Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Neurol ; 14: 1129138, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37034097

RESUMO

Introduction: FIRES is a rare epileptic encephalopathy induced by acute unremitting seizures that occur suddenly in healthy children or young adults after a febrile illness in the preceding 2 weeks. This condition results in high mortality, neurological disability, and drug-resistant epilepsy. The development of new therapeutics is hampered by the lack of validated experimental models. Our goal was to address this unmet need by providing a simple tool for rapid throughput screening of new therapies that target pathological inflammatory mechanisms in FIRES. The model was not intended to mimic the etiopathogenesis of FIRES which is still unknown, but to reproduce salient features of its clinical presentation such as the age, the cytokine storm and the refractoriness of epileptic activity to antiseizure medications (ASMs). Methods: We refined an in vitro model of mouse hippocampal/temporal cortex acute slices where drug-resistant epileptic activity is induced by zero Mg2+/100 µM 4-aminopirydine. Clinical evidence suggests that acute unremitting seizures in FIRES are promoted by neuroinflammation triggered in the brain by the preceding infection. We mimicked this inflammatory component by exposing slices for 30 min to 10 µg/ml lipopolysaccharide (LPS). Results: LPS induced a sustained neuroinflammatory response, as shown by increased mRNA levels of IL-1ß, CXCL1 (IL-8), TNF, and increased IL-1ß/IL-1Ra ratio. Epileptiform activity was exacerbated by neuroinflammation, also displaying increased resistance to maximal therapeutic concentrations of midazolam (100 µM), phenytoin (50 µM), sodium valproate (800 µM), and phenobarbital (100 µM). Treatment of LPS-exposed slices with two immunomodulatory drugs, a mouse anti-IL-6 receptor antibody (100 µM) corresponding to tocilizumab in humans, or anakinra (1.3 µM) which blocks the IL-1 receptor type 1, delayed the onset of epileptiform events and strongly reduced the ASM-resistant epileptiform activity evoked by neuroinflammation. These drugs were shown to reduce ASM-refractory seizures in FIRES patients. Discussion: The neuroinflammatory component and the pharmacological responsiveness of epileptiform events provide a proof-of-concept validation of this in vitro model for the rapid selection of new treatments for acute ASM-refractory seizures in FIRES.

2.
J Cereb Blood Flow Metab ; 43(5): 680-693, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36655331

RESUMO

Brain ischemia is a common acute injury resulting from impaired blood flow to the brain. Translation of effective drug candidates from experimental models to patients has systematically failed. The use of human induced pluripotent stem cells (iPSC) offers new opportunities to gain translational insights into diseases including brain ischemia. We used a human 3D self-assembling iPSC-derived model (human cortical organoids, hCO) to characterize the effects of ischemia caused by oxygen-glucose deprivation (OGD). hCO exposed to 2 h or 8 h of OGD had neuronal death and impaired neuronal network complexity, measured in whole-mounting microtubule-associated protein 2 (MAP-2) immunostaining. Neuronal vulnerability was reflected by a reduction in MAP-2 mRNA levels, and increased release of neurofilament light chain (NfL) in culture media, proportional to OGD severity. Glial fibrillary acidic protein (GFAP) gene or protein levels did not change in hCO, but their release in medium increased after prolonged OGD. In conclusion, this human 3D iPSC-based in vitro model of brain ischemic injury is characterized by marked neuronal injury reflected by the release of the translational biomarker NfL which is relevant for testing neuroprotective strategies.


Assuntos
Isquemia Encefálica , Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Isquemia Encefálica/metabolismo , Oxigênio/metabolismo , Morte Celular , Glucose/farmacologia , Organoides/metabolismo , Células Cultivadas
3.
Parkinsonism Relat Disord ; 106: 105229, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36462409

