Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Neuroscience ; 155(1): 263-9, 2008 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-18534764

RESUMO

Neurovascular regulation, which is critical to the efficient functioning of the brain, is impaired in Alzheimer's disease and in transgenic mice overexpressing Abeta. Although senile plaques and neurofibrillary tangles represent neuropathological hallmarks of Alzheimer's disease, deposition of Abeta in cerebral blood vessels also likely plays a significant role in this debilitating and fatal disease. Further, soluble Abeta, which shows greater correlation with disease progression and severity than deposited plaques or tangles, displays strong vasoactive properties. The aim of this study was to develop a non-invasive model of cerebral vasoactivity that would ultimately be translatable to Alzheimer's disease as a marker for disease-modifying efficacy of novel small molecule and biologics drugs. Relative changes in cerebral blood volume following relevant doses of soluble Abeta(1-40) (0.01 or 0.1 mg/mouse), PBS, or the reverse peptide, Abeta(40-1) (0.01 or 0.1 mg/mouse), were monitored non-invasively by contrast-enhanced functional magnetic resonance imaging in anesthetized C57BL/6 mice. Experiments were performed on a 7T horizontal bore scanner using gradient echo echo-planar imaging. As expected, PBS and Abeta(40-1) did not induce any significant change in vascular response. In contrast, Abeta(1-40) significantly decreased CBV in a quantifiable, dose-related and region-specific manner. These data demonstrate for the first time the feasibility of characterizing pathogenic Abeta(1-40)-induced vascular dysfunction in vivo using a non-invasive approach. Further, this technique can be readily applied to preclinical screening in a longitudinal manner for novel drugs or antibodies targeting disease modification.


Assuntos
Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Imageamento por Ressonância Magnética/métodos , Fragmentos de Peptídeos , Animais , Mapeamento Encefálico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Processamento de Imagem Assistida por Computador/métodos , Camundongos , Oxigênio/sangue
2.
Eur J Pain ; 20(4): 607-14, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26392119

RESUMO

BACKGROUND: The receptor for advanced glycation end products (RAGE) is a multi-ligand receptor in the immunoglobulin superfamily. RAGE is localized throughout ascending sensory pathways (skin, peripheral nerve, dorsal root ganglion, spinal cord), and in cell types interacting with sensory neurons (endothelial cells, smooth muscle cells, monocytes and macrophages). Neuronal RAGE expression increases in pathological pain states in humans and rodents, and soluble RAGE attenuates thermal hypoalgesia in diabetic mice. The objective of the present study was to investigate whether pharmacological modulation of RAGE could attenuate mechanical allodynia in rodent pain models. METHODS: We developed an anti-RAGE monoclonal antibody (11E6) that binds to the C2 immunoglobulin domain of human RAGE, binds to mouse RAGE, and presumably to the same domain in mouse RAGE. The antinociceptive activity of 11E6 was investigated in mouse models of inflammatory (complete Freund's adjuvant) and neuropathic (chronic constriction injury of the sciatic nerve) pain. Mice were dosed intraperitoneally with 11E6 or IgG (negative control). RESULTS: Increased mechanical thresholds were observed following a single dose of 11E6 in both inflammatory and neuropathic pain models. Similar treatment with IgG did not alter nociceptive sensitivity. Repeated dosing with 11E6 significantly attenuated established mechanical hypersensitivity in a neuropathic pain model in a dose-related fashion. CONCLUSIONS: These data demonstrate that specific modulation of RAGE effectively attenuates nociceptive sensitivity associated with chronic inflammatory and neuropathic pain states.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Fatores Imunológicos/uso terapêutico , Neuralgia/tratamento farmacológico , Receptor para Produtos Finais de Glicação Avançada/antagonistas & inibidores , Animais , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Modelos Animais de Doenças , Feminino , Adjuvante de Freund , Gânglios Espinais/metabolismo , Humanos , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/etiologia , Neurônios Aferentes/metabolismo , Receptor para Produtos Finais de Glicação Avançada/imunologia , Nervo Isquiático/metabolismo , Medula Espinal/metabolismo
3.
Eur J Cell Biol ; 66(2): 136-50, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7774600

RESUMO

We have investigated the effect of stepwise truncating the carboxyterminal domain ("tail") of the intermediate filament (IF) protein vimentin of the clawed toad, Xenopus laevis, on filament assembly in vitro and, using cell transfection, in vivo and also on the cellular topology of the structures formed. All truncations examined, except the minimal one missing the last 11 amino acids which made the protein more sensitive to changes of ionic strength, did not significantly alter IF assembly in vitro, as judged by electron microscopy, viscometry and determination of viscoelastic properties with a laser-operated torsion pendulum. Stable transfections of vimentin-free mammalian cells with cDNAs encoding these mutations resulted at 28 degrees C, i.e. the permissive temperature for assembly of Xenopus vimentin, in the formation of extended IF bundle arrays. At 37 degrees C, however, the mutants lacking more than the last 35 amino acids could leave the cytoplasm and accumulated in the nucleus, indicating a certain topogenic element is located in the tail and directs cytoplasmic restriction in the wild-type protein although this does not form IFs under these conditions. Transfer to the nucleus is, however, abolished if the IF-consensus motif at the end of the rod domain is removed, suggesting that this part of the molecule also contributes to nuclear location. Similar results were obtained with human vimentin: While the rod entered the nucleus, headless vimentin, unable to form IFs, remained restricted to the cytoplasm owing to its tail domain. In contrast, tailless human vimentin and tailless mouse desmin, which are fully assembly-competent in vitro, both formed extensive IF arrays in the cytoplasm but did not accumulate in the nucleus. We conclude that in class III IF proteins stepwise deletions in the tail, while not considerably altering IF assembly in vitro, can change the topogenesis of IF proteins and structures in the living cell.


Assuntos
Filamentos Intermediários/química , Mutagênese/fisiologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Vimentina/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Núcleo Celular/metabolismo , Demecolcina/farmacologia , Desmina/química , Humanos , Dados de Sequência Molecular , Transfecção , Viscosidade , Xenopus laevis
4.
J Neurosci Methods ; 98(2): 145-54, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10880828

RESUMO

Cultures of primary neurons or thin brain slices are typically prepared from immature animals. We introduce a method to prepare hippocampal slice cultures from mature rats aged 20-30 days. Mature slice cultures retain hippocampal cytoarchitecture and synaptic connections up to 3 months in vitro. Spontaneous epileptiform activity is rarely observed suggesting long-term retention of normal neuronal excitability and of excitatory and inhibitory synaptic networks. Picrotoxin, a GABAergic Cl(-) channel antagonist, induced characteristic interictal-like bursts that originated in the CA3 region, but not in the CA1 region. These data suggest that mature slice cultures displayed long-term retention of GABAergic inhibitory synapses that effectively suppressed synchronized burst activity via recurrent excitatory synapses of CA3 pyramidal cells. Mature slice cultures lack the reactive synaptogenesis, spontaneous epileptiform activity, and short life span that limit the use of slice cultures isolated from immature rats. Mature slice cultures are anticipated to be a useful addition for the in vitro study of normal and pathological hippocampal function.


Assuntos
Hipocampo/fisiologia , Técnicas de Cultura de Órgãos/métodos , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Fatores Etários , Animais , Eletrofisiologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/citologia , Microscopia Eletrônica , Fibras Musgosas Hipocampais/fisiologia , Fibras Musgosas Hipocampais/ultraestrutura , Neuroglia/fisiologia , Neuroglia/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Piperazinas/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
5.
Br J Pharmacol ; 169(6): 1203-10, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23638877

RESUMO

Degenerative dementia is mainly caused by Alzheimer's disease and/or cerebrovascular abnormalities. Disturbance of the intracellular calcium homeostasis is central to the pathophysiology of neurodegeneration. In Alzheimer's disease, enhanced calcium load may be brought about by extracellular accumulation of amyloid-ß. Recent studies suggest that soluble forms facilitate influx through calcium-conducting ion channels in the plasma membrane, leading to excitotoxic neurodegeneration. Calcium channel blockade attenuates amyloid-ß-induced neuronal decline in vitro and is neuroprotective in animal models. Vascular dementia, on the other hand, is caused by cerebral hypoperfusion and may benefit from calcium channel blockade due to relaxation of the cerebral vasculature. Several calcium channel blockers have been tested in clinical trials of dementia and the outcome is heterogeneous. Nimodipine as well as nilvadipine prevent cognitive decline in some trials, whereas other calcium channel blockers failed. In trials with a positive outcome, BP reduction did not seem to play a role in preventing dementia, indicating a direct protecting effect on neurons. An optimization of calcium channel blockers for the treatment of dementia may involve an increase of selectivity for presynaptic calcium channels and an improvement of the affinity to the inactivated state. Novel low molecular weight compounds suitable for proof-of-concept studies are now available.


Assuntos
Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Demência/tratamento farmacológico , Nootrópicos/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Canais de Cálcio/química , Canais de Cálcio Tipo L/química , Canais de Cálcio Tipo L/metabolismo , Demência/metabolismo , Demência/fisiopatologia , Progressão da Doença , Drogas em Investigação/uso terapêutico , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transmissão Sináptica/efeitos dos fármacos
6.
Br J Pharmacol ; 167(4): 741-59, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22670568

RESUMO

P/Q-type calcium channels are high-voltage-gated calcium channels contributing to vesicle release at synaptic terminals. A number of neurological diseases have been attributed to malfunctioning of P/Q channels, including ataxia, migraine and Alzheimer's disease. To date, only two specific P/Q-type blockers are known: both are peptides deriving from the spider venom of Agelenopsis aperta, ω-agatoxins. Other peptidic calcium channel blockers with activity at P/Q channels are available, albeit with less selectivity. A number of low molecular weight compounds modulate P/Q-type currents with different characteristics, and some exhibit a peculiar bidirectional pattern of modulation. Interestingly, there are a number of therapeutics in clinical use, which also show P/Q channel activity. Because selectivity as well as the exact mode of action is different between all P/Q-type channel modulators, the interpretation of clinical and experimental data is complicated and needs a comprehensive understanding of their target profile. The situation is further complicated by the fact that information on potency varies vastly in the literature, which may be the result of different experimental systems, conditions or the splice variants of the P/Q channel. This review attempts to provide a comprehensive overview of the compounds available that affect the P/Q-type channel and should help with the interpretation of results of in vitro experiments and animal models. It also aims to explain some clinical observations by implementing current knowledge about P/Q channel modulation of therapeutically used non-selective drugs. Chances and challenges of the development of P/Q channel-selective molecules are discussed.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo P/fisiologia , Canais de Cálcio Tipo Q/fisiologia , Toxinas Biológicas/farmacologia , Animais , Bloqueadores dos Canais de Cálcio/uso terapêutico , Humanos , Toxinas Biológicas/uso terapêutico
7.
Br J Pharmacol ; 165(5): 1572-83, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21883149

RESUMO

BACKGROUND AND PURPOSE: ß-amyloid (Aß) oligomers have been implicated in the early pathophysiology of Alzheimer's disease (AD). While the precise nature of the molecular target has not been fully revealed, a number of studies have indicated that Aß oligomers modulate neuron-specific ion channels. We recently provided evidence that Aß oligomers suppress isolated P/Q-type calcium currents in cultured nerve cells. Using a heterologous expression system, we aimed to prove a direct effect on the membrane channel mediating such current. EXPERIMENTAL APPROACH: The effects of a synthetically generated Aß oligomer, Aß globulomer, were investigated on P/Q-type currents recorded from Xenopus laevis oocytes expressing the full P/Q-type calcium channel or the pore-forming subunit only. We also examined the effects of Aß globulomer on recombinant NMDA receptor currents. Finally, we compared the modulation by Aß globulomer with that induced by a synthetic monomeric Aß. KEY RESULTS: Aß globulomer directly and dose-dependently modulated P/Q-type calcium channels. A leftward shift of the current-voltage curve indicated that the threshold for channel opening was reduced. The effect of Aß globulomer was also present when only the α1A subunit of the normally tripartite channel was expressed. In contrast, the monomeric Aß had no effect on P/Q current. Also globulomer Aß had no effect on glutamate-induced NMDA currents. CONCLUSIONS AND IMPLICATIONS: The α1A subunit of the P/Q-type calcium channel is directly modulated by oligomeric Aß. Threshold reduction as well as an increase in current at synaptic terminals may facilitate vesicle release and could trigger excitotoxic events in the brains of patients with AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Canais de Cálcio Tipo P/metabolismo , Canais de Cálcio Tipo Q/metabolismo , Animais , Células Cultivadas , Feminino , Ácido Glutâmico/metabolismo , Potenciais da Membrana/fisiologia , N-Metilaspartato/metabolismo , Neurônios/metabolismo , Oócitos/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Xenopus laevis
8.
Br J Pharmacol ; 159(7): 1523-31, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20233208

RESUMO

BACKGROUND AND PURPOSE: Alzheimer's disease (AD) is a multifactorial, neurodegenerative disease, which is in part caused by an impairment of synaptic function, probably mediated by oligomeric forms of amyloid-beta (Abeta). While the Abeta pathology mainly affects the physiology of neurotransmission, neuronal decline is caused by excitotoxic cell death, which is mediated by the NMDA receptor. A comprehensive therapeutic approach should address both Abeta-induced synaptic deficits, as well as NMDA receptor-mediated neurodegeneration, via one molecular target. This study was designed to test whether calpain could be involved in both pathological pathways, which would offer a promising avenue for new treatments. EXPERIMENTAL APPROACH: Application of the specific, water-soluble calpain inhibitor A-705253 was used to inhibit calpain in hippocampal slice cultures. We examined whether inhibition of calpain would prevent Abeta-induced deficits in neurotransmission in CA1, as well as NMDA-induced neuronal cell death. KEY RESULTS: A-705253 dose-dependently prevented excitotoxicity-induced neurodegeneration at low nanomolar concentrations, determined by propidium iodide histochemistry. Inhibition of the NMDA receptor similarly protected from neuronal damage. Caspase staining indicated that calpain inhibition was protective by reducing apoptosis. Electrophysiological analysis revealed that inhibition of calpain by A-705253 also fully prevented Abeta oligomer-induced deficits in neurotransmission. The protective effect of calpain was compared to the clinically available NMDA receptor antagonist memantine, which was also effective in this model. CONCLUSIONS AND IMPLICATIONS: We suggest that inhibition of calpain exhibits a promising strategy to address several aspects of the pathology of AD that may go beyond the available therapeutic intervention by memantine.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Benzamidas/farmacologia , Calpaína/antagonistas & inibidores , Morte Celular/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , N-Metilaspartato/farmacologia , Sinapses/efeitos dos fármacos , Animais , Hipocampo/fisiopatologia , Técnicas In Vitro , Masculino , Ratos , Ratos Wistar , Sinapses/fisiologia , Transmissão Sináptica
9.
Neuroscience ; 162(4): 1081-90, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19477243

RESUMO

One of the hallmarks of Alzheimer's disease is the accumulation of amyloid plaques in brains of affected patients. Several recent studies provided evidence that soluble oligomer forms of amyloid-beta (Abeta) rather than plaques determine cognitive decline. In vitro studies using artificial Abeta oligomer preparations suggest that such pathophysiology is caused by a specific impairment of synaptic function. We examined whether synaptic deficits occur before deposition of insoluble fibrillar Abeta by analyzing brain slices taken from young Tg2576 mice overexpressing mutant amyloid precursor protein. Excitatory synaptic transmission in the hippocampal CA1 region was strongly impaired before plaque development, suggesting a dissociation of an early synaptic impairment, probably caused by soluble oligomeric amyloid-beta, from subsequent plaque formation. At higher age neurotransmission was also decreased in wild type mice, paralleling a cognitive decline of normal aged animals. Memory formation in rats is accompanied by distinct hippocampal network oscillations. It has recently been shown that hippocampal gamma oscillations, a network correlate of exploratory behavior, are impaired in amyloid precursor protein (APP)-overexpressing mice. We determined whether sharp wave-ripple complexes, which contribute to memory consolidation during slow wave-sleep, are modified in Tg2576 mice. Interestingly, neither sharp waves nor superimposed ripples were changed at pre-plaque or plaque stages. During aging, however, there was a strong reduction of sharp wave frequency and ripple energy in wild type and APP-overexpressing animals. This indicates that the reported changes in network oscillations following APP-overexpression are specific for gamma oscillations, whereas aging has a more general effect on network properties. Taken together our data suggest that non-fibrillar forms of Abeta--possibly Abeta oligomers--specifically interfere with synaptic function in Tg2576, but do not globally alter memory-related network properties. We propose that mechanisms leading to Abeta-related cognitive decline are different from those related to aging.


Assuntos
Envelhecimento/fisiologia , Precursor de Proteína beta-Amiloide/genética , Comportamento Animal , Potenciais Pós-Sinápticos Excitadores , Placa Amiloide/patologia , Transmissão Sináptica , Precursor de Proteína beta-Amiloide/biossíntese , Animais , Feminino , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Técnicas In Vitro , Memória , Camundongos , Camundongos Transgênicos
10.
J Cell Sci ; 113 ( Pt 13): 2471-83, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10852826

RESUMO

Plakophilin 1 and 2 (PKP1, PKP2) are members of the arm-repeat protein family. They are both constitutively expressed in most vertebrate cells, in two splice forms named a and b, and display a remarkable dual location: they occur in the nuclei of cells and, in epithelial cells, at the plasma membrane within the desmosomal plaques. We have shown by solid phase-binding assays that both PKP1a and PKP2a bind to intermediate filament (IF) proteins, in particular to cytokeratins (CKs) from epidermal as well as simple epithelial cells and, to some extent, to vimentin. In line with this we show that recombinant PKP1a binds strongly to IFs assembled in vitro from CKs 8/18, 5/14, vimentin or desmin and integrates them into thick (up to 120 nm in diameter) IF bundles extending for several microm. The basic amino-terminal, non-arm-repeat domain of PKP1a is necessary and sufficient for this specific interaction as shown by blot overlay and centrifugation experiments. In particular, the binding of PKP1a to IF proteins is saturable at an approximately equimolar ratio. In extracts from HaCaT cells, distinct soluble complexes containing PKP1a and desmoplakin I (DPI) have been identified by co-immunoprecipitation and sucrose density fractionation. The significance of these interactions of PKP1a with IF proteins on the one hand and desmoplakin on the other is discussed in relation to the fact that PKP1a is not bound - and does not bind - to extended IFs in vivo. We postulate that (1) effective cellular regulatory mechanisms exist that prevent plakophilins from unscheduled IF-binding, and (2) specific desmoplakin interactions with either PKP1, PKP2 or PKP3, or combinations thereof, are involved in the selective recruitment of plakophilins to the desmosomal plaques.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Proteínas/metabolismo , Extratos Celulares/imunologia , Centrifugação , Desmoplaquinas , Desmossomos/metabolismo , Técnicas In Vitro , Proteínas de Filamentos Intermediários/ultraestrutura , Microscopia Eletrônica , Placofilinas , Ensaio Radioligante , Ressonância de Plasmônio de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA