Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Alzheimers Dis ; 62(1): 213-226, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29439327

RESUMO

BACKGROUND: With current treatments for Alzheimer's disease (AD) only providing temporary symptomatic benefits, disease modifying drugs are urgently required. This approach relies on improved understanding of the early pathophysiology of AD. A new hypothesis has emerged, in which early memory loss is considered a synapse failure caused by soluble amyloid-ß oligomers (Aßo). These small soluble Aßo, which precede the formation of larger fibrillar assemblies, may be the main cause of early AD pathologies. OBJECTIVE: The aim of the current study was to investigate the effect of acute administration of stabilized low-n amyloid-ß1-42 oligomers (Aßo1-42) on cognitive, inflammatory, synaptic, and neuronal markers in the rat. METHODS: Female and male Lister Hooded rats received acute intracerebroventricular (ICV) administration of either vehicle or 5 nmol of Aßo1-42 (10µL). Cognition was assessed in the novel object recognition (NOR) paradigm at different time points. Levels of inflammatory (IL-1ß, IL-6, TNF-α), synaptic (PSD-95, SNAP-25), and neuronal (n-acetylaspartate, parvalbumin-positive cells) markers were investigated in different brain regions (prefrontal and frontal cortex, striatum, dorsal and ventral hippocampus). RESULTS: Acute ICV administration of Aßo1-42 induced robust and enduring NOR deficits. These deficits were reversed by acute administration of donepezil and rolipram but not risperidone. Postmortem analysis revealed an increase in inflammatory markers, a decrease in synaptic markers and parvalbumin containing interneurons in the frontal cortex, with no evidence of widespread neuronal loss. CONCLUSION: Taken together the results suggest that acute administration of soluble low-n Aßo may be a useful model to study the early mechanisms involved in AD and provide us with a platform for testing novel therapeutic approaches that target the early underlying synaptic pathology.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Cognição/fisiologia , Inflamação/metabolismo , Transtornos da Memória/metabolismo , Neurônios/metabolismo , Fragmentos de Peptídeos/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Donepezila/farmacologia , Feminino , Inflamação/tratamento farmacológico , Inflamação/patologia , Masculino , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Nootrópicos/farmacologia , Distribuição Aleatória , Ratos , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Risperidona/farmacologia , Rolipram/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia
2.
Nat Commun ; 7: 12504, 2016 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-27509875

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit cyclooxygenase-1 (COX-1) and COX-2 enzymes. The NLRP3 inflammasome is a multi-protein complex responsible for the processing of the proinflammatory cytokine interleukin-1ß and is implicated in many inflammatory diseases. Here we show that several clinically approved and widely used NSAIDs of the fenamate class are effective and selective inhibitors of the NLRP3 inflammasome via inhibition of the volume-regulated anion channel in macrophages, independently of COX enzymes. Flufenamic acid and mefenamic acid are efficacious in NLRP3-dependent rodent models of inflammation in air pouch and peritoneum. We also show therapeutic effects of fenamates using a model of amyloid beta induced memory loss and a transgenic mouse model of Alzheimer's disease. These data suggest that fenamate NSAIDs could be repurposed as NLRP3 inflammasome inhibitors and Alzheimer's disease therapeutics.


Assuntos
Doença de Alzheimer/prevenção & controle , Anti-Inflamatórios não Esteroides/farmacologia , Ácido Flufenâmico/farmacologia , Inflamassomos/metabolismo , Ácido Mefenâmico/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doença de Alzheimer/metabolismo , Animais , Células da Medula Óssea/metabolismo , Morte Celular , Canais de Cloreto/metabolismo , Cisteína/metabolismo , Feminino , Genótipo , Inflamação , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Transtornos da Memória/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Reconhecimento Visual de Modelos/efeitos dos fármacos , Ratos
3.
FASEB J ; 29(9): 3713-25, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26018677

RESUMO

Deficiency in the methyl donors vitamin B12 and folate during pregnancy and postnatal life impairs proper brain development. We studied the consequences of this combined deficiency on cerebellum plasticity in offspring from rat mothers subjected to deficient diet during gestation and lactation and in rat neuroprogenitor cells expressing cerebellum markers. The major proteomic change in cerebellum of 21-d-old deprived females was a 2.2-fold lower expression of synapsins, which was confirmed in neuroprogenitors cultivated in the deficient condition. A pathway analysis suggested that these proteomic changes were related to estrogen receptor α (ER-α)/Src tyrosine kinase. The influence of impaired ER-α pathway was confirmed by abnormal negative geotaxis test at d 19-20 and decreased phsophorylation of synapsins in deprived females treated by ER-α antagonist 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride (MPP). This effect was consistent with 2-fold decreased expression and methylation of ER-α and subsequent decreased ER-α/PPAR-γ coactivator 1 α (PGC-1α) interaction in deficiency condition. The impaired ER-α pathway led to decreased expression of synapsins through 2-fold decreased EGR-1/Zif-268 transcription factor and to 1.7-fold reduced Src-dependent phosphorylation of synapsins. The treatment of neuroprogenitors with either MPP or PP1 (4-(4'-phenoxyanilino)-6,7-dimethoxyquinazoline, 6,7-dimethoxy-N-(4-phenoxyphenyl)-4-quinazolinamine, SKI-1, Src-l1) Src inhibitor produced similar effects. In conclusion, the deficiency during pregnancy and lactation impairs the expression of synapsins through a deregulation of ER-α pathway.


Assuntos
Encéfalo/metabolismo , Receptor alfa de Estrogênio/metabolismo , Deficiência de Ácido Fólico , Regulação da Expressão Gênica no Desenvolvimento , Lactação , Sinapsinas/biossíntese , Deficiência de Vitamina B 12 , Animais , Encéfalo/embriologia , Encéfalo/patologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/antagonistas & inibidores , Feminino , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , PPAR gama/metabolismo , Gravidez , Ratos
4.
FASEB J ; 27(6): 2185-97, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23401564

RESUMO

We examined the gastric mucosa structure and inflammatory status in control well-nourished Wistar dams and in Wistar dams deprived of choline, folate, and vitamin B12 during gestation and suckling periods, and in their offspring just before birth and at weaning. In this model of methyl donor deficiency (MDD), structural protein (E-cadherin and actin) N-homocysteinylation was measured through immunoprecipitation and proximity ligation assays. Cellular stress, inflammation, and apoptosis were estimated by the analysis of the NF-κB pathway, and the expression of superoxide dismutase, cyclooxygenase-2, tumor necrosis factor α, caspases 3 and 9, and TUNEL assay. Aberrant gastric mucosa formation and signs of surface layer erosion were detected in MDD fetuses and weanlings. E-cadherin and actin were N-homocysteinylated (+215 and +249% vs. controls, respectively; P<0.001). Expression of ß-catenin staining drastically decreased (-98%; P<0.01). NF-κB pathway was activated (+124%; P<0.01). Expressions of all inflammatory factors (+70%; P<0.01), superoxide dismutase (+55%; P<0.01), and caspases (+104%; P<0.01) were markedly increased. These changes were also observed in dams, to a lesser extent. Early MDD induced gastric mucosa injury similar to atrophic gastritis through structural protein N-homocysteinylation, marked inflammation, and apoptosis, despite activation of repair machinery.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Gastrite/etiologia , Gastrite/patologia , Homocisteína/metabolismo , Inflamação/etiologia , Inflamação/patologia , Estresse Oxidativo/fisiologia , Animais , Animais Recém-Nascidos , Animais Lactentes , Caderinas/metabolismo , Feminino , Feto , Gastrite/metabolismo , Inflamação/metabolismo , Metilação , Mães , Gravidez , Ratos , Ratos Wistar , Índice de Gravidade de Doença , Transdução de Sinais/fisiologia
5.
PLoS One ; 7(11): e48828, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23173039

RESUMO

Whereas brief acute or intermittent episodes of hypoxia have been shown to exert a protective role in the central nervous system and to stimulate neurogenesis, other studies suggest that early hypoxia may constitute a risk factor that influences the future development of mental disorders. We therefore investigated the effects of a neonatal "conditioning-like" hypoxia (100% N2, 5 min) on the brain and the cognitive outcomes of rats until 720 days of age (physiologic senescence). We confirmed that such a short hypoxia led to brain neurogenesis within the ensuing weeks, along with reduced apoptosis in the hippocampus involving activation of Erk1/2 and repression of p38 and death-associated protein (DAP) kinase. At 21 days of age, increased thicknesses and cell densities were recorded in various subregions, with strong synapsin activation. During aging, previous exposure to neonatal hypoxia was associated with enhanced memory retrieval scores specifically in males, better preservation of their brain integrity than controls, reduced age-related apoptosis, larger hippocampal cell layers, and higher expression of glutamatergic and GABAergic markers. These changes were accompanied with a marked expression of synapsin proteins, mainly of their phosphorylated active forms which constitute major players of synapse function and plasticity, and with increases of their key regulators, i.e. Erk1/2, the transcription factor EGR-1/Zif-268 and Src kinase. Moreover, the significantly higher interactions between PSD-95 scaffolding protein and NMDA receptors measured in the hippocampus of 720-day-old male animals strengthen the conclusion of increased synaptic functional activity and plasticity associated with neonatal hypoxia. Thus, early non-injurious hypoxia may trigger beneficial long term effects conferring higher resistance to senescence in aged male rats, with a better preservation of cognitive functions.


Assuntos
Envelhecimento/fisiologia , Encéfalo/fisiopatologia , Hipóxia/fisiopatologia , Envelhecimento/metabolismo , Animais , Animais Recém-Nascidos , Gasometria , Encéfalo/metabolismo , Encéfalo/patologia , Morte Celular , Proliferação de Células , Sobrevivência Celular , Feminino , Hipóxia/sangue , Hipóxia/metabolismo , Hipóxia/patologia , Locomoção/fisiologia , Masculino , Memória/fisiologia , Neurogênese , Plasticidade Neuronal/fisiologia , Ratos , Ratos Wistar , Sinapses/metabolismo
6.
J Neurochem ; 123(4): 467-76, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22909011

RESUMO

Brain lipid homeostasis is important for maintenance of brain cell function and synaptic communications, and is intimately linked to age-related cognitive decline. Because of the blood-brain barrier's limiting nature, this tissue relies on a complex system for the synthesis and receptor-mediated uptake of lipids between the different networks of neurons and glial cells. Using immunofluorescence, we describe the region-specific expression of the lipolysis-stimulated lipoprotein receptor (LSR), in the mouse hippocampus, cerebellum Purkinje cells, the ependymal cell interface between brain parenchyma and cerebrospinal fluid, and the choroid plexus. Colocalization with cell-specific markers revealed that LSR was expressed in neurons, but not astrocytes. Latency in arms of the Y-maze exhibited by young heterozygote LSR(+/-) mice was significantly different as compared to control LSR(+/+), and increased in older LSR(+/-) mice. Filipin and Nile red staining revealed membrane cholesterol content accumulation accompanied by significantly altered distribution of LSR in the membrane, and decreased intracellular lipid droplets in the cerebellum and hippocampus of old LSR(+/-) mice, as compared to control littermates as well as young LSR(+/-) animals. These data therefore suggest a potential role of LSR in brain cholesterol distribution, which is particularly important in preserving neuronal integrity and thereby cognitive functions during aging.


Assuntos
Envelhecimento/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Colesterol/metabolismo , Neurônios/metabolismo , Receptores de LDL/metabolismo , Fatores Etários , Animais , Plexo Corióideo/metabolismo , Filipina/metabolismo , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfopiruvato Hidratase/metabolismo , Receptores de LDL/deficiência
7.
FASEB J ; 26(10): 3980-92, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22713523

RESUMO

Despite the key role in neuronal development of a deficit in the methyl donor folate, little is known on the underlying mechanisms. We therefore studied the consequences of folate deficiency on proliferation, differentiation, and plasticity of the rat H19-7 hippocampal cell line. Folate deficit reduced proliferation (17%) and sensitized cells to differentiation-associated apoptosis (+16%). Decreased production (-58%) of S-adenosylmethionine (the universal substrate for transmethylation reactions) and increased expression of histone deacetylases (HDAC4,6,7) would lead to epigenomic changes that may impair the differentiation process. Cell polarity, vesicular transport, and synaptic plasticity were dramatically affected, with poor neurite outgrowth (-57%). Cell treatment by an HDAC inhibitor (SAHA) led to a noticeable improvement of cell polarity and morphology, with longer processes. Increased homocysteine levels (+55%) consecutive to folate shortage produced homocysteinylation, evidenced by coimmunoprecipitations and mass spectrometry, and aggregation of motor proteins dynein and kinesin, along with functional alterations, as reflected by reduced interactions with partner proteins. Prominent homocysteinylation of key neuronal proteins and subsequent aggregation certainly constitute major adverse effects of folate deficiency, affecting normal development with possible long-lasting consequences.


Assuntos
Deficiência de Ácido Fólico/metabolismo , Ácido Fólico/farmacologia , Hipocampo/citologia , Homocisteína/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Hep G2 , Humanos , Imuno-Histoquímica , Neurônios/metabolismo , Ligação Proteica , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Vitamina B 12/farmacologia
8.
Neurobiol Aging ; 33(6): 1123.e17-29, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22188721

RESUMO

Soluble beta-amyloid (Aß) oligomers are considered to putatively play a critical role in the early synapse loss and cognitive impairment observed in Alzheimer's disease. We previously demonstrated that Aß oligomers activate cytosolic phospholipase A(2) (cPLA(2)), which specifically releases arachidonic acid from membrane phospholipids. We here observed that cPLA(2) gene inactivation prevented the alterations of cognitive abilities and the reduction of hippocampal synaptic markers levels noticed upon a single intracerebroventricular injection of Aß oligomers in wild type mice. We further demonstrated that the Aß oligomer-induced sphingomyelinase activation was suppressed and that phosphorylation of Akt/protein kinase B (PKB) was preserved in neuronal cells isolated from cPLA(2)(-/-) mice. Interestingly, expression of the Aß precursor protein (APP) was reduced in hippocampus homogenates and neuronal cells from cPLA(2)(-/-) mice, but the relationship with the resistance of these mice to the Aß oligomer toxicity requires further investigation. These results therefore show that cPLA(2) plays a key role in the Aß oligomer-associated neurodegeneration, and as such represents a potential therapeutic target for the treatment of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Transtornos da Memória/enzimologia , Transtornos da Memória/genética , Doenças Neurodegenerativas/enzimologia , Fragmentos de Peptídeos/toxicidade , Fosfolipases A2 Citosólicas/fisiologia , Animais , Células Cultivadas , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/induzido quimicamente , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Doenças Neurodegenerativas/induzido quimicamente
9.
FASEB J ; 24(11): 4218-28, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20647547

RESUMO

As a hepatic receptor for triglyceride-rich lipoproteins, the lipolysis-stimulated lipoprotein receptor (LSR) may be involved in the dynamics of lipid distribution between the liver and peripheral tissues. Here, we explore the potential role of leptin in regulating LSR. At physiological concentrations (1-10 ng/ml), leptin increased LSR protein and mRNA levels in Hepa1-6 cells through an ERK1/2-dependent and α-amanitin-sensitive pathway. In vivo, leptin treatment of C57BL6/Rj mice (1 µg 2×/d, 8 d) led to a significant increase in hepatic LSR mRNA and protein, decreased liver triglycerides and increased VLDL secretion as compared to controls. LSR(+/-) mice with elevated postprandial lipemia placed on a high-fat (60% kcal) diet exhibited accelerated weight gain and increased fat mass as compared to controls. While plasma leptin levels were increased 3-fold, hepatic leptin receptor protein levels and phosphorylation of ERK1/2 were significantly reduced. Therefore, leptin is an important regulator of LSR protein levels providing the means for the control of hepatic uptake of lipids during the postprandial phase. However, this may no longer be functional in LSR(+/-) mice placed under a chronic dietary fat load, suggesting that this animal model could be useful for the study of molecular mechanisms involved in peripheral leptin resistance.


Assuntos
Leptina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipólise/efeitos dos fármacos , Fígado/efeitos dos fármacos , Período Pós-Prandial , Receptores de Lipoproteínas/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Western Blotting , Peso Corporal/efeitos dos fármacos , Linhagem Celular , Imunofluorescência , Leptina/sangue , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase
10.
J Neurosci ; 30(22): 7516-27, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20519526

RESUMO

The development of novel therapeutic strategies for Alzheimer's disease (AD) represents one of the biggest unmet medical needs today. Application of neurotrophic factors able to modulate neuronal survival and synaptic connectivity is a promising therapeutic approach for AD. We aimed to determine whether the loco-regional delivery of ciliary neurotrophic factor (CNTF) could prevent amyloid-beta (Abeta) oligomer-induced synaptic damages and associated cognitive impairments that typify AD. To ensure long-term administration of CNTF in the brain, we used recombinant cells secreting CNTF encapsulated in alginate polymers. The implantation of these bioreactors in the brain of Abeta oligomer-infused mice led to a continuous secretion of recombinant CNTF and was associated with the robust improvement of cognitive performances. Most importantly, CNTF led to full recovery of cognitive functions associated with the stabilization of synaptic protein levels in the Tg2576 AD mouse model. In vitro as well as in vivo, CNTF activated a Janus kinase/signal transducer and activator of transcription-mediated survival pathway that prevented synaptic and neuronal degeneration. These preclinical studies suggest that CNTF and/or CNTF receptor-associated pathways may have AD-modifying activity through protection against progressive Abeta-related memory deficits. Our data also encourage additional exploration of ex vivo gene transfer for the prevention and/or treatment of AD.


