kinin system in patients with TLE, as it relates to the maintenance of blood-brain barrier. Human hippocampal sclerotic tissues removed after surgery for seizure control, plasma, and serum were used in the following assays: immunostaining for white blood cells in the TLE hippocampus, C-reactive protein in serum, quantification of plasma kallikrein (PKal) and cathepsin B (CatB) activity in serum and plasma, quantification of C1-inhibitor, analysis of high-molecular-weight kininogen (H-kininogen) fragments, and activation of plasma prekallikrein for comparison with healthy controls. Infiltration of white blood cells in the sclerotic hippocampus and a significant increase in the neutrophil/lymphocyte ratio in the blood of TLE patients were observed. High levels of C-reactive protein (TLE = 1.4 ± 0.3 µg/mL), PKal (TLE = 5.4 ± 0.4 U/mL), and CatB (TLE = 4.9 ± 0.4 U/mL) were also evident in the serum of TLE patients comparing to controls. A strong linear correlation was observed between active CatB and PKal in the serum of TLE patients (r = 0.88). High levels of cleaved H-kininogen and free PKal, and low levels of C1-inhibitor (TLE = 188 ± 12 µg/mL) were observed in the serum of TLE patients. Our data demonstrated that the plasma kallikrein-kinin system is activated in patients with TLE. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Priscila S R Simões
- Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | - Alexia O Zanelatto
- Departamento de Bioquímica, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | - Mirian C Assis
- Departamento de Bioquímica, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | - Pedro Paulo V Varella
- Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil.,Diagnóstico da América Sociedade Anônima (DASA), Barueri, SP, Brasil
| | - Elza Marcia Yacubian
- Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | - Henrique Carrete
- Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | - Ricardo Centeno
- Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | - Mariana S Araujo
- Departamento de Bioquímica, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | - Esper A Cavalheiro
- Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | | | - Guacyara Motta
- Departamento de Bioquímica, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| | - Maria da Graça Naffah-Mazzacoratti
- Departamento de Neurologia e Neurocirurgia, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil.,Departamento de Bioquímica, Escola Paulista de Medicina (UNIFESP), São Paulo, SP, Brasil
| |
Collapse
|
37
|
Rüber T, David B, Lüchters G, Nass RD, Friedman A, Surges R, Stöcker T, Weber B, Deichmann R, Schlaug G, Hattingen E, Elger CE. Evidence for peri-ictal blood-brain barrier dysfunction in patients with epilepsy. Brain 2019; 141:2952-2965. [PMID: 30239618 DOI: 10.1093/brain/awy242] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
Epilepsy has been associated with a dysfunction of the blood-brain barrier. While there is ample evidence that a dysfunction of the blood-brain barrier contributes to epileptogenesis, blood-brain barrier dysfunction as a consequence of single epileptic seizures has not been systematically investigated. We hypothesized that blood-brain barrier dysfunction is temporally and anatomically associated with epileptic seizures in patients and used a newly-established quantitative MRI protocol to test our hypothesis. Twenty-three patients with epilepsy undergoing inpatient monitoring as part of their presurgical evaluation were included in this study (10 females, mean age ± standard deviation: 28.78 ± 8.45). For each patient, we acquired quantitative T1 relaxation time maps (qT1) after both ictal and interictal injection of gadolinium-based contrast agent. The postictal enhancement of contrast agent was quantified by subtracting postictal qT1 from interictal qT1 and the resulting ΔqT1 was used as a surrogate imaging marker of peri-ictal blood-brain barrier dysfunction. Additionally, the serum concentrations of MMP9 and S100, both considered biomarkers of blood-brain barrier dysfunction, were assessed in serum samples obtained prior to and after the index seizure. Fifteen patients exhibited secondarily generalized tonic-clonic seizures and eight patients exhibited focal seizures at ictal injection of contrast agent. By comparing ΔqT1 of the generalized tonic-clonic seizures and focal seizures groups, the anatomical association between ictal epileptic activity and postictal enhancement of contrast agent could be probed. The generalized tonic-clonic seizures group showed significantly higher ΔqT1 in the whole brain as compared to the focal seizures group. Specific analysis of scans acquired later than 3 h after the onset of the seizure revealed higher ΔqT1 in the generalized tonic-clonic seizures group as compared to the focal seizures group, which was strictly lateralized to the hemisphere of seizure onset. Both MMP9 and S100 showed a significantly increased postictal concentration. The current study provides evidence for the occurrence of a blood-brain barrier dysfunction, which is temporally and anatomically associated with epileptic seizures. qT1 after ictal contrast agent injection is rendered as valuable imaging marker of seizure-associated blood-brain barrier dysfunction and may be measured hours after the seizure. The observation of the strong anatomical association of peri-ictal blood-brain barrier dysfunction may spark the development of new functional imaging modalities for the post hoc visualization of brain areas affected by the seizure.