RESUMO

BACKGROUND AND PURPOSE: Parkinson's disease remains orphan of valuable therapies capable to interfere with the disease pathogenesis despite the large number of symptomatic approaches adopted in clinical practice to manage this disease. Treatments simultaneously affecting α-synuclein (α-syn) oligomerization and neuroinflammation may counteract Parkinson's disease and related disorders. Recent data demonstrate that Doxycycline, a tetracycline antibiotic, can inhibit α-syn aggregation as well as neuroinflammation. We herein investigate, for the first time, the potential therapeutic properties of Doxy in a human α-syn A53T transgenic Parkinson's disease mouse model evaluating behavioural, biochemical and histopathological parameters. EXPERIMENTAL APPROACH: Human α-syn A53T transgenic mice were treated with Doxycycline (10 mg/kg daily ip) for 30 days. The effect of treatment on motor, cognitive and daily live activity performances were examined. Neuropathological and neurophysiological parameters were assessed through immunocytochemical, electrophysiological and biochemical analysis of cerebral tissue. KEY RESULTS: Doxy treatment abolished cognitive and daily life activity deficiencies in A53T mice. The effect on cognitive functions was associated with neuroprotection, inhibition of α-syn oligomerization and gliosis both in the cortex and hippocampus. Doxy treatment restored hippocampal long-term potentiation in association with the inhibition of pro-inflammatory cytokines expression. Moreover, Doxy ameliorated motor impairment and reduced striatal glial activation in A53T mice. CONCLUSIONS AND IMPLICATIONS: Our findings promote Doxy as a valuable multi-target therapeutic approach counteracting both symptoms and neuropathology in the complex scenario of α-synucleinopathies.


Assuntos
Doxiciclina , Doença de Parkinson , Sinucleinopatias , Animais , Humanos , Camundongos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Modelos Animais de Doenças , Doxiciclina/farmacologia , Doxiciclina/uso terapêutico , Camundongos Transgênicos , Doenças Neuroinflamatórias , Doença de Parkinson/metabolismo , Sinucleinopatias/tratamento farmacológico , Reposicionamento de Medicamentos
4.
Neuropathol Appl Neurobiol ; 48(1): e12758, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34388852

RESUMO

AIMS: The causes of distinct patterns of reduced cortical thickness in the common human epilepsies, detectable on neuroimaging and with important clinical consequences, are unknown. We investigated the underlying mechanisms of cortical thinning using a systems-level analysis. METHODS: Imaging-based cortical structural maps from a large-scale epilepsy neuroimaging study were overlaid with highly spatially resolved human brain gene expression data from the Allen Human Brain Atlas. Cell-type deconvolution, differential expression analysis and cell-type enrichment analyses were used to identify differences in cell-type distribution. These differences were followed up in post-mortem brain tissue from humans with epilepsy using Iba1 immunolabelling. Furthermore, to investigate a causal effect in cortical thinning, cell-type-specific depletion was used in a murine model of acquired epilepsy. RESULTS: We identified elevated fractions of microglia and endothelial cells in regions of reduced cortical thickness. Differentially expressed genes showed enrichment for microglial markers and, in particular, activated microglial states. Analysis of post-mortem brain tissue from humans with epilepsy confirmed excess activated microglia. In the murine model, transient depletion of activated microglia during the early phase of the disease development prevented cortical thinning and neuronal cell loss in the temporal cortex. Although the development of chronic seizures was unaffected, the epileptic mice with early depletion of activated microglia did not develop deficits in a non-spatial memory test seen in epileptic mice not depleted of microglia. CONCLUSIONS: These convergent data strongly implicate activated microglia in cortical thinning, representing a new dimension for concern and disease modification in the epilepsies, potentially distinct from seizure control.


Assuntos
Epilepsia , Microglia , Animais , Encéfalo , Células Endoteliais , Epilepsia/metabolismo , Camundongos , Microglia/metabolismo , Convulsões
5.
Brain ; 144(12): 3710-3726, 2021 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-34972208

RESUMO

Aggregation and cytoplasmic mislocalization of TDP-43 are pathological hallmarks of amyotrophic lateral sclerosis and frontotemporal dementia spectrum. However, the molecular mechanism by which TDP-43 aggregates form and cause neurodegeneration remains poorly understood. Cyclophilin A, also known as peptidyl-prolyl cis-trans isomerase A (PPIA), is a foldase and molecular chaperone. We previously found that PPIA interacts with TDP-43 and governs some of its functions, and its deficiency accelerates disease in a mouse model of amyotrophic lateral sclerosis. Here we characterized PPIA knock-out mice throughout their lifespan and found that they develop a neurodegenerative disease with key behavioural features of frontotemporal dementia, marked TDP-43 pathology and late-onset motor dysfunction. In the mouse brain, deficient PPIA induces mislocalization and aggregation of the GTP-binding nuclear protein Ran, a PPIA interactor and a master regulator of nucleocytoplasmic transport, also for TDP-43. Moreover, in absence of PPIA, TDP-43 autoregulation is perturbed and TDP-43 and proteins involved in synaptic function are downregulated, leading to impairment of synaptic plasticity. Finally, we found that PPIA was downregulated in several patients with amyotrophic lateral sclerosis and amyotrophic lateral sclerosis-frontotemporal dementia, and identified a PPIA loss-of-function mutation in a patient with sporadic amyotrophic lateral sclerosis . The mutant PPIA has low stability, altered structure and impaired interaction with TDP-43. These findings strongly implicate that defective PPIA function causes TDP-43 mislocalization and dysfunction and should be considered in future therapeutic approaches.