Assuntos
Doença de Alzheimer/complicações , Fator Neurotrófico Ciliar/biossíntese , Fator Neurotrófico Ciliar/uso terapêutico , Transtornos da Memória/etiologia , Transtornos da Memória/terapia , Sinapses/efeitos dos fármacos , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Animais , Apoptose/genética , Encéfalo/patologia , Contagem de Células/métodos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Fator Neurotrófico Ciliar/administração & dosagem , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Humanos , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sinapses/metabolismo , Sinaptossomos/metabolismo , Sinaptossomos/patologia , Sinaptossomos/ultraestrutura , Fatores de Tempo , Transfecção/métodos
11.
Hippocampus ; 20(8): 949-61, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19714568

RESUMO

In the context of their potential implication in regenerative strategies, we characterized cell mechanisms underlying the fate of embryonic rat hippocampal H19-7 progenitors in culture upon induction of their differentiation, and tested their capacities to integrate into a neuronal network in vitro. Without addition of growth factors, nearly 100% of cells expressed various neuronal markers, with a progressive rise of the expression of Synapsin I and II, suggesting that cells developed as mature neurons with synaptogenic capacities. Fully differentiated neurons were identified as glutamatergic and expressed the receptor-associated protein PSD-95. Quantification of ATP showed that 60% of cells died within 24 h after differentiation. Cell death was shown to imply Erk1/2-dependent intrinsic mitochondrial apoptosis signaling pathway, with activation of caspase-9 and -3, finally leading to single-strand DNA. Surviving neurons displayed high levels of Akt, phospho-Akt, and antiapoptotic proteins such as Bcl-2 and Bcl-XL, with decreased caspase activation. In the absence of trophic support, the proapoptotic death-associated protein (DAP) kinase was dramatically stimulated by 24 h postdifferentiation, along with increased levels of p38 and phospho-p38, and caspase reactivation. These findings show that different signaling pathways are sequentially triggered by differentiation, and highlight that ultimate cell death would involve p38 and DAP kinase activation. This was supported by the improvement of cell survival at 24-h postdifferentiation when cells were treated by PD169316, a specific inhibitor of p38. Finally, when seeded on rat hippocampal primary cultured neurons, a significant number of differentiated H19-7 cells were able to survive and to develop cell-cell communication.


Assuntos
Diferenciação Celular/fisiologia , Hipocampo/citologia , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Células-Tronco/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Caspases/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura/métodos , Proteína 4 Homóloga a Disks-Large , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Imidazóis/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Membranas Mitocondriais/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Fatores de Tempo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
12.
Am J Pathol ; 176(1): 270-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19948829

RESUMO

Methyl donor deficiency (MDD) during pregnancy influences intrauterine development. Ghrelin is expressed in the stomach of fetuses and influences fetal growth, but MDD influence on gastric ghrelin is unknown. We examined the gastric ghrelin system in MDD-induced intrauterine growth retardation. By using specific markers and approaches (such as periodic acid-Schiff, bromodeoxyuridine, homocysteine, terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling, immunostaining, reverse transcription-polymerase chain reaction), we studied the gastric oxyntic mucosa cellular organization and ghrelin gene expression in the mucosa in 20-day-old fetuses and weanling pups, and plasma ghrelin concentration in weanling rat pups of dams either normally fed or deprived of choline, folate, vitamin B6, and vitamin B12 during gestation and suckling periods. MDD fetuses weighed less than controls; the weight deficit reached 57% at weaning (P < 0.001). Both at the end of gestation and at weaning, they presented with an aberrant gastric oxyntic mucosa formation with loss of cell polarity, anarchic cell migration, abnormal progenitor differentiation, apoptosis, and signs of surface layer erosion. Ghrelin cells were abnormally located in the pit region of oxyntic glands. At weaning, plasma ghrelin levels were decreased (-28%; P < 0.001) despite unchanged mRNA expression in the stomach. This decrease was associated with lower body weight. Taken together, these data indicate that one mechanism through which MDD influences fetal programming is the remodeling of gastric cellular organization, leading to dysfunction of the ghrelin system and dramatic effects on growth.


Assuntos
Deficiências Nutricionais/embriologia , Deficiências Nutricionais/fisiopatologia , Desenvolvimento Fetal , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Grelina/metabolismo , Animais , Peso Corporal , Linhagem da Célula , Colina/metabolismo , Células Enteroendócrinas/metabolismo , Feminino , Ácido Fólico/metabolismo , Grelina/sangue , Hormônio do Crescimento/sangue , Homocisteína/sangue , Imuno-Histoquímica , Gravidez , Ratos , Ratos Wistar , Vitamina B 12/metabolismo , Desmame
13.
Biochimie ; 91(6): 804-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19303044

RESUMO

In the absence of efficient diagnostic and therapeutic tools, Alzheimer's disease (AD) is a major public health concern due to longer life expectancy in the Western countries. Although the precise cause of AD is still unknown, soluble beta-amyloid (Abeta) oligomers are considered the proximate effectors of the synaptic injury and neuronal death occurring in the early stages of AD. Abeta oligomers may directly interact with the synaptic membrane, leading to impairment of synaptic functions and subsequent signalling pathways triggering neurodegeneration. Therefore, membrane structure and lipid status should be considered determinant factors in Abeta-oligomer-induced synaptic and cell injuries, and therefore AD progression. Numerous epidemiological studies have highlighted close relationships between AD incidence and dietary patterns. Among the nutritional factors involved, lipids significantly influence AD pathogenesis. It is likely that maintenance of adequate membrane lipid content could prevent the production of Abeta peptide as well as its deleterious effects upon its interaction with synaptic membrane, thereby protecting neurons from Abeta-induced neurodegeneration. As major constituents of neuronal lipids, n-3 polyunsaturated fatty acids are of particular interest in the prevention of AD valuable diet ingredients whose neuroprotective properties could be essential for designing preventive nutrition-based strategies. In this review, we discuss the functional relevance of neuronal membrane features with respect to susceptibility to Abeta oligomers and AD pathogenesis, as well as the prospective capacities of lipids to prevent or to delay the disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Metabolismo dos Lipídeos/fisiologia , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Apoptose/fisiologia , Ácidos Graxos Ômega-3/metabolismo , Humanos , Microdomínios da Membrana/metabolismo , Modelos Biológicos , Transdução de Sinais/fisiologia
14.
Neurobiol Aging ; 29(9): 1319-33, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17459527

RESUMO

N-terminal-truncated forms of amyloid-beta (A beta) peptide have been recently suggested to play a pivotal role early in Alzheimer's disease (AD). Among them, A beta 3(pE)-42 peptide, starting with pyroglutamyl at residue Glu-3, is considered as the predominant A beta species in AD plaques and pre-amyloid lesions. Its abundance is reported to be directly proportional to the severity of the clinical phenotype. The present study investigates the effects of soluble oligomeric A beta 3(pE)-42 after intracerebroventricular injection on mice learning ability and the molecular mechanisms of its in vitro neurotoxicity. Mice injected with soluble A beta 3(pE)-42 or A beta(l-42) displayed impaired spatial working memory and delayed memory acquisition in Y-maze and Morris water maze tests, while those injected with soluble A beta(42-1) showed no effect. These cognitive alterations were associated with free radical overproduction in the hippocampus and olfactory bulbs, but not in the cerebral cortex or cerebellum. In vitro, A beta 3(pE)-42 oligomers induced a redox-sensitive neuronal apoptosis involving caspase activation and an arachidonic acid-dependent pro-inflammatory pathway. These data suggest that A beta 3(pE)-42 could mediate the neurodegenerative process and subsequent cognitive alteration occurring in preclinical AD stages.


Assuntos
Peptídeos beta-Amiloides/administração & dosagem , Aprendizagem em Labirinto/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fragmentos de Peptídeos/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
J Neurol Sci ; 262(1-2): 27-36, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17681547

RESUMO

Alzheimer's disease (AD) is a major public health concern in all countries. Although the precise cause of AD is still unknown, a growing body of evidence supports the notion that soluble amyloid beta-peptide (Abeta) may be the proximate cause of synaptic injuries and neuronal death early in the disease. AD patients display lower levels of docosahexaenoic acid (DHA, C22:6 ; n-3) in plasma and brain tissues as compared to age-matched controls. Furthermore, epidemiological studies suggest that high DHA intake might have protective properties against neurodegenerative diseases. These observations are supported by in vivo studies showing that DHA-rich diets limits the synaptic loss and cognitive defects induced by Abeta peptide. Although the molecular basis of these neuroprotective effects remains unknown, several mechanisms have been proposed such as (i) regulation of the expression of potentially protective genes, (ii) activation of anti-inflammatory pathways, (iii) modulation of functional properties of the synaptic membranes along with changes in their physicochemical and structural features.


Assuntos
Doença de Alzheimer/dietoterapia , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/antagonistas & inibidores , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ácidos Docosa-Hexaenoicos/uso terapêutico , Encefalite/dietoterapia , Fármacos Neuroprotetores/uso terapêutico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/fisiopatologia , Ácidos Docosa-Hexaenoicos/metabolismo , Encefalite/metabolismo , Encefalite/fisiopatologia , Alimentos Formulados/normas , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/fisiologia , Fármacos Neuroprotetores/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
16.
Neurobiol Dis ; 23(1): 178-89, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16626961

RESUMO

Recent data have revealed that soluble oligomeric amyloid-beta peptide (Abeta) may be the proximate effectors of neuronal injuries and death in Alzheimer's disease (AD) by unknown mechanisms. Consistently, we recently demonstrated the critical role of a redox-sensitive cytosolic calcium-dependent phospholipase A2 (cPLA2)-arachidonic acid (AA) pathway in Abeta oligomer-induced cell death. According to the involvement of oxidative stress and polyunsaturated fatty acids like AA in the regulation of sphingomyelinase (SMase) activity, the present study underlines the role of SMases in soluble Abeta-induced apoptosis. Soluble Abeta oligomers induced the activation of both neutral and acidic SMases, as demonstrated by the direct measurement of their enzymatic activities, by the inhibitory effects of both specific neutral and acidic SMase inhibitors, and by gene knockdown using antisense oligonucleotides. Furthermore, soluble Abeta-mediated activation of SMases and subsequent cell death were found to be inhibited by antioxidant molecules and a cPLA2-specific inhibitor or antisense oligonucleotide. We also demonstrate that sphingosine-1-phosphate is a potent neuroprotective factor against soluble Abeta oligomer-induced cell death and apoptosis by inhibiting soluble Abeta-induced activation of acidic sphingomyelinase. These results suggest that Abeta oligomers induce neuronal death by activating neutral and acidic SMases in a redox-sensitive cPLA2-AA pathway.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apoptose/fisiologia , Ceramidas/metabolismo , Neurônios/patologia , Fosfolipases A/metabolismo , Esfingomielina Fosfodiesterase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Ácido Araquidônico/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Lisofosfolipídeos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oligonucleotídeos Antissenso , Oxirredução , Fosfolipases A2 , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados
17.
J Neurochem ; 96(2): 385-95, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16300635

RESUMO

A growing body of evidence supports the notion that soluble oligomers of amyloid-beta (Abeta) peptide interact with the neuronal plasma membrane, leading to cell injury and inducing death-signalling pathways that could account for the increased neurodegeneration occurring in Alzheimer's disease (AD). Docosahexaenoic acid (DHA, C22:6, n-3) is an essential polyunsaturated fatty acid in the CNS and has been shown in several epidemiological and in vivo studies to have protective effects against AD and cognitive alterations. However, the molecular mechanisms involved remain unknown. We hypothesized that DHA enrichment of plasma membranes could protect neurones from apoptosis induced by soluble Abeta oligomers. DHA pre-treatment was observed to significantly increase neuronal survival upon Abeta treatment by preventing cytoskeleton perturbations, caspase activation and apoptosis, as well as by promoting extracellular signal-related kinase (ERK)-related survival pathways. These data suggest that DHA enrichment probably induces changes in neuronal membrane properties with functional outcomes, thereby increasing protection from soluble Abeta oligomers. Such neuroprotective effects could be of major interest in the prevention of AD and other neurodegenerative diseases.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/farmacologia , Apoptose/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/farmacologia , Neurônios/fisiologia , Fármacos Neuroprotetores/farmacologia , Animais , Caspases/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Citoesqueleto/efeitos dos fármacos , Citosol/enzimologia , Ativação Enzimática/efeitos dos fármacos , Ácidos Graxos Ômega-3/farmacologia , Oligodendroglia/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Fosfolipases A/metabolismo , Ratos , Ratos Wistar , Solubilidade
18.
J Biol Chem ; 281(1): 229-40, 2006 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-16234245

RESUMO

A growing body of evidence supports the notion that soluble oligomeric forms of the amyloid beta-peptide (Abeta) may be the proximate effectors of neuronal injuries and death in the early stages of Alzheimer disease. However, the molecular mechanisms associated with neuronal apoptosis induced by soluble Abeta remain to be elucidated. We recently demonstrated the involvement of an early reactive oxygen species-dependent perturbation of the microtubule network (Sponne, I., Fifre, A., Drouet, B., Klein, C., Koziel, V., Pincon-Raymond, M., Olivier, J.-L., Chambaz, J., and Pillot, T. (2003) J. Biol. Chem. 278, 3437-3445). Because microtubule-associated proteins (MAPs) are responsible for the polymerization, stabilization, and dynamics of the microtubule network, we investigated whether MAPs might represent the intracellular targets that would enable us to explain the microtubule perturbation involved in soluble Abeta-mediated neuronal apoptosis. The data presented here show that soluble Abeta oligomers induce a time-dependent degradation of MAP1A, MAP1B, and MAP2 involving a perturbation of Ca2+ homeostasis with subsequent calpain activation that, on its own, is sufficient to induce the proteolysis of isoforms MAP2a, MAP2b, and MAP2c. In contrast, MAP1A and MAP1B sequential proteolysis results from the Abeta-mediated activation of caspase-3 and calpain. The prevention of MAP1A, MAP1B, and MAP2 proteolysis by antioxidants highlights the early reactive oxygen species generation in the perturbation of the microtubule network induced by soluble Abeta. These data clearly demonstrate the impact of cytoskeletal perturbations on soluble Abeta-mediated cell death and support the notion of microtubule-stabilizing agents as effective Alzheimer disease drugs.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Apoptose/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fragmentos de Peptídeos/toxicidade , Animais , Apoptose/fisiologia , Cálcio/metabolismo , Calpaína/metabolismo , Caspase 3 , Caspase 9 , Caspases/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Homeostase/efeitos dos fármacos , Isomerismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/química , Estresse Oxidativo/fisiologia
19.
FASEB J ; 19(1): 85-7, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15486059

RESUMO

Recent data have revealed that soluble oligomeric forms of amyloid peptide (Abeta) may be the proximate effectors of the neuronal injury and death occurring in Alzheimer's disease (AD). However, the molecular mechanisms associated with the neuronal cell death induced by the nonfibrillar Abeta remain to be elucidated. In this study, we investigated the role of the cytosolic Ca2+-dependent phospholipase A2 (cPLA2), and its associated metabolic pathway, i.e., the arachidonic acid (AA) cascade, in the apoptotic cell death induced by soluble oligomers of Abeta. The treatment of rat cortical neurons with low concentrations of soluble Abeta(1-40) or Abeta(1-42) peptide resulted in an early calcium-dependent release of AA associated with a transient relocalization of cPLA2. Both cPLA2 antisense oligonucleotides and a selective inhibitor of cPLA2 activity abolished the release of AA from neurons and also protected cells against apoptosis induced by Abeta. Furthermore, inhibitors of the PKC, p38, and MEK/ERK pathways that are involved in cPLA2 phosphorylation and activation reduced Abeta-induced cell death. Finally, we demonstrate that inhibitors of cyclooxygenase-2 reduced the Abeta-induced cell death by 55%. Our studies suggest a novel neuronal response of soluble oligomers of Abeta, which occurs through a cPLA2 signaling cascade and an AA-dependent death pathway. This may prove to be crucial in AD processes and could provide important targets for drug development.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apoptose/fisiologia , Citosol/enzimologia , Neurônios/enzimologia , Neurônios/fisiologia , Fragmentos de Peptídeos/metabolismo , Fosfolipases A/metabolismo , Animais , Ácido Araquidônico/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Ciclo-Oxigenase 2 , Humanos , Proteínas de Membrana , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfolipases A2 , Prostaglandina-Endoperóxido Sintases/metabolismo , Ratos , Solubilidade
20.
Glia ; 47(1): 1-8, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15139007

RESUMO

Neurodegenerative prion diseases, characterized by a progressive dementia, are associated with the accumulation of abnormal forms of the prion (PrPc) protein, potentially due to an aberrant regulation of PrPc biogenesis and/or topology. One of these forms, termed ctmPrP, displays a transmembrane conformation and might trigger neuronal cell death in Gerstmann-Straüssler-Scheinker (GSS) syndrome and other prion-associated diseases in humans. Although the primary target cells involved in the progression of prion diseases remain unidentified, it was recently suggested that modifications of the oligodendroglial cells occur early in prion diseases. In the present study, we demonstrate that a putative transmembrane domain of the human PrPc, i.e., amino acids 118-135, induces oligodendrocyte (OLG) death in vitro in a time- and dose-dependent manner. The process leading to OLG death and induced by the PrP[118-135] peptide was characterized by DNA fragmentation, cytoskeletal disruption, and caspase activation. Protection against the PrP[118-135] peptide-induced OLG apoptosis by several antioxidant molecules, such as probucol, propylgallate, and promethazine, suggests that oxidative injuries contribute to the PrP[118-135] cytotoxicity to OLGs. These results suggest a potential pathophysiological role of the ctmPrP- and/or PrP fragment-mediated OLG cytotoxicity in spongiform encephalopathies.


Assuntos
Apoptose , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Fragmentos de Peptídeos/metabolismo , Doenças Priônicas/metabolismo , Príons/metabolismo , Animais , Animais Recém-Nascidos , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Caspases/efeitos dos fármacos , Caspases/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Fragmentação do DNA/efeitos dos fármacos , Fragmentação do DNA/fisiologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Fragmentos de Peptídeos/toxicidade , Proteínas PrPC/metabolismo , Doenças Priônicas/patologia , Doenças Priônicas/fisiopatologia , Príons/toxicidade , Estrutura Terciária de Proteína/fisiologia , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...