Collapse
Affiliation(s)
- Theodor Rüber
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| | - Bastian David
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| | - Guido Lüchters
- Center for Development Research, University of Bonn, Bonn, Germany
| | - Robert D Nass
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Canada.,Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rainer Surges
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany.,Section of Epileptology, Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | - Tony Stöcker
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Bernd Weber
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| | - Ralf Deichmann
- Brain Imaging Center, Goethe University Frankfurt, Frankfurt, Germany
| | - Gottfried Schlaug
- Stroke Recovery Laboratory, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Elke Hattingen
- Department of Radiology, University of Bonn Medical Center, Bonn, Germany
| | - Christian E Elger
- Department of Epileptology, University of Bonn Medical Center, Bonn, Germany
| |
Collapse
|
38
|
Klement W, Blaquiere M, Zub E, deBock F, Boux F, Barbier E, Audinat E, Lerner-Natoli M, Marchi N. A pericyte-glia scarring develops at the leaky capillaries in the hippocampus during seizure activity. Epilepsia 2019; 60:1399-1411. [PMID: 31135065 DOI: 10.1111/epi.16019] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/28/2019] [Accepted: 04/28/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Inflammatory cerebrovascular damage occurs in epilepsy. Here, we tested the hypothesis that a pericyte-glia scar forms around the outer wall of hippocampal capillaries in a model of temporal lobe epilepsy associated with hippocampal sclerosis. We studied the participation of stromal cells expressing platelet-derived growth factor receptor beta (PDGFRβ) and extracellular matrix modifications to the perivascular scar during epileptogenesis. METHODS We used NG2DsRed/C57BL6 mice and induced status epilepticus (SE) followed by epileptogenesis and spontaneous recurrent seizures (SRS) by means of unilateral intrahippocampal injection of kainic acid (KA). For pharmacological assessment, we used organotypic hippocampal cultures (OHCs) where ictal electrographic activity was elicited by KA or bicuculline. RESULTS NG2DsRed pericytes, GFAP astroglia, and IBA1 microglia are reactive and converge to form a pericapillary multicellular scar in the CA hippocampal regions during epileptogenesis and at SRS. The capillaries are leaky as indicated by fluorescein entering the parenchyma from the peripheral blood. Concomitantly, PDGFRβ transcript and protein levels were significantly increased. Within the regional scar, a fibrotic-like PDGFRβ mesh developed around the capillaries, peaking at 1 week post-SE and regressing, but not resolving, at SRS. Abnormal distribution or accumulation of extracellular matrix collagens III/IV occurred in the CA regions during seizure progression. PDGFRβ/DAPI cells were in direct contact with or adjacent to the damaged NG2DsRed pericytes at the capillary interface, consistent with the notion of stromal cell reactivity or fibroblast formation. Inducing electrographic activity in OHCs was sufficient to augment PDGFRβ reactivity around the capillaries. The latter effect was pharmacologically mimicked by treating OHCs with the PDGFRβ agonist PDGF-BB and it was diminished by the PDGFRβ inhibitor imatinib. SIGNIFICANCE The reported multicellular activation and scar are traits of perivascular inflammation and hippocampal sclerosis in experimental epilepsy, with an implication for neurovascular dysfunction. Modulation of PDGFRβ could be exploited to target inflammation in this chronic disease setting.
Collapse
Affiliation(s)
- Wendy Klement
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Marine Blaquiere
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Emma Zub
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Frederic deBock
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Fabien Boux
- Grenoble Neuroscience Institute, GIN, Inserm U 1216 - Grenoble University, La Tronche, France
| | - Emmanuel Barbier
- Grenoble Neuroscience Institute, GIN, Inserm U 1216 - Grenoble University, La Tronche, France
| | - Etienne Audinat
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Mireille Lerner-Natoli
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Nicola Marchi
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
39
|
Gorter JA, Aronica E, van Vliet EA. The Roof is Leaking and a Storm is Raging: Repairing the Blood-Brain Barrier in the Fight Against Epilepsy. Epilepsy Curr 2019; 19:177-181. [PMID: 31037960 PMCID: PMC6610387 DOI: 10.1177/1535759719844750] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A large body of evidence that has accumulated over the past decade strongly supports the role of both blood–brain barrier (BBB) dysfunction and perivascular inflammation in the pathophysiology of epilepsy. Recent preclinical studies indicate that prolonged seizure- or brain injury-induced BBB dysfunction and subsequent perivascular inflammation may play an important role in post-traumatic epileptogenesis. In turn, perivascular inflammation can further sustain BBB dysfunction. In genetic epilepsies, such as tuberous sclerosis complex and other related epileptogenic developmental pathologies, there is an association between the underlying gene mutation, BBB dysfunction, and perivascular inflammation, but evidence for a causal link to epilepsy is lacking. Future neuroimaging studies might shed light on the role of BBB function in different epilepsies and address the potential for disease modification by targeting both the BBB and perivascular inflammation in acquired and genetic epilepsies.
Collapse
Affiliation(s)
- J A Gorter
- 1 Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - E Aronica
- 2 Department of (Neuro)pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,3 Stichting Epilepsie Instellingen Nederland (SEIN), the Netherlands
| | - E A van Vliet
- 1 Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands.,2 Department of (Neuro)pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
40
|
Neuroimaging Biomarkers of Experimental Epileptogenesis and Refractory Epilepsy. Int J Mol Sci 2019; 20:ijms20010220. [PMID: 30626103 PMCID: PMC6337422 DOI: 10.3390/ijms20010220] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 12/31/2018] [Accepted: 01/03/2019] [Indexed: 11/17/2022] Open
Abstract
This article provides an overview of neuroimaging biomarkers in experimental epileptogenesis and refractory epilepsy. Neuroimaging represents a gold standard and clinically translatable technique to identify neuropathological changes in epileptogenesis and longitudinally monitor its progression after a precipitating injury. Neuroimaging studies, along with molecular studies from animal models, have greatly improved our understanding of the neuropathology of epilepsy, such as the hallmark hippocampus sclerosis. Animal models are effective for differentiating the different stages of epileptogenesis. Neuroimaging in experimental epilepsy provides unique information about anatomic, functional, and metabolic alterations linked to epileptogenesis. Recently, several in vivo biomarkers for epileptogenesis have been investigated for characterizing neuronal loss, inflammation, blood-brain barrier alterations, changes in neurotransmitter density, neurovascular coupling, cerebral blood flow and volume, network connectivity, and metabolic activity in the brain. Magnetic resonance imaging (MRI) is a sensitive method for detecting structural and functional changes in the brain, especially to identify region-specific neuronal damage patterns in epilepsy. Positron emission tomography (PET) and single-photon emission computerized tomography are helpful to elucidate key functional alterations, especially in areas of brain metabolism and molecular patterns, and can help monitor pathology of epileptic disorders. Multimodal procedures such as PET-MRI integrated systems are desired for refractory epilepsy. Validated biomarkers are warranted for early identification of people at risk for epilepsy and monitoring of the progression of medical interventions.
Collapse
|
41
|
Presta I, Vismara M, Novellino F, Donato A, Zaffino P, Scali E, Pirrone KC, Spadea MF, Malara N, Donato G. Innate Immunity Cells and the Neurovascular Unit. Int J Mol Sci 2018; 19:E3856. [PMID: 30513991 PMCID: PMC6321635 DOI: 10.3390/ijms19123856] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022] Open
Abstract
Recent studies have clarified many still unknown aspects related to innate immunity and the blood-brain barrier relationship. They have also confirmed the close links between effector immune system cells, such as granulocytes, macrophages, microglia, natural killer cells and mast cells, and barrier functionality. The latter, in turn, is able to influence not only the entry of the cells of the immune system into the nervous tissue, but also their own activation. Interestingly, these two components and their interactions play a role of great importance not only in infectious diseases, but in almost all the pathologies of the central nervous system. In this paper, we review the main aspects in the field of vascular diseases (cerebral ischemia), of primitive and secondary neoplasms of Central Nervous System CNS, of CNS infectious diseases, of most common neurodegenerative diseases, in epilepsy and in demyelinating diseases (multiple sclerosis). Neuroinflammation phenomena are constantly present in all diseases; in every different pathological state, a variety of innate immunity cells responds to specific stimuli, differentiating their action, which can influence the blood-brain barrier permeability. This, in turn, undergoes anatomical and functional modifications, allowing the stabilization or the progression of the pathological processes.
Collapse
Affiliation(s)
- Ivan Presta
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Marco Vismara
- Department of Cell Biotechnologies and Hematology, University "La Sapienza" of Rome, 00185 Rome, Italy.
| | - Fabiana Novellino
- Institute of Molecular Bioimaging and Physiology, National Research Council, 88100 Catanzaro, Italy.
| | - Annalidia Donato
- Department of Medical and Surgical Sciences, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Paolo Zaffino
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Elisabetta Scali
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Krizia Caterina Pirrone
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Maria Francesca Spadea
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Natalia Malara
- Department of Clinical and Experimental Medicine, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Giuseppe Donato
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| |
Collapse
|
42
|
Blood-Brain Barrier Leakage during Early Epileptogenesis Is Associated with Rapid Remodeling of the Neurovascular Unit. eNeuro 2018; 5:eN-NWR-0123-18. [PMID: 29854942 PMCID: PMC5975718 DOI: 10.1523/eneuro.0123-18.2018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/01/2018] [Indexed: 01/26/2023] Open
Abstract
Increased permeability of the blood-brain barrier (BBB) following cerebral injury results in regional extravasation of plasma proteins and can critically contribute to the pathogenesis of epilepsy. Here, we comprehensively explore the spatiotemporal evolution of a main extravasation component, albumin, and illuminate associated responses of the neurovascular unit (NVU) contributing to early epileptogenic neuropathology. We applied translational in vivo MR imaging and complementary immunohistochemical analyses in the widely used rat pilocarpine post-status epilepticus (SE) model. The observed rapid BBB leakage affected major epileptogenesis-associated brain regions, peaked between 1 and 2 d post-SE, and rapidly declined thereafter, accompanied by cerebral edema generally following the same time course. At peak of BBB leakage, serum albumin colocalized with NVU constituents, such as vascular components, neurons, and brain immune cells. Surprisingly, astroglial markers did not colocalize with albumin, and aquaporin-4 (AQP4) was clearly reduced in areas of leaky BBB, indicating a severe disturbance of astrocyte-mediated endothelial-neuronal coupling. In addition, a distinct adaptive reorganization process of the NVU vasculature apparently takes place at sites of albumin presence, substantiated by reduced immunoreactivity of endothelial and changes in vascular basement membrane markers. Taken together, degenerative events at the level of the NVU, affecting vessels, astrocytes, and neurons, seem to outweigh reconstructive processes. Considering the rapidly occurring BBB leakage and subsequent impairment of the NVU, our data support the necessity of a prompt BBB-restoring treatment as one component of rational therapeutic intervention to prevent epileptogenesis and the development of other detrimental sequelae of SE.