Assuntos
Esclerose Lateral Amiotrófica/genética , Ciclofilina A/genética , Demência Frontotemporal/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Ciclofilina A/deficiência , Proteínas de Ligação a DNA/metabolismo , Demência Frontotemporal/patologia , Humanos , Camundongos , Camundongos Knockout
6.
Epilepsia ; 62(8): 1931-1945, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34128226

RESUMO

OBJECTIVE: Microgliosis occurs in animal models of acquired epilepsy and in patients. It includes cell proliferation that is associated with seizure frequency and decreased neuronal cells in human epilepsy. The role of microglia proliferation in the development of acquired epilepsy is unknown; thus, we examined its contribution to spontaneous seizure, neurodegeneration, and cognitive deficits in different disease phases. METHODS: We used a model of acquired epilepsy triggered by intra-amygdala kainic acid in C57BL6N adult male mice. Mice were electroencephalographically (EEG) monitored (24/7) during status epilepticus and in early and chronic disease. Microglia proliferation was blocked by GW2580, a selective CSF1 receptor inhibitor, supplemented in the diet for 21 days from status epilepticus onset. Then, mice were returned to placebo diet until experiment completion. Control mice were exposed to status epilepticus and fed with placebo diet. Experimental mice were tested in the novel object recognition test (NORT) and in Barnes maze, and compared to control and sham mice. At the end of the behavioral test, mice were killed for brain histopathological analysis. Additionally, seizure baseline was monitored in chronic epileptic mice, then mice were fed for 14 days with GW2580 or placebo diet under 24/7 EEG recording. RESULTS: GW2580 prevented microglia proliferation in mice undergoing epilepsy, whereas it did not affect microglia or basal excitatory neurotransmission in the hippocampus of naive mice. Mice with occluded microglia proliferation during early disease development underwent status epilepticus and subsequent epilepsy similar to placebo diet mice, and were similarly impaired in NORT, with improvement in Barnes maze. GW2580-treated mice displayed neuroprotection in the hippocampus. In contrast, blockade of microglia proliferation in chronic epileptic mice resulted in spontaneous seizure reduction versus placebo mice. SIGNIFICANCE: Microglia proliferation during early disease contributes to neurodegeneration, whereas in late chronic disease it contributes to seizures. Timely pharmacological interference with microglia proliferation may offer a potential target for improving disease outcomes.


Assuntos
Epilepsia , Estado Epiléptico , Animais , Proliferação de Células , Modelos Animais de Doenças , Epilepsia/etiologia , Hipocampo , Humanos , Ácido Caínico/toxicidade , Masculino , Camundongos , Microglia , Convulsões , Estado Epiléptico/induzido quimicamente
7.
Prog Mol Biol Transl Sci ; 177: 175-202, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33453941

RESUMO

After Alzheimer's disease, Parkinson's disease is the most frequent neurodegenerative disorder. Although numerous treatments have been developed to control the disease symptomatology, with some successes, an efficacious therapy affecting the causes of PD is still a goal to pursue. The genetic evidence and the identification of α-synuclein as the main component of intracellular Lewy bodies, the neuropathological hallmark of PD and related disorders, have changed the approach to these disorders. More recently, the detrimental role of α-synuclein has been further extended to explain the wide spread of cerebral pathology through its oligomers. To emphasize the central pathogenic role of these soluble aggregates, we have defined synucleinopathies and other neurodegenerative disorders associated with protein misfolding as oligomeropathies. Another common element in the pathogenesis of oligomeropathies is the role played by inflammation, both at the peripheral and cerebral levels. In the brain parenchyma, inflammatory reaction has been considered an obvious consequence of neuronal degeneration, but recent observations indicate a direct contribution of glial alteration in the early phase of the disease. Furthermore, systemic inflammation also influences the development of neuronal dysfunction caused by specific elements, ß amyloid, α-synuclein, tau or prion. However, each disorder has its own specific pathological process and within the same pathological condition, it is possible to find inter-individual differences. This heterogeneity might explain the difficulties developing efficacious therapeutic approaches, even though the possibility of intervention is supported by robust biological evidence. We have recently demonstrated that peripheral inflammation can amplify the neuronal dysfunction induced by α-synuclein oligomers and the neuropathological consequences observed in a Parkinson's disease model. In both cases, activation of microglia was incremented by the "double hit" process, compared to the single treatment. In contrast, astrocyte activation was attenuated and these cells appeared damaged when chronic inflammation was combined with α-synuclein exposure. This evidence might indicate a more specific anti-inflammatory strategy rather than the generic anti-inflammatory treatment.


Assuntos
Doença de Alzheimer , Doença de Parkinson , Humanos , Inflamação , Doença de Parkinson/terapia , alfa-Sinucleína
8.
Front Aging Neurosci ; 11: 284, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31680937

RESUMO

Alzheimer's disease (AD) is a complex, multi-factorial disease affecting various brain systems. This complexity implies that successful therapies must be directed against several core neuropathological targets rather than single ones. The scientific community has made great efforts to identify the right AD targets beside the historic amyloid-ß (Aß). Neuroinflammation is re-emerging as determinant in the neuropathological process of AD. A new theory, still in its infancy, highlights the role of gut microbiota (GM) in the control of brain development, but also in the onset and progression of neurodegenerative diseases. Bidirectional communication between the central and the enteric nervous systems, called gut-brain axes, is largely influenced by GM and the immune system is a potential key mediator of this interaction. Growing evidence points to the role of GM in the maturation and activation of host microglia and peripheral immune cells. Several recent studies have found abnormalities in GM (dysbiosis) in AD populations. These observations raise the intriguing question whether and how GM dysbiosis could contribute to AD development through action on the immune system and whether, in a therapeutic prospective, the development of strategies preserving a healthy GM might become a valuable approach to prevent AD. Here, we review the evidence from animal models and humans of the role of GM in neuroinflammation and AD.

9.
Brain Behav Immun ; 69: 591-602, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29458199

RESUMO

Alpha-synuclein oligomers (α-synOs) are emerging as crucial factors in the pathogenesis of synucleinopathies. Although the connection between neuroinflammation and α-syn still remains elusive, increasing evidence suggests that extracellular moieties activate glial cells leading to neuronal damage. Using an acute mouse model, we explored whether α-synOs induce memory impairment in association to neuroinflammation, addressing Toll-like receptors 2 and 4 (TLR2 and TLR4) involvement. We found that α-synOs abolished mouse memory establishment in association to hippocampal glial activation. On brain slices α-synOs inhibited long-term potentiation. Indomethacin and Ibuprofen prevented the α-synOs-mediated detrimental actions. Furthermore, while the TLR2 functional inhibitor antibody prevented the memory deficit, oligomers induced memory deficits in the TLR4 knockout mice. In conclusion, solely α-synOs induce memory impairment likely inhibiting synaptic plasticity. α-synOs lead to hippocampal gliosis that is involved in memory impairment. Moreover, while the oligomer-mediated detrimental actions are TLR2 dependent, the involvement of TLR4 was ruled out.


Assuntos
Hipocampo/efeitos dos fármacos , Memória/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Receptor 2 Toll-Like/metabolismo , alfa-Sinucleína/farmacologia , Animais , Hipocampo/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Camundongos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Reconhecimento Psicológico/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
10.
Biol Psychiatry ; 81(3): 179-192, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27587266

RESUMO

BACKGROUND: Dysregulation of Ras-extracellular signal-related kinase (ERK) signaling gives rise to RASopathies, a class of neurodevelopmental syndromes associated with intellectual disability. Recently, much attention has been directed at models bearing mild forms of RASopathies whose behavioral impairments can be attenuated by inhibiting the Ras-ERK cascade in the adult. Little is known about the brain mechanisms in severe forms of these disorders. METHODS: We performed an extensive characterization of a new brain-specific model of severe forms of RASopathies, the KRAS12V mutant mouse. RESULTS: The KRAS12V mutation results in a severe form of intellectual disability, which parallels mental deficits found in patients bearing mutations in this gene. KRAS12V mice show a severe impairment of both short- and long-term memory in a number of behavioral tasks. At the cellular level, an upregulation of ERK signaling during early phases of postnatal development, but not in the adult state, results in a selective enhancement of synaptogenesis in gamma-aminobutyric acidergic interneurons. The enhancement of ERK activity in interneurons at this critical postnatal time leads to a permanent increase in the inhibitory tone throughout the brain, manifesting in reduced synaptic transmission and long-term plasticity in the hippocampus. In the adult, the behavioral and electrophysiological phenotypes in KRAS12V mice can be temporarily reverted by inhibiting gamma-aminobutyric acid signaling but not by a Ras-ERK blockade. Importantly, the synaptogenesis phenotype can be rescued by a treatment at the developmental stage with Ras-ERK inhibitors. CONCLUSIONS: These data demonstrate a novel mechanism underlying inhibitory synaptogenesis and provide new insights in understanding mental dysfunctions associated to RASopathies.


Assuntos
Encéfalo/fisiologia , Neurônios GABAérgicos/fisiologia , Deficiência Intelectual/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Sinapses/fisiologia , Animais , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Neurônios GABAérgicos/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiologia , Potenciais Pós-Sinápticos Inibidores , Potenciação de Longa Duração , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Receptores de GABA/metabolismo , Comportamento Social , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
11.
Neurobiol Dis ; 99: 12-23, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27939857

RESUMO

We recently discovered that forebrain activation of the IL-1 receptor/Toll-like receptor (IL-1R1/TLR4) innate immunity signal plays a pivotal role in neuronal hyperexcitability underlying seizures in rodents. Since this pathway is activated in neurons and glia in human epileptogenic foci, it represents a potential target for developing drugs interfering with the mechanisms of epileptogenesis that lead to spontaneous seizures. The lack of such drugs represents a major unmet clinical need. We tested therefore novel therapies inhibiting the IL-1R1/TLR4 signaling in an established murine model of acquired epilepsy. We used an epigenetic approach by injecting a synthetic mimic of micro(mi)RNA-146a that impairs IL1R1/TLR4 signal transduction, or we blocked receptor activation with antiinflammatory drugs. Both interventions when transiently applied to mice after epilepsy onset, prevented disease progression and dramatically reduced chronic seizure recurrence, while the anticonvulsant drug carbamazepine was ineffective. We conclude that IL-1R1/TLR4 is a novel potential therapeutic target for attaining disease-modifications in patients with diagnosed epilepsy.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Anticonvulsivantes/administração & dosagem , Epilepsia/terapia , MicroRNAs/administração & dosagem , Receptores Tipo I de Interleucina-1/antagonistas & inibidores , Receptor 4 Toll-Like/antagonistas & inibidores , Animais , Carbamazepina/farmacologia , Cianobactérias , Dipeptídeos/administração & dosagem , Modelos Animais de Doenças , Epilepsia/tratamento farmacológico , Epilepsia/fisiopatologia , Hipocampo/fisiopatologia , Ácido Caínico , Lipopolissacarídeos/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL , Oligonucleotídeos/administração & dosagem , Distribuição Aleatória , Receptores Tipo I de Interleucina-1/metabolismo , Fatores de Tempo , Receptor 4 Toll-Like/metabolismo , para-Aminobenzoatos/administração & dosagem
12.
Sci Rep ; 6: 36441, 2016 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-27819308

RESUMO

Some mutant forms of the cellular prion protein (PrPC) carrying artificial deletions or point mutations associated with familial human prion diseases are capable of inducing spontaneous ionic currents across the cell membrane, conferring hypersensitivity to certain antibiotics to a wide range of cultured cells and primary cerebellar granular neurons (CGNs). These effects are abrogated when the wild type (WT) form is co-expressed, suggesting that they might be related to a physiological activity of PrPC. Interestingly, the prion protein family member Shadoo (Sho) makes cells hypersensitive to the same antibiotics as mutant PrP-s, an effect that is diminished by the co-expression of WT-PrP. Here, we report that Sho engages in another mutant PrP-like activity: it spontaneously induces large ionic currents in cultured SH-SY5Y cells, as detected by whole-cell patch clamping. These currents are also decreased by the co-expression of WT-PrP. Furthermore, deletion of the N-terminal (RXXX)8 motif of Sho, mutation of the eight arginine residues of this motif to glutamines, or replacement of the hydrophobic domain by that of PrP, also diminish Sho-induced ionic currents. Our results suggest that the channel activity that is also characteristic to some pathogenic PrP mutants may be linked to a physiological function of Sho.


Assuntos
Potenciais de Ação/fisiologia , Proteínas Ligadas por GPI/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Motivos de Aminoácidos , Antibacterianos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteínas Ligadas por GPI/genética , Células HEK293 , Humanos , Mutagênese , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Plasmídeos/genética , Plasmídeos/metabolismo , Domínios Proteicos
13.
Sci Rep ; 6: 23180, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26976106

RESUMO

Prion diseases are rare neurodegenerative conditions associated with the conformational conversion of the cellular prion protein (PrP(C)) into PrP(Sc), a self-replicating isoform (prion) that accumulates in the central nervous system of affected individuals. The structure of PrP(Sc) is poorly defined, and likely to be heterogeneous, as suggested by the existence of different prion strains. The latter represents a relevant problem for therapy in prion diseases, as some potent anti-prion compounds have shown strain-specificity. Designing therapeutics that target PrP(C) may provide an opportunity to overcome these problems. PrP(C) ligands may theoretically inhibit the replication of multiple prion strains, by acting on the common substrate of any prion replication reaction. Here, we characterized the properties of a cationic tetrapyrrole [Fe(III)-TMPyP], which was previously shown to bind PrP(C), and inhibit the replication of a mouse prion strain. We report that the compound is active against multiple prion strains in vitro and in cells. Interestingly, we also find that Fe(III)-TMPyP inhibits several PrP(C)-related toxic activities, including the channel-forming ability of a PrP mutant, and the PrP(C)-dependent synaptotoxicity of amyloid-ß (Aß) oligomers, which are associated with Alzheimer's Disease. These results demonstrate that molecules binding to PrP(C) may produce a dual effect of blocking prion replication and inhibiting PrP(C)-mediated toxicity.


Assuntos
Metaloporfirinas/química , Proteínas PrPC/metabolismo , Proteínas Priônicas/antagonistas & inibidores , Tetrapirróis/química , Peptídeos beta-Amiloides/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Células HEK293 , Humanos , Metaloporfirinas/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Porfirinas , Proteínas PrPC/química , Proteínas PrPC/genética , Proteínas Priônicas/química , Ligação Proteica , Proteínas Recombinantes/metabolismo , Tetrapirróis/farmacologia
14.
Cell Rep ; 13(7): 1353-1365, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26549453

RESUMO

The basal ganglia play a critical role in shaping motor behavior. For this function, the activity of medium spiny neurons (MSNs) of the striatonigral and striatopallidal pathways must be integrated. It remains unclear whether the activity of the two pathways is primarily coordinated by synaptic plasticity mechanisms. Using a model of Parkinson's disease, we determined the circuit and behavioral effects of concurrently regulating cell-type-specific forms of corticostriatal long-term synaptic depression (LTD) by inhibiting small-conductance Ca(2+)-activated K(+) channels (SKs) of the dorsolateral striatum. At striatopallidal synapses, SK channel inhibition rescued the disease-linked deficits in endocannabinoid (eCB)-dependent LTD. At striatonigral cells, inhibition of these channels counteracted a form of adenosine-mediated LTD by activating the ERK cascade. Interfering with eCB-, adenosine-, and ERK signaling in vivo alleviated motor abnormalities, which supports that synaptic modulation of striatal pathways affects behavior. Thus, our results establish a central role of coordinated synaptic plasticity at MSN subpopulations in motor control.


Assuntos
Corpo Estriado/patologia , Plasticidade Neuronal , Substância Negra/patologia , Adenosina/fisiologia , Animais , Córtex Cerebral/citologia , Córtex Cerebral/patologia , Dopamina/fisiologia , Neurônios Dopaminérgicos/fisiologia , Potenciais Pós-Sinápticos Excitadores , Depressão Sináptica de Longo Prazo , Sistema de Sinalização das MAP Quinases , Camundongos , Atividade Motora , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/patologia , Receptor A1 de Adenosina/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo
15.
Biol Psychiatry ; 77(2): 106-15, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24844602

RESUMO

BACKGROUND: Bidirectional long-term plasticity at the corticostriatal synapse has been proposed as a central cellular mechanism governing dopamine-mediated behavioral adaptations in the basal ganglia system. Balanced activity of medium spiny neurons (MSNs) in the direct and the indirect pathways is essential for normal striatal function. This balance is disrupted in Parkinson's disease and in l-3,4-dihydroxyphenylalanine (l-DOPA)-induced dyskinesia (LID), a common motor complication of current pharmacotherapy of Parkinson's disease. METHODS: Electrophysiological recordings were performed in mouse cortico-striatal slice preparation. Synaptic plasticity, such as long-term potentiation (LTP) and depotentiation, was investigated. Specific pharmacological inhibitors or genetic manipulations were used to modulate the Ras-extracellular signal-regulated kinase (Ras-ERK) pathway, a signal transduction cascade implicated in behavioral plasticity, and synaptic activity in different subpopulations of striatal neurons was measured. RESULTS: We found that the Ras-ERK pathway, is not only essential for long-term potentiation induced with a high frequency stimulation protocol (HFS-LTP) in the dorsal striatum, but also for its reversal, synaptic depotentiation. Ablation of Ras-guanine nucleotide-releasing factor 1 (Ras-GRF1), a neuronal activator of Ras proteins, causes a specific loss of HFS-LTP in the medium spiny neurons in the direct pathway without affecting LTP in the indirect pathway. Analysis of LTP in animals with unilateral 6-hydroxydopamine lesions (6-OHDA) rendered dyskinetic with chronic L-DOPA treatment reveals a complex, Ras-GRF1 and pathway-independent, apparently stochastic involvement of ERK. CONCLUSIONS: These data not only demonstrate a central role for Ras-ERK signaling in striatal LTP, depotentiation, and LTP restored after L-DOPA treatment but also disclose multifaceted synaptic adaptations occurring in response to dopaminergic denervation and pulsatile administration of L-DOPA.


Assuntos
Corpo Estriado/fisiopatologia , Discinesia Induzida por Medicamentos/fisiopatologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Plasticidade Neuronal/fisiologia , ras-GRF1/metabolismo , Animais , Antiparkinsonianos/toxicidade , Butadienos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiopatologia , Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Levodopa/toxicidade , Camundongos Knockout , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Nitrilas/farmacologia , Oxidopamina , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Técnicas de Cultura de Tecidos , ras-GRF1/genética
16.
Neurobiol Learn Mem ; 105: 63-80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23827407

RESUMO

The striatum is the input structure of the basal ganglia system. By integrating glutamatergic signals from cortical and subcortical regions and dopaminergic signals from mesolimbic nuclei the striatum functions as an important neural substrate for procedural and motor learning as well as for reward-guided behaviors. In addition, striatal activity is significantly altered in pathological conditions in which either a loss of dopamine innervation (Parkinson's disease) or aberrant dopamine-mediated signaling (drug addiction and L-DOPA induced dyskinesia) occurs. Here we discuss cellular mechanisms of striatal synaptic plasticity and aspects of cell signaling underlying striatum-dependent behavior, with a major focus on the neuromodulatory action of the endocannabinoid system and on the role of the Ras-ERK cascade.


Assuntos
Corpo Estriado/fisiologia , Plasticidade Neuronal , Receptores Dopaminérgicos/fisiologia , Animais , Humanos , Camundongos , Transdução de Sinais/fisiologia
17.
Nat Neurosci ; 15(2): 284-93, 2012 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-22231426

RESUMO

Endocannabinoids (eCBs) regulate neuronal activity in the dorso-lateral striatum (DLS), a brain region that is involved in habitual behaviors. How synaptic eCB signaling contributes to habitual behaviors under physiological and pathological conditions remains unclear. Using a mouse model of cannabinoid tolerance, we found that persistent activation of the eCB pathway impaired eCB-mediated long-term depression (LTD) and synaptic depotentiation in the DLS. The loss of eCB LTD, occurring preferentially at cortical connections to striatopallidal neurons, was associated with a shift in behavioral control from goal-directed action to habitual responding. eCB LTD and behavioral alterations were rescued by in vivo modulation of small-conductance calcium activated potassium channel (SK channel) activity in the DLS, which potentiates eCB signaling. Our results reveal a direct relationship between drug tolerance and changes in control of instrumental performance by establishing a central role for eCB LTD in habit expression. In addition, SK channels emerge as molecular targets to fine tune the eCB pathway under pathological conditions.


Assuntos
Canabinoides/administração & dosagem , Corpo Estriado/efeitos dos fármacos , Tolerância a Medicamentos/fisiologia , Hábitos , Depressão Sináptica de Longo Prazo/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Animais , Apamina/farmacologia , Benzamidas/farmacologia , Biofísica , Canabinoides/agonistas , Canabinoides/antagonistas & inibidores , Carbamatos/farmacologia , Condicionamento Operante/efeitos dos fármacos , Corpo Estriado/citologia , Cicloexanóis/farmacocinética , Relação Dose-Resposta a Droga , Dronabinol/farmacologia , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Ácido Okadáico/farmacologia , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Ligação Proteica/efeitos dos fármacos , Pirazóis/farmacologia , Rimonabanto , Bloqueadores dos Canais de Sódio/farmacologia , Trítio/farmacocinética
18.
Proc Natl Acad Sci U S A ; 107(50): 21824-9, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21115823

