Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Differ ; 29(3): 657-669, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34625662

RESUMO

The sequential activation of Nucleotide-binding oligomerization domain, Leucine rich Repeat and Pyrin domain containing protein 1 (Nlrp1) inflammasome, Caspase-1 (Casp1), and Caspase-6 (Casp6) is implicated in primary human neuron cultures and Alzheimer Disease (AD) neurodegeneration. To validate the Nlrp1-Casp1-Casp6 pathway in vivo, the APPSwedish/Indiana J20 AD transgenic mouse model was generated on either a Nlrp1, Casp1 or Casp6 null genetic background and mice were studied at 4-5 months of age. Episodic memory deficits assessed with novel object recognition were normalized by genetic ablation of Nlrp1, Casp1, or Casp6 in J20 mice. Spatial learning deficits, assessed with the Barnes Maze, were normalized in genetically ablated J20, whereas memory recall was normalized in J20/Casp1-/- and J20/Casp6-/-, and improved in J20/Nlrp1-/- mice. Hippocampal CA1 dendritic spine density of the mushroom subtype was reduced in J20, and normalized in genetically ablated J20 mice. Reduced J20 hippocampal dentate gyrus and CA3 synaptophysin levels were normalized in genetically ablated J20. Increased Iba1+-microglia in the hippocampus and cortex of J20 brains were normalized by Casp1 and Casp6 ablation and reduced by Nlrp1 ablation. Increased pro-inflammatory cytokines, TNF-α and CXCL1, in the J20 hippocampus were normalized by Nlrp1 or Casp1 genetic ablation. CXCL1 was also normalized by Casp6 genetic ablation. IFN-γ was increased and total amyloid ß peptide was decreased in genetically ablated Nlrp1, Casp1 or Casp6 J20 hippocampi. We conclude that Nlrp1, Casp1, or Casp6 are implicated in AD-related cognitive impairment, inflammation, and amyloidogenesis. These results indicate that Nlrp1, Casp1, and Casp6 represent rational therapeutic targets against cognitive impairment and inflammation in AD.


Assuntos
Doença de Alzheimer , Caspase 1/metabolismo , Caspase 6/metabolismo , Disfunção Cognitiva , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Caspase 6/genética , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Inflamassomos/metabolismo , Inflamação , Camundongos , Camundongos Transgênicos
3.
Cell Death Dis ; 12(3): 227, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33649324

RESUMO

Active Caspase-6 (Casp6) and Tau cleaved by Casp6 at amino acids 402 (Tau∆D402) and 421 (Tau∆D421) are present in early Alzheimer disease intraneuronal neurofibrillary tangles, which are made primarily of filamentous Tau aggregates. To assess whether Casp6 cleavage of Tau contributes to Tau pathology and Casp6-mediated age-dependent cognitive impairment, we generated transgenic knock-in mouse models that conditionally express full-length human Tau (hTau) 0N4R only (CTO) or together with human Casp6 (hCasp6) (CTC). Region-specific hippocampal and cortical hCasp6 and hTau expression were confirmed with western blot and immunohistochemistry in 2-25-month-old brains. Casp6 activity was confirmed with Tau∆D421 and Tubulin cleaved by Casp6 immunopositivity in 3-25-month-old CTC, but not in CTO, brains. Immunoprecipitated Tau∆D402 was detected in both CTC and CTO brains, but was more abundant in CTC brains. Intraneuronal hippocampal Tau hyperphosphorylation at S202/T205, S422, and T231, and Tau conformational change were absent in both CTC and CTO brains. A slight accumulation of Tau phosphorylated at S396/404 and S202 was observed in Cornu Ammonis 1 (CA1) hippocampal neuron soma of CTC compared to CTO brains. Eighteen-month-old CTC brains showed rare argentophilic deposits that increased by 25 months, whereas CTO brains only displayed them sparsely at 25 months. Tau microtubule binding was equivalent in CTC and CTO hippocampi. Episodic and spatial memory measured with novel object recognition and Barnes maze, respectively, remained normal in 3-25-month-old CTC and CTO mice, in contrast to previously observed impairments in ACL mice expressing equivalent levels of hCasp6 only. Consistently, the CTC and CTO hippocampal CA1 region displayed equivalent dendritic spine density and no glial inflammation. Together, these results reveal that active hCasp6 co-expression with hTau generates Tau cleavage and rare age-dependent argentophilic deposits but fails to induce cognitive deficits, neuroinflammation, and Tau pathology.