Collapse
|
43
|
Giannoni P, Badaut J, Dargazanli C, Fayd'Herbe De Maudave A, Klement W, Costalat V, Marchi N. The pericyte-glia interface at the blood-brain barrier. Clin Sci (Lond) 2018; 132:361-374. [PMID: 29439117 DOI: 10.1042/cs20171634] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 12/30/2022]
Abstract
The cerebrovasculature is a multicellular structure with varying rheological and permeability properties. The outer wall of the brain capillary endothelium is enclosed by pericytes and astrocyte end feet, anatomically assembled to guarantee barrier functions. We, here, focus on the pericyte modifications occurring in disease conditions, reviewing evidence supporting the interplay amongst pericytes, the endothelium, and glial cells in health and pathology. Deconstruction and reactivity of pericytes and glial cells around the capillary endothelium occur in response to traumatic brain injury, epilepsy, and neurodegenerative disorders, impacting vascular permeability and participating in neuroinflammation. As this represents a growing field of research, addressing the multicellular reorganization occurring at the outer wall of the blood-brain barrier (BBB) in response to an acute insult or a chronic disease could disclose novel disease mechanisms and therapeutic targets.
Collapse
Affiliation(s)
| | - Jerome Badaut
- Laboratory of Brain Molecular Imaging, CNRS UMR5287, University of Bordeaux, France
- Basic Science Departments, Loma Linda University School of Medicine, CA, U.S.A
| | - Cyril Dargazanli
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Alexis Fayd'Herbe De Maudave
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Wendy Klement
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Vincent Costalat
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Nicola Marchi
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
44
|
Cerebrovascular heterogeneity and neuronal excitability. Neurosci Lett 2018; 667:75-83. [DOI: 10.1016/j.neulet.2017.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 01/01/2023]
|
45
|
Curatolo P, Moavero R, van Scheppingen J, Aronica E. mTOR dysregulation and tuberous sclerosis-related epilepsy. Expert Rev Neurother 2018; 18:185-201. [DOI: 10.1080/14737175.2018.1428562] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital, Rome, Italy
| | - Romina Moavero
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital, Rome, Italy
- Child Neurology Unit, Neuroscience and Neurorehabilitation Department, “Bambino Gesù” Children’s Hospital, IRCCS, Rome, Italy
| | - Jackelien van Scheppingen
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), The Netherlands
| |
Collapse
|
46
|
Van Skike CE, Galvan V. A Perfect sTORm: The Role of the Mammalian Target of Rapamycin (mTOR) in Cerebrovascular Dysfunction of Alzheimer's Disease: A Mini-Review. Gerontology 2018; 64:205-211. [PMID: 29320772 PMCID: PMC5876078 DOI: 10.1159/000485381] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/16/2017] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular dysfunction is detected prior to the onset of cognitive and histopathological changes in Alzheimer's disease (AD). Increasing evidence indicates a critical role of cerebrovascular dysfunction in the initiation and progression of AD. Recent studies identified the mechanistic/mammalian target of rapamycin (mTOR) as a critical effector of cerebrovascular dysfunction in AD. mTOR has a key role in the regulation of metabolism, but some mTOR-dependent mechanisms are uniquely specific to the regulation of cerebrovascular function. These include the regulation of cerebral blood flow, blood-brain barrier integrity and maintenance, neurovascular coupling, and cerebrovascular reactivity. This article examines the available evidence for a role of mTOR-driven cerebrovascular dysfunction in the pathogenesis of AD and of vascular cognitive impairment and dementia (VCID) and highlights the therapeutic potential of targeting mTOR and/or specific downstream effectors for vasculoprotection in AD, VCID, and other age-associated neurological diseases with cerebrovascular etiology.
Collapse
Affiliation(s)
- Candice E Van Skike
- Department of Cellular and Integrative Physiology and The Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX, USA
| | | |
Collapse
|
47
|
Neuroimaging in animal models of epilepsy. Neuroscience 2017; 358:277-299. [DOI: 10.1016/j.neuroscience.2017.06.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/27/2017] [Accepted: 06/28/2017] [Indexed: 02/06/2023]
|
48
|
Aronica E, Bauer S, Bozzi Y, Caleo M, Dingledine R, Gorter JA, Henshall DC, Kaufer D, Koh S, Löscher W, Louboutin JP, Mishto M, Norwood BA, Palma E, Poulter MO, Terrone G, Vezzani A, Kaminski RM. Neuroinflammatory targets and treatments for epilepsy validated in experimental models. Epilepsia 2017; 58 Suppl 3:27-38. [PMID: 28675563 DOI: 10.1111/epi.13783] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2017] [Indexed: 12/16/2022]
Abstract
A large body of evidence that has accumulated over the past decade strongly supports the role of inflammation in the pathophysiology of human epilepsy. Specific inflammatory molecules and pathways have been identified that influence various pathologic outcomes in different experimental models of epilepsy. Most importantly, the same inflammatory pathways have also been found in surgically resected brain tissue from patients with treatment-resistant epilepsy. New antiseizure therapies may be derived from these novel potential targets. An essential and crucial question is whether targeting these molecules and pathways may result in anti-ictogenesis, antiepileptogenesis, and/or disease-modification effects. Therefore, preclinical testing in models mimicking relevant aspects of epileptogenesis is needed to guide integrated experimental and clinical trial designs. We discuss the most recent preclinical proof-of-concept studies validating a number of therapeutic approaches against inflammatory mechanisms in animal models that could represent novel avenues for drug development in epilepsy. Finally, we suggest future directions to accelerate preclinical to clinical translation of these recent discoveries.
Collapse
Affiliation(s)
- Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, Amsterdam, The Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience University of Amsterdam, Amsterdam, The Netherlands.,SEIN-Stichting Epilepsie Instellingen Nederland, Heemstede, The Netherlands
| | - Sebastian Bauer
- Department of Neurology, Philipps University, Marburg, Germany.,Department of Neurology, Epilepsy Center Frankfurt Rhine-Main, Goethe University, Frankfurt am Main, Germany
| | - Yuri Bozzi
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy.,Laboratory of Molecular Neuropathology, Centre for Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Matteo Caleo
- Neuroscience Institute, National Research Council (CNR), Pisa, Italy
| | - Raymond Dingledine
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, U.S.A
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience University of Amsterdam, Amsterdam, The Netherlands
| | - David C Henshall
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, California, U.S.A
| | - Sookyong Koh
- Department of Pediatrics, Emory University, Atlanta, Georgia, U.S.A
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Jean-Pierre Louboutin
- Department of Basic Medical Sciences, University of the West Indies, Kingston, Jamaica.,Gene Therapy Program, University of Pennsylvania, Philadelphia, Pennsylvania, U.S.A
| | - Michele Mishto
- Charite University Medicine Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Braxton A Norwood
- Department of Neurology, Philipps University, Marburg, Germany.,Neuroscience Division, Expesicor LLC, Kalispell, Montana, U.S.A
| | - Eleonora Palma
- Department of Physiology and Pharmacology, University of Rome La Sapienza, Rome, Italy
| | - Michael O Poulter
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Gaetano Terrone
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Annamaria Vezzani
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | | |
Collapse
|
49
|
Bar-Klein G, Lublinsky S, Kamintsky L, Noyman I, Veksler R, Dalipaj H, Senatorov VV, Swissa E, Rosenbach D, Elazary N, Milikovsky DZ, Milk N, Kassirer M, Rosman Y, Serlin Y, Eisenkraft A, Chassidim Y, Parmet Y, Kaufer D, Friedman A. Imaging blood-brain barrier dysfunction as a biomarker for epileptogenesis. Brain 2017; 140:1692-1705. [PMID: 28444141 DOI: 10.1093/brain/awx073] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 01/31/2017] [Indexed: 12/30/2022] Open
Abstract
A biomarker that will enable the identification of patients at high-risk for developing post-injury epilepsy is critically required. Microvascular pathology and related blood-brain barrier dysfunction and neuroinflammation were shown to be associated with epileptogenesis after injury. Here we used prospective, longitudinal magnetic resonance imaging to quantitatively follow blood-brain barrier pathology in rats following status epilepticus, late electrocorticography to identify epileptic animals and post-mortem immunohistochemistry to confirm blood-brain barrier dysfunction and neuroinflammation. Finally, to test the pharmacodynamic relevance of the proposed biomarker, two anti-epileptogenic interventions were used; isoflurane anaesthesia and losartan. Our results show that early blood-brain barrier pathology in the piriform network is a sensitive and specific predictor (area under the curve of 0.96, P < 0.0001) for epilepsy, while diffused pathology is associated with a lower risk. Early treatments with either isoflurane anaesthesia or losartan prevented early microvascular damage and late epilepsy. We suggest quantitative assessment of blood-brain barrier pathology as a clinically relevant predictive, diagnostic and pharmaco!dynamics biomarker for acquired epilepsy.
Collapse
Affiliation(s)
- Guy Bar-Klein
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Svetlana Lublinsky
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lyn Kamintsky
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Iris Noyman
- Pediatric Neurology and Epilepsy, Pediatric Division, Soroka Medical Center, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ronel Veksler
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hotjensa Dalipaj
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Vladimir V Senatorov
- Department of Integrative Biology and the Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| | - Evyatar Swissa
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Dror Rosenbach
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Netta Elazary
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Dan Z Milikovsky
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nadav Milk
- The Israel Defense Force Medical Corps, Tel Hashomer, Israel
| | | | - Yossi Rosman
- The Israel Defense Force Medical Corps, Tel Hashomer, Israel.,Sackler School of Medicine, Tel Aviv Uneversity, Tel Aviv, Israel
| | - Yonatan Serlin
- Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Arik Eisenkraft
- The Israel Defense Force Medical Corps, Tel Hashomer, Israel.,NBC Protection Division, Ministry of Defense, Tel-Aviv, Israel.,The Institute for Research in Military Medicine, Hebrew University, Jerusalem, Israel
| | - Yoash Chassidim
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yisrael Parmet
- Department of Industrial Engineering and Management, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Daniela Kaufer
- Department of Integrative Biology and the Helen Wills Neuroscience Institute, University of California, Berkeley, California, USA
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Brain and Cognitive Sciences, Zlowotski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Medical Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
50
|
Broekaart DWM, van Scheppingen J, Geijtenbeek KW, Zuidberg MRJ, Anink JJ, Baayen JC, Mühlebner A, Aronica E, Gorter JA, van Vliet EA. Increased expression of (immuno)proteasome subunits during epileptogenesis is attenuated by inhibition of the mammalian target of rapamycin pathway. Epilepsia 2017. [DOI: 10.1111/epi.13823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Diede W. M. Broekaart
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Jackelien van Scheppingen
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Karlijne W. Geijtenbeek
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Mark R. J. Zuidberg
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Jasper J. Anink
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Johannes C. Baayen
- Department of Neurosurgery; VU University Medical Center; Vrije Universiteit; Amsterdam The Netherlands
| | - Angelika Mühlebner
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
- Swammerdam Institute for Life Sciences; Center for Neuroscience; University of Amsterdam; Amsterdam The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN); Heemstede The Netherlands
| | - Jan A. Gorter
- Swammerdam Institute for Life Sciences; Center for Neuroscience; University of Amsterdam; Amsterdam The Netherlands
| | - Erwin A. van Vliet
- Department of (Neuro)Pathology; Academic Medical Center; University of Amsterdam; Amsterdam The Netherlands
| |
Collapse
|