RESUMO

L-dopa-induced dyskinesia (LID) is a common debilitating complication of dopamine replacement therapy in Parkinson's disease. Recent evidence suggests that LID may be linked causally to a hyperactivation of the Ras-ERK signaling cascade in the basal ganglia. We set out to determine whether specific targeting of Ras-guanine nucleotide-releasing factor 1 (Ras-GRF1), a brain-specific activator of the Ras-ERK pathway, may provide a therapy for LID. On the rodent abnormal involuntary movements scale, Ras-GRF1-deficient mice were significantly resistant to the development of dyskinesia during chronic L-dopa treatment. Furthermore, in a nonhuman primate model of LID, lentiviral vectors expressing dominant negative forms of Ras-GRF1 caused a dramatic reversion of dyskinesia severity leaving intact the therapeutic effect of L-dopa. These data reveal the central role of Ras-GRF1 in governing striatal adaptations to dopamine replacement therapy and validate a viable treatment for LID based on intracellular signaling modulation.


Assuntos
Corpo Estriado/efeitos dos fármacos , Corpo Estriado/fisiologia , Discinesia Induzida por Medicamentos/fisiopatologia , Levodopa/farmacologia , Transdução de Sinais/fisiologia , ras-GRF1/metabolismo , Animais , Corpo Estriado/citologia , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , ras-GRF1/genética
19.
J Neurosci Methods ; 185(1): 1-14, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19699233

RESUMO

The use of viral vectors as gene transfer tools for the central nervous system has seen a significant growth in the last decade. Improvements in the safety, efficiency and specificity of vectors for clinical applications have proven to be beneficial also for basic neuroscience research. This review will discuss the viral systems currently available to neuroscientists and some of the recent achievements in the study of synaptic function, memory and drug addiction.


Assuntos
Encéfalo/metabolismo , Técnicas de Transferência de Genes/tendências , Vetores Genéticos/genética , Vírus/genética , Animais , Química Encefálica/genética , Regulação da Expressão Gênica , Humanos , Aprendizagem/fisiologia , Plasticidade Neuronal/genética , Transtornos Relacionados ao Uso de Substâncias/genética
20.
J Neurosci ; 26(21): 5810-8, 2006 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-16723539

RESUMO

Chronic exposure to Delta9-tetrahydrocannabinol (THC) induces tolerance to cannabinoid-induced locomotor effects, which are mediated by cannabinoid receptors (CB1Rs) located in motor control regions, including the cerebellum. There is substantial evidence of cerebellar CB1R molecular adaptation and modifications in receptor signaling after prolonged cannabinoid exposure. However, very little is known about the effects of chronic cannabinoid administration on cerebellar synaptic plasticity, which may contribute to the development of cannabinoid behavioral tolerance. In the cerebellar cortex, activation of CB1R inhibits excitatory synaptic transmission at parallel fiber (PF)-Purkinje cell (PC) synapses by decreasing neurotransmitter release. Our study aimed to investigate the neurophysiological adaptive responses occurring at cerebellar PF-PC cell synapses after repeated THC exposure. In THC-tolerant mice, an increase of the basal release probability was found at PF-PC synapses, in parallel with a facilitation of slow mGluR1 (metabotropic glutamate receptor type 1)-mediated excitatory postsynaptic currents and a reduced sensitivity to the inhibitory effects of the CB1R agonist CP55,940 [(-)-cis-3-[2-hydroxy-4-(1,1-dimethylheptyl)phenyl]-trans-4-(3-hydroxypropyl)cyclohexanol]. Additionally, after repeated THC exposures, presynaptic PF-PC long-term potentiation was blocked by A1R (adenosine receptor-1) activation. Inhibition of the extracellular signal regulated kinase (ERK) pathway prevented these alterations of cerebellar synaptic transmission and plasticity. In summary, we provide evidence for ERK-dependent modulatory mechanisms at PF-PC synapses after chronic THC administration. This contributes to generation of forms of pathological synaptic plasticity that might play a role in cannabinoid dependence.


Assuntos
Cerebelo/efeitos dos fármacos , Cerebelo/fisiologia , Dronabinol/administração & dosagem , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Transmissão Sináptica/fisiologia , Animais , Células Cultivadas , Tolerância a Medicamentos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Transmissão Sináptica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...