Assuntos
Doença de Alzheimer/enzimologia , Comportamento Animal , Encéfalo/enzimologia , Caspase 6/metabolismo , Cognição , Disfunção Cognitiva/enzimologia , Degeneração Neural , Neuroglia/enzimologia , Neurônios/enzimologia , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Animais , Encéfalo/patologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Caspase 6/genética , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Disfunção Cognitiva/psicologia , Modelos Animais de Doenças , Locomoção , Memória , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Emaranhados Neurofibrilares/enzimologia , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/patologia , Neuroglia/patologia , Neurônios/patologia , Teste de Campo Aberto , Fosforilação , Agregados Proteicos , Agregação Patológica de Proteínas , Proteínas tau/genética
4.
Nat Commun ; 11(1): 4571, 2020 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-32917871

RESUMO

Early therapeutic interventions are essential to prevent Alzheimer Disease (AD). The association of several inflammation-related genetic markers with AD and the early activation of pro-inflammatory pathways in AD suggest inflammation as a plausible therapeutic target. Inflammatory Caspase-1 has a significant impact on AD-like pathophysiology and Caspase-1 inhibitor, VX-765, reverses cognitive deficits in AD mouse models. Here, a one-month pre-symptomatic treatment of Swedish/Indiana mutant amyloid precursor protein (APPSw/Ind) J20 and wild-type mice with VX-765 delays both APPSw/Ind- and age-induced episodic and spatial memory deficits. VX-765 delays inflammation without considerably affecting soluble and aggregated amyloid beta peptide (Aß) levels. Episodic memory scores correlate negatively with microglial activation. These results suggest that Caspase-1-mediated inflammation occurs early in the disease and raise hope that VX-765, a previously Food and Drug Administration-approved drug for human CNS clinical trials, may be a useful drug to prevent the onset of cognitive deficits and brain inflammation in AD.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Disfunção Cognitiva/metabolismo , Serpinas/metabolismo , Proteínas Virais/metabolismo , Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Animais , Comportamento Animal , Disfunção Cognitiva/tratamento farmacológico , Citocinas/metabolismo , Dipeptídeos/sangue , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Encefalite/metabolismo , Encefalite/patologia , Feminino , Humanos , Inflamação/metabolismo , Masculino , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Serpinas/sangue , Serpinas/farmacologia , Memória Espacial/fisiologia , Proteínas Virais/sangue , Proteínas Virais/farmacologia , para-Aminobenzoatos/sangue , para-Aminobenzoatos/farmacologia
5.
Acta Neuropathol Commun ; 7(1): 210, 2019 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-31843022

RESUMO

Activated Caspase-6 (Casp6) is associated with age-dependent cognitive impairment and Alzheimer disease (AD). Mice expressing human Caspase-6 in hippocampal CA1 neurons develop age-dependent cognitive deficits, neurodegeneration and neuroinflammation. This study assessed if methylene blue (MB), a phenothiazine that inhibits caspases, alters Caspase-6-induced neurodegeneration and cognitive impairment in mice. Aged cognitively impaired Casp6-overexpressing mice were treated with methylene blue in drinking water for 1 month. Methylene blue treatment did not alter Caspase-6 levels, assessed by RT-PCR, western blot and immunohistochemistry, but inhibited fluorescently-labelled Caspase-6 activity in acute brain slice intact neurons. Methylene blue treatment rescued Caspase-6-induced episodic and spatial memory deficits measured by novel object recognition and Barnes maze, respectively. Methylene blue improved synaptic function of hippocampal CA1 neurons since theta-burst long-term potentiation (LTP), measured by field excitatory postsynaptic potentials (fEPSPs) in acute brain slices, was successfully induced in the Schaffer collateral-CA1 pathway in methylene blue-treated, but not in vehicle-treated, Caspase-6 mice. Increased neuroinflammation, measured by ionized calcium binding adaptor molecule 1 (Iba1)-positive microglia numbers and subtypes, and glial fibrillary acidic protein (GFAP)-positive astrocytes, were decreased by methylene blue treatment. Therefore, methylene blue reverses Caspase-6-induced cognitive deficits by inhibiting Caspase-6, and Caspase-6-mediated neurodegeneration and neuroinflammation. Our results indicate that Caspase-6-mediated damage is reversible months after the onset of cognitive deficits and suggest that methylene blue could benefit Alzheimer disease patients by reversing Caspase-6-mediated cognitive decline.


Assuntos
Envelhecimento/metabolismo , Caspase 6/metabolismo , Inibidores de Caspase/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/enzimologia , Azul de Metileno/uso terapêutico , Envelhecimento/efeitos dos fármacos , Envelhecimento/patologia , Animais , Inibidores de Caspase/farmacologia , Disfunção Cognitiva/patologia , Feminino , Humanos , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Inflamação/patologia , Masculino , Azul de Metileno/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
6.
Nat Commun ; 9(1): 3916, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30254377

RESUMO

Alzheimer's disease (AD) is an intractable progressive neurodegenerative disease characterized by cognitive decline and dementia. An inflammatory neurodegenerative pathway, involving Caspase-1 activation, is associated with human age-dependent cognitive impairment and several classical AD brain pathologies. Here, we show that the nontoxic and blood-brain barrier permeable small molecule Caspase-1 inhibitor VX-765 dose-dependently reverses episodic and spatial memory impairment, and hyperactivity in the J20 mouse model of AD. Cessation of VX-765 results in the reappearance of memory deficits in the mice after 1 month and recommencement of treatment re-establishes normal cognition. VX-765 prevents progressive amyloid beta peptide deposition, reverses brain inflammation, and normalizes synaptophysin protein levels in mouse hippocampus. Consistent with these findings, Caspase-1 null J20 mice are protected from episodic and spatial memory deficits, neuroinflammation and Aß accumulation. These results provide in vivo proof of concept for Caspase-1 inhibition against AD cognitive deficits and pathologies.


Assuntos
Doença de Alzheimer/prevenção & controle , Caspase 1/metabolismo , Inibidores de Caspase/farmacologia , Transtornos Cognitivos/prevenção & controle , Modelos Animais de Doenças , Transtornos da Memória/prevenção & controle , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Caspase 1/genética , Cognição/efeitos dos fármacos , Cognição/fisiologia , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/fisiopatologia , Dipeptídeos/farmacologia , Humanos , Memória/efeitos dos fármacos , Memória/fisiologia , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos Knockout , para-Aminobenzoatos/farmacologia
7.
Cell Death Differ ; 25(7): 1319-1335, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29352267

RESUMO

Active cysteinyl protease Caspase-6 is associated with early Alzheimer and Huntington diseases. Higher entorhinal cortex and hippocampal Caspase-6 levels correlate with lower cognitive performance in aged humans. Caspase-6 induces axonal degeneration in human primary neuron cultures and causes inflammation and neurodegeneration in mouse hippocampus, and age-dependent memory impairment. To assess whether Caspase-6 causes damage to another neuronal system, a transgenic knock-in mouse overexpressing a self-activated form of Caspase-6 five-fold in the striatum, the area affected in Huntington disease, and 2.5-fold in the hippocampus and cortex, was generated. Detection of Tubulin cleaved by Caspase-6 confirmed Caspase-6 activity. The Caspase-6 expressing mice and control littermates were subjected to behavioral tests to assess Huntington disease-relevant psychiatric, motor, and cognitive deficits. Depression was excluded with the forced swim and sucrose consumption tests. Motor deficits were absent in the nesting, clasping, rotarod, vertical pole, gait, and open field analyzes. However, Caspase-6 mice developed age-dependent episodic and spatial memory deficits identified by novel object recognition, Barnes maze and Morris water maze assays. Neuron numbers were maintained in the striatum, hippocampus, and cortex. Microglia and astrocytes were increased in the hippocampal stratum lacunosum molecular and in the cortex, but not in the striatum. Synaptic mRNA profiling identified two differentially expressed genes in transgenic hippocampus, but none in striatum. Caspase-6 impaired synaptic transmission and induced neurodegeneration in hippocampal CA1 neurons, but not in striatal medium spiny neurons. These data revealed that active Caspase-6 in the striatal medium spiny neurons failed to induce inflammation, neurodegeneration or behavioral abnormalities, whereas active Caspase-6 in the cortex and hippocampus impaired episodic and spatial memories, and induced inflammation, neuronal dysfunction, and neurodegeneration. The results indicate age and neuronal subtype-dependent Caspase-6 toxicity and highlight the importance of targeting the correct neuronal subtype to identify underlying molecular mechanisms of neurodegenerative diseases.


Assuntos
Caspase 6/metabolismo , Córtex Cerebral/enzimologia , Corpo Estriado/enzimologia , Hipocampo/enzimologia , Doença de Huntington/enzimologia , Transtornos da Memória/enzimologia , Neurônios/enzimologia , Animais , Caspase 6/genética , Córtex Cerebral/patologia , Corpo Estriado/patologia , Hipocampo/patologia , Humanos , Doença de Huntington/patologia , Transtornos da Memória/genética , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Neurônios/patologia
8.
Biochimie ; 137: 158-164, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28365364

RESUMO

Sphingolipid-related issues are increasingly discussed to contribute to the neuropathological process of Alzheimer's disease (AD). In this study, gangliosides and related-sphingolipids (ceramides, neutral glycosphingolipids and sphingomyelins) were analyzed in neuroglioma (H4) cells expressing the Swedish mutation of the human amyloid precursor protein (H4APPsw) and compared with those of wild-type control H4 cells. These cells were chosen since H4APPsw cells were previously reported to reproduce well some essential features of AD. We found that H4APPsw cells exhibited a striking elevation of the simplest ganglioside GM3, an abnormality that was consistently reported in AD patients and animal models of AD. Concomitantly, the levels of both lactosylceramide (the immediate metabolic precursor of GM3) and ganglioside GD1a increased, suggesting a deregulation in the biosynthesis of gangliosides in the H4APPsw cells. Moreover, while the total ceramide level remained unaltered in H4APPsw cells, a shift in ceramide composition from long chain - to very long chain fatty acid-ceramide species was recorded. Because sphingolipid alterations occurring in H4APPsw cells were similar to those observed in transgenic mice and in human brains, this cellular model might be useful to further explore the complex role of sphingolipids in AD pathogenesis.


Assuntos
Doença de Alzheimer/patologia , Neoplasias Encefálicas/patologia , Gangliosídeos/metabolismo , Glioma/patologia , Esfingolipídeos/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Ceramidas/metabolismo , Glioma/metabolismo , Humanos , Camundongos , Mutação/genética
9.
Mol Neurodegener ; 12(1): 22, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28241839

RESUMO

BACKGROUND: The activation of the aspartate-specific cysteinyl protease, Caspase-6, is proposed as an early pathogenic event of Alzheimer disease (AD) and Huntington's disease. Caspase-6 inhibitors could be useful against these neurodegenerative diseases but most Caspase-6 inhibitors have been exclusively studied in vitro or show acute liver toxicity in humans. Here, we assessed vinyl sulfone small molecule peptide caspase inhibitors for potential use in vivo. METHODS: The IC50 of NWL vinyl sulfone small molecule caspase inhibitors were determined on Caspase-1 to 10, and Caspase-6-transfected human colon carcinoma HCT116 cells. Inhibition of Caspase-6-mediated axonal degeneration was assessed in serum-deprived or amyloid precursor protein-transfected primary human CNS neurons. Cellular toxicity was measured by phase contrast microscopy, mitochondrial and lactate dehydrogenase colorimetric activity assays, or flow cytometry. Caspase inhibition was measured by fluorogenic activity assays, fluorescence microscopy, and western blot analyses. The effect of inhibitors on age-dependent cognitive deficits in Caspase-6 transgenic mice was assessed by the novel object recognition task. Liquid chromatography coupled to tandem mass spectrometry assessed the blood-brain barrier permeability of inhibitors in Caspase-6 mice. RESULTS: Vinyl sulfone NWL-117 caspase inhibitor has a higher selectivity against Caspase-6, -4, -8, -9, and -10 whereas NWL-154 has higher selectivity against Caspase-6, -8, and -10. The half-maximal inhibitory concentrations (IC50) of NWL-117 and NWL-154 is 192 nM and 100 nM against Caspase-6 in vitro, and 4.82 µM and 3.63 µM in Caspase-6-transfected HCT116 cells, respectively. NWL inhibitors are not toxic to HCT116 cells or to human primary neurons. NWL-117 and NWL-154 inhibit serum deprivation-induced Caspase-6 activity and prevent amyloid precursor protein-mediated neurite degeneration in human primary CNS neurons. NWL-117 crosses the blood brain barrier and reverses age-dependent episodic memory deficits in Caspase-6 mice. CONCLUSIONS: NWL peptidic vinyl methyl sulfone inhibitors are potent, non-toxic, blood-brain barrier permeable, and irreversible caspase inhibitors with neuroprotective effects in HCT116 cells, in primary human CNS neurons, and in Caspase-6 mice. These results highlight the therapeutic potential of vinyl sulfone inhibitors as caspase inhibitors against neurodegenerative diseases and sanction additional work to improve their selectivity against different caspases.


