Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cell ; 133(7): 1149-61, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18585350

RESUMO

Alzheimer's disease (AD) is a genetically heterogeneous disorder characterized by early hippocampal atrophy and cerebral amyloid-beta (Abeta) peptide deposition. Using TissueInfo to screen for genes preferentially expressed in the hippocampus and located in AD linkage regions, we identified a gene on 10q24.33 that we call CALHM1. We show that CALHM1 encodes a multipass transmembrane glycoprotein that controls cytosolic Ca(2+) concentrations and Abeta levels. CALHM1 homomultimerizes, shares strong sequence similarities with the selectivity filter of the NMDA receptor, and generates a large Ca(2+) conductance across the plasma membrane. Importantly, we determined that the CALHM1 P86L polymorphism (rs2986017) is significantly associated with AD in independent case-control studies of 3404 participants (allele-specific OR = 1.44, p = 2 x 10(-10)). We further found that the P86L polymorphism increases Abeta levels by interfering with CALHM1-mediated Ca(2+) permeability. We propose that CALHM1 encodes an essential component of a previously uncharacterized cerebral Ca(2+) channel that controls Abeta levels and susceptibility to late-onset AD.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Cálcio/metabolismo , Predisposição Genética para Doença , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Polimorfismo Genético , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Canais de Cálcio , Membrana Celular/metabolismo , Cromossomos Humanos Par 10 , Citosol/metabolismo , Feminino , Genoma Humano , Humanos , Masculino , Glicoproteínas de Membrana/química , Pessoa de Meia-Idade , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência
2.
Neurobiol Dis ; 129: 217-233, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30928644

RESUMO

Alzheimer's Disease is a devastating dementing disease involving amyloid deposits, neurofibrillary tangles, progressive and irreversible cognitive impairment. Today, only symptomatic drugs are available and therapeutic treatments, possibly acting at a multiscale level, are thus urgently needed. To that purpose, we designed multi-effects compounds by synthesizing drug candidates derived by substituting a novel N,N'-disubstituted piperazine anti-amyloid scaffold and adding acetylcholinesterase inhibition property. Two compounds were synthesized and evaluated. The most promising hybrid molecule reduces both the amyloid pathology and the Tau pathology as well as the memory impairments in a preclinical model of Alzheimer's disease. In vitro also, the compound reduces the phosphorylation of Tau and inhibits the release of Aß peptides while preserving the processing of other metabolites of the amyloid precursor protein. We synthetized and tested the first drug capable of ameliorating both the amyloid and Tau pathology in animal models of AD as well as preventing the major brain lesions and associated memory impairments. This work paves the way for future compound medicines against both Alzheimer's-related brain lesions development and the associated cognitive impairments.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/efeitos dos fármacos , Degeneração Neural/patologia , Fármacos Neuroprotetores/farmacologia , Piperazinas/farmacologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Humanos , Memória/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Placa Amiloide/patologia
3.
J Biomed Sci ; 26(1): 89, 2019 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-31666073

RESUMO

BACKGROUND: Effective neurorestorative therapies of neurodegenerative diseases must be developed. There is increasing interest in using human platelet lysates, rich in neurotrophic factors, as novel disease-modifying strategy of neurodegeneration. To ensure virus safety, pathogen reduction treatments should be incorporated in the preparation process of the platelet concentrates used as source material. We therefore investigated whether platelet concentrates (PC) pathogen-inactivated using a licensed photo-inactivation treatment combining photosensitive psoralen (amotosalen) and UVA irradiation (Intercept) can serve as source material to prepare platelet lysates with preserved neuroprotective activity in Parkinson's disease models. METHODS: Intercept treated-PCs were centrifuged, when reaching expiry day (7 days after collection), to remove plasma and platelet additive solution. The platelet pellet was re-suspended and concentrated in phosphate buffer saline, subjected to 3 freeze-thaw cycles (- 80 °C/37 °C) then centrifuged to remove cell debris. The supernatant was recovered and further purified, or not, by heat-treatment as in our previous investigations. The content in proteins and neurotrophic factors was determined and the toxicity and neuroprotective activity of the platelet lysates towards LUHMES cells or primary cortical/hippocampal neurons were assessed using ELISA, flow cytometry, cell viability and cytotoxicity assays and proteins analysis by Western blot. RESULTS: Platelet lysates contained the expected level of total proteins (ca. 7-14 mg/mL) and neurotrophic factors. Virally inactivated and heat-treated platelet lysates did not exert detectable toxic effects on neither Lund human mesencephalic dopaminergic LUHMES cell line nor primary neurons. When used at doses of 5 and 0.5%, they enhanced the expression of tyrosine hydroxylase and neuron-specific enolase in LUHMES cells and did not significantly impact synaptic protein expression in primary neurons, respectively. Furthermore, virally-inactivated platelet lysates tested were found to exert very strong neuroprotection effects on both LUHMES and primary neurons exposed to erastin, an inducer of ferroptosis cell death. CONCLUSION: Outdated Intercept pathogen-reduced platelet concentrates can be used to prepare safe and highly neuroprotective human heat-treated platelet pellet lysates. These data open reassuring perspectives in the possibility to develop an effective biotherapy using virally-inactivated platelet lysates rich in functional neurotrophins for neuroregenerative medicine, and for further bio-industrial development. However, the data should be confirmed in animal models.


Assuntos
Plaquetas/fisiologia , Furocumarinas/farmacologia , Temperatura Alta , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , Animais , Materiais Biocompatíveis/efeitos da radiação , Plaquetas/efeitos da radiação , Linhagem Celular , Humanos , Camundongos , Neurônios/efeitos dos fármacos , Doença de Parkinson/metabolismo , Raios Ultravioleta
4.
Nature ; 495(7440): 223-6, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23467090

RESUMO

Recognition of sweet, bitter and umami tastes requires the non-vesicular release from taste bud cells of ATP, which acts as a neurotransmitter to activate afferent neural gustatory pathways. However, how ATP is released to fulfil this function is not fully understood. Here we show that calcium homeostasis modulator 1 (CALHM1), a voltage-gated ion channel, is indispensable for taste-stimuli-evoked ATP release from sweet-, bitter- and umami-sensing taste bud cells. Calhm1 knockout mice have severely impaired perceptions of sweet, bitter and umami compounds, whereas their recognition of sour and salty tastes remains mostly normal. Calhm1 deficiency affects taste perception without interfering with taste cell development or integrity. CALHM1 is expressed specifically in sweet/bitter/umami-sensing type II taste bud cells. Its heterologous expression induces a novel ATP permeability that releases ATP from cells in response to manipulations that activate the CALHM1 ion channel. Knockout of Calhm1 strongly reduces voltage-gated currents in type II cells and taste-evoked ATP release from taste buds without affecting the excitability of taste cells by taste stimuli. Thus, CALHM1 is a voltage-gated ATP-release channel required for sweet, bitter and umami taste perception.


Assuntos
Canais de Cálcio/metabolismo , Transmissão Sináptica , Paladar/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Canais de Cálcio/deficiência , Canais de Cálcio/genética , Feminino , Células HeLa , Humanos , Ativação do Canal Iônico , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Receptores Purinérgicos/metabolismo , Análise de Célula Única , Paladar/genética , Papilas Gustativas/citologia , Papilas Gustativas/metabolismo
5.
Int J Mol Sci ; 19(12)2018 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-30467274

RESUMO

Long-term memory formation depends on the expression of immediate early genes (IEGs). Their expression, which is induced by synaptic activation, is mainly regulated by the 3',5'-cyclic AMP (cAMP)-dependent protein kinase/cAMP response element binding protein (cAMP-dependent protein kinase (PKA)/ cAMP response element binding (CREB)) signaling pathway. Synaptic activation being highly energy demanding, neurons must maintain their energetic homeostasis in order to successfully induce long-term memory formation. In this context, we previously demonstrated that the expression of IEGs required the activation of AMP-activated protein kinase (AMPK) to sustain the energetic requirements linked to synaptic transmission. Here, we sought to determine the molecular mechanisms by which AMPK regulates the expression of IEGs. To this end, we assessed the involvement of AMPK in the regulation of pathways involved in the expression of IEGs upon synaptic activation in differentiated primary neurons. Our data demonstrated that AMPK regulated IEGs transcription via the PKA/CREB pathway, which relied on the activity of the soluble adenylyl cyclase. Our data highlight the interplay between AMPK and PKA/CREB signaling pathways that allows synaptic activation to be transduced into the expression of IEGs, thus exemplifying how learning and memory mechanisms are under metabolic control.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Adenilil Ciclases/genética , Proteína de Ligação a CREB/genética , Proteínas Quinases Dependentes de AMP Cíclico/genética , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/metabolismo , 4-Aminopiridina/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Adenilil Ciclases/metabolismo , Animais , Bicuculina/farmacologia , Proteína de Ligação a CREB/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Embrião de Mamíferos , Genes Precoces , Memória de Longo Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Cultura Primária de Células , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transmissão Sináptica
6.
Mol Med ; 22: 841-849, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27900387

RESUMO

Strategies aimed at reducing cerebral accumulation of the amyloid-ß (Aß) peptides have therapeutic potential in Alzheimer's disease (AD). Aß immunization has proven to be effective at promoting Aß clearance in animal models but adverse effects have hampered its clinical evaluation. The first anti-Aß immunization clinical trial, which assessed a full-length Aß1-42 vaccine, increased the risk of encephalitis most likely because of autoimmune pro-inflammatory T helper 1 (Th1) response against all forms of Aß. Immunization against less abundant but potentially more pathologically relevant Aß products, such as N-terminally-truncated pyroglutamate-3 Aß (AßpE3), could provide efficacy and improve tolerability in Aß immunotherapy. Here, we describe a selective vaccine against AßpE3, which uses the diphtheria toxin mutant CRM197 as carrier protein for epitope presentation. CRM197 is currently used in licensed vaccines and has demonstrated excellent immunogenicity and safety in humans. In mice, our AßpE3:CRM197 vaccine triggered the production of specific anti-AßpE3 antibodies that did not cross-react with Aß1-42, non-cyclized AßE3, or N-terminally-truncated pyroglutamate-11 Aß (AßpE11). AßpE3:CRM197 antiserum strongly labeled AßpE3 in insoluble protein extracts and decorated cortical amyloid plaques in human AD brains. Anti-