Assuntos
Caspase 6/efeitos dos fármacos , Inibidores de Caspase/farmacologia , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Sulfonas/farmacologia , Animais , Axônios/efeitos dos fármacos , Axônios/enzimologia , Western Blotting , Cromatografia Líquida , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Microscopia de Contraste de Fase , Neurônios/enzimologia , Espectrometria de Massas em Tandem
10.
Mol Cell Neurosci ; 75: 63-70, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27373967

RESUMO

Evidence has suggested that ganglioside abnormalities may be linked to the proteolytic processing of amyloid precursor protein (APP) in Alzheimer's disease (AD) and that pharmacological inhibition of ganglioside synthesis may reduce amyloid ß-peptide (Aß) production. In this study, we assessed the usefulness of two well-established glycosphingolipid (GSL) synthesis inhibitors, the synthetic ceramide analog D-PDMP (1-phenyl 2-decanoylamino-3-morpholino-1-propanol) and the iminosugar N-butyldeoxynojirimycin (NB-DNJ or miglustat), as anti-amyloidogenic drugs in a human cellular model of AD. We found that both GSL inhibitors were able to markedly inhibit Aß production, although affecting differently the APP cleavage. Surprisingly, the L-enantiomer of PDMP, which promotes ganglioside accumulation, acted similarly to D-PDMP to inhibit Aß production. Concurrently, both D- and L-PDMP strongly and equally reduced the levels of long-chain ceramides. Altogether, our data suggested that the anti-amyloidogenic effects of PDMP agents are independent of the altered cellular ganglioside composition, but may result, at least in part, from their ability to reduce ceramide levels. Moreover, our current study established for the first time that NB-DNJ, a drug already used as a therapeutic for Gaucher disease (a lysosomal storage disorder), was also able to reduce Aß production in our cellular model. Therefore, our study provides novel information regarding the possibilities to target amyloidogenic processing of APP through modulation of sphingolipid metabolism and emphasizes the potential of the iminosugar NB-DNJ as a disease modifying therapy for AD.


Assuntos
1-Desoxinojirimicina/análogos & derivados , Peptídeos beta-Amiloides/metabolismo , Inibidores Enzimáticos/farmacologia , Gangliosídeos/biossíntese , Morfolinas/farmacologia , 1-Desoxinojirimicina/farmacologia , Linhagem Celular Tumoral , Gangliosídeos/metabolismo , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo
11.
Life Sci ; 158: 14-21, 2016 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-27343974

RESUMO

AIMS: GSK3ß activation in Aß conditions leading to tau phosphorylation at pathological sites is a well-known phenomenon. However, the serine/tyrosine phosphorylation processes implied in Aß-induced GSK3ß activation and responsible for tau phosphorylation, especially at the GSK3ß specific Ser396/Ser404 (PHF-1) site, are still debated. MAIN METHODS: Experiments were performed on SH-SY5Y cells exposed to 20µM Aß1-42 in a time ranging from 5min to 8h. The phophorylated forms (Ser9 and Tyr216) of GSK3ß and pTau at PHF-1 epitope were measured by immunoblotting in nuclear extracts. KEY FINDINGS: We showed a superimposable time-dependent increase of nuclear pGSK3ßTyr216 and nuclear pTau at PHF-1 site, both reaching their maximal level after 8h of Aß1-42 exposure. In addition, nuclear accumulation of pTau is accompanied by its cytoplasmic decrease suggesting that pTau is translocated in response to Aß treatment. Besides, our experiments showed that specific pGSK3ßTyr216 inhibition is required to drop nuclear pTau, ensuring the involvement of Tyr216 phosphorylation in Aß-mediated tau phosphorylation at PHF-1 epitope. SIGNIFICANCE: These data suggested that in response to Aß exposure in SH-SY5Y cells, GSK3ß activation is performed through Tyr216 phosphorylation and resulted in tau phosphorylation at PHF-1 epitope and in its translocation.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Anticorpos Monoclonais/imunologia , Núcleo Celular/metabolismo , Epitopos/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Tirosina/metabolismo , Proteínas tau/metabolismo , Linhagem Celular Tumoral , Epitopos/imunologia , Humanos , Fosforilação
12.
Am J Pathol ; 186(3): 552-67, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26857506

RESUMO

Mild traumatic brain injury (mTBI) is an emerging risk for chronic behavioral, cognitive, and neurodegenerative conditions. Athletes absorb several hundred mTBIs each year; however, rodent models of repeat mTBI (rmTBI) are often limited to impacts in the single digits. Herein, we describe the effects of 30 rmTBIs, examining structural and pathological changes in mice up to 365 days after injury. We found that single mTBI causes a brief loss of consciousness and a transient reduction in dendritic spines, reflecting a loss of excitatory synapses. Single mTBI does not cause axonal injury, neuroinflammation, or cell death in the gray or white matter. Thirty rmTBIs with a 1-day interval between each mTBI do not cause dendritic spine loss; however, when the interinjury interval is increased to 7 days, dendritic spine loss is reinstated. Thirty rmTBIs cause white matter pathology characterized by positive silver and Fluoro-Jade B staining, and microglial proliferation and activation. This pathology continues to develop through 60 days, and is still apparent at 365 days, after injury. However, rmTBIs did not increase ß-amyloid levels or tau phosphorylation in the 3xTg-AD mouse model of Alzheimer disease. Our data reveal that single mTBI causes a transient loss of synapses, but that rmTBIs habituate to repetitive injury within a short time period. rmTBI causes the development of progressive white matter pathology that continues for months after the final impact.


Assuntos
Lesões Encefálicas/patologia , Espinhas Dendríticas/patologia , Substância Branca/patologia , Amiloide/metabolismo , Animais , Comportamento Animal , Concussão Encefálica/patologia , Lesões Encefálicas/metabolismo , Traumatismos Craniocerebrais/metabolismo , Traumatismos Craniocerebrais/patologia , Espinhas Dendríticas/metabolismo , Modelos Animais de Doenças , Fluoresceínas , Complexo de Golgi , Humanos , Inflamação , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Recidiva , Inconsciência , Substância Branca/metabolismo , Proteínas tau/metabolismo
13.
Exp Neurol ; 271: 308-18, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26115843

RESUMO

Accumulating evidence implicates ganglioside and/or related-sphingolipid disturbance in the pathogenesis of Alzheimer's disease (AD). However, it is not known whether these lipidic alterations are connected with other important features of AD, such as deregulated insulin/Akt/GSK3 signaling. In this study, we have treated neuroglioma cells expressing the double Swedish mutation of human amyloid precursor protein (H4APPsw) with several glycosphingolipid (GSL)-modulating agents, and we have analyzed the impact of the aberrant ganglioside composition on the GSK3 activation state. We found that both ceramide analogs D- and L-PDMP (1-phenyl 2-decanoylamino-3-morpholino-1-propanol), which have opposite effects on ganglioside synthesis, selectively inhibited GSK3ß via Ser9 phosphorylation independently of the upstream insulin/Akt pathway. Conversely, the iminosugar N-butyldeoxynojirimycin (NB-DNJ) which displayed similar reduction of gangliosides as D-PDMP, did not affect the phosphorylation state of GSK3ß. Concurrently, while NB-DNJ did not modify the cellular ceramide content, both PDMP enantiomers strongly and equally reduced the levels of long-chain ceramide species. Altogether, our findings led us to hypothesize that the PDMP-induced altered ganglioside composition is not the principal mechanism involved in the inhibition of GSK3ß, but seems to implicate, at least in part, their ability to reduce ceramide levels. Nevertheless, this study provides new information regarding the possibilities to target GSK3ß through modulation of sphingolipid metabolism.


Assuntos
Gangliosídeos/metabolismo , Regulação Enzimológica da Expressão Gênica/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Esfingolipídeos/metabolismo , Precursor de Proteína beta-Amiloide/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glioma/patologia , Glicogênio Sintase Quinase 3 beta , Humanos , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Morfolinas/farmacologia , Mutação/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
14.
Neurobiol Aging ; 36(8): 2414-28, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26058840

RESUMO

There is developing interest in the potential association between anesthesia and the onset and progression of Alzheimer's disease. Several anesthetics have, thus, been demonstrated to induce tau hyperphosphorylation, an effect mostly mediated by anesthesia-induced hypothermia. Here, we tested the hypothesis that acute normothermic administration of dexmedetomidine (Dex), an intravenous sedative used in intensive care units, would result in tau hyperphosphorylation in vivo and in vitro. When administered to nontransgenic mice, Dex-induced tau hyperphosphorylation persisting up to 6 hours in the hippocampus for the AT8 epitope. Pretreatment with atipamezole, a highly specific α2-adrenergic receptor antagonist, blocked Dex-induced tau hyperphosphorylation. Furthermore, Dex dose-dependently increased tau phosphorylation at AT8 in SH-SY5Y cells, impaired mice spatial memory in the Barnes maze and promoted tau hyperphosphorylation and aggregation in transgenic hTau mice. These findings suggest that Dex: (1) increases tau phosphorylation, in vivo and in vitro, in the absence of anesthetic-induced hypothermia and through α2-adrenergic receptor activation, (2) promotes tau aggregation in a mouse model of tauopathy, and (3) impacts spatial reference memory.


Assuntos
Dexmedetomidina/efeitos adversos , Hipnóticos e Sedativos/efeitos adversos , Proteínas tau/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Células Cultivadas , Dexmedetomidina/administração & dosagem , Dexmedetomidina/antagonistas & inibidores , Relação Dose-Resposta a Droga , Hipocampo/metabolismo , Humanos , Hipnóticos e Sedativos/administração & dosagem , Hipotermia Induzida , Técnicas In Vitro , Infusões Intravenosas , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Agregação Patológica de Proteínas/induzido quimicamente , Memória Espacial/efeitos dos fármacos
15.
Hum Mol Genet ; 24(1): 86-99, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25205109

RESUMO

Huntington's disease (HD) is an autosomal-dominant neurodegenerative disorder caused by polyglutamine expansions in the amino-terminal region of the huntingtin (Htt) protein. At the cellular level, neuronal death is accompanied by the proteolytic cleavage, misfolding and aggregation of huntingtin. Abnormal hyperphosphorylation of tau protein is a characteristic feature of a class of neurodegenerative diseases called tauopathies. As a number of studies have reported tau pathology in HD patients, we investigated whether HD pathology may promote tau hyperphosphorylation and if so tackle some of its underlying mechanisms. For that purpose, we used the R6/2 mouse, a well-characterized model of HD, and analyzed tau phosphorylation before and after the onset of HD-like symptoms. We found a significant increase in tau hyperphosphorylation at the PHF-1 epitope in pre-symptomatic R6/2 mice, whereas symptomatic mice displayed tau hyperphosphorylation at multiple tau phosphoepitopes (AT8, CP13, PT205 and PHF-1). There was no activation of major tau kinases that could explain this observation. However, when we examined tau phosphatases, we found that calcineurin/PP2B was downregulated by 30% in pre-symptomatic and 50% in symptomatic R6/2 mice, respectively. We observed similar changes in tau phosphorylation and calcineurin expression in Q175 mice, another HD model. Calcineurin was also reduced in Q111 compared with Q7 cells. Finally, pharmacological or genetic inhibition of endogenous calcineurin was sufficient to promote tau hyperphosphorylation in neuronal cells. Taken together, our data suggest that mutant huntingtin can induce abnormal tau hyperphosphorylation in vivo, via the deregulation of calcineurin.


Assuntos
Encéfalo/citologia , Calcineurina/metabolismo , Doença de Huntington/metabolismo , Neurônios/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas tau/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Doença de Huntington/genética , Camundongos , Camundongos Transgênicos , Fosforilação
16.
Neurobiol Aging ; 36(2): 901-2, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25491074

RESUMO

Alzheimer's disease is characterized by the deposition of intracellular aggregates of hyperphosphorylated tau protein. Tau hyperphosphorylation has been attributed in part to the deregulation of kinases and phosphatases activities. Extracellular signal regulated-kinases 1/2 (ERK1/2) were reported to be activated in the first stages of Alzheimer's disease and were proposed as a potential therapeutic target. However, although the phosphorylation of tau by ERK1/2 has been demonstrated in cell-free system, it remains controversial in vivo. Here, we showed that pharmacologic inhibition of ERK1/2 in mice and SH-SY5Y cells did not reduce basal levels of phospho-tau or hypothermia-induced tau hyperphosphorylation. We also found that activating ERK1/2 by hyperthermia did not correlate with increased tau phosphorylation. Finally, ERK1/2 was inhibited, but tau phosphorylation was not altered in Mek1-/- mice. In conclusion, these results do not support the involvement of ERK1/2 in tau phosphorylation under physiological conditions.


Assuntos
Doença de Alzheimer/etiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Proteínas tau/metabolismo , Doença de Alzheimer/terapia , Animais , Células Cultivadas , Humanos , Hipertermia Induzida , Hipotermia Induzida , Técnicas In Vitro , Camundongos Endogâmicos C57BL , Camundongos Knockout , Terapia de Alvo Molecular , Fosforilação
17.
J Neurosci Res ; 89(5): 755-63, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21360572

RESUMO

Glycogen synthase kinase 3ß (GSK3ß) activity is regulated by phosphorylation processes and regulates in turn through phosphorylation several proteins, including eukaryotic initiation factor 2B (eIF2B). Serine 9 phosphorylation of GSK3ß (pGSK3ßSer9), usually promoted by activation of the PI3K/Akt survival pathway, triggers GSK3ß inhibition. By contrast, tyrosine 216 phosphorylation of GSK3ß (pGSK3ßTyr216) increases under apoptotic conditions, leading to GSK3ß activation. Lithium chloride (LiCl) is usually described to increase pGSK3ßSer9 through the PI3K/Akt pathway, resulting in GSK3ß inhibition. The purpose of this study is to demonstrate that in some cases LiCl is also able to increase pGSK3ßTyr216, resulting in GSK3ß activation. For this, we used SH-SY5Y cells and primary neuronal cultures and investigated the effects of LiCl on the two phosphorylated forms of GSK3ß under staurosporine (STS)-intoxicated conditions. The ratios between the phosphorylated and total forms of GSK3ß and eIF2B were determined by Western blotting. Our results revealed that, besides its ability to increase pGSK3ßSer9, LiCl is also able to increase pGSK3ßTyr216 greatly in STS-intoxicated SH-SY5Y cells but not in STS-intoxicated primary neuronal cultures. This accumulation of both Ser9 and Tyr216 phosphorylation results in GSK3ß activation in STS-intoxicated SH-SY5Y cells in spite of the presence of LiCl. These findings indicate that LiCl treatment is not necessarily correlated with GSK3ß inhibition even though it generates Ser9 phosphorylation. Consequently, the ratio pGSK3ßSer9/pGSK3ßTyr216, which takes into account the balance between the two inactive (Ser9) and active (Tyr216) forms of GSK3ß, could be more useful for predicting GSK3ß inhibition.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Cloreto de Lítio/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Estaurosporina/farmacologia , Antimaníacos/farmacologia , Linhagem Celular Tumoral , Células Cultivadas , Glicogênio Sintase Quinase 3 beta , Humanos , Neuroblastoma/enzimologia , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia
18.
Int J Alzheimers Dis ; 2011: 920958, 2010 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-21234372

RESUMO

There is evidence linking sphingolipid abnormalities, APP processing, and neuronal death in Alzheimer's disease (AD). We previously reported a strong elevation of ceramide levels in the brain of the APP(SL)/PS1Ki mouse model of AD, preceding the neuronal death. To extend these findings, we analyzed ceramide and related-sphingolipid contents in brain from two other mouse models (i.e., APP(SL) and APP(SL)/PS1(M146L)) in which the time-course of pathology is closer to that seen in most currently available models. Conversely to our previous work, ceramides did not accumulate in disease-associated brain regions (cortex and hippocampus) from both models. However, the APP(SL)/PS1Ki model is unique for its drastic neuronal loss coinciding with strong accumulation of neurotoxic Aß isoforms, not observed in other animal models of AD. Since there are neither neuronal loss nor toxic Aß species accumulation in APP(SL) mice, we hypothesized that it might explain the lack of ceramide accumulation, at least in this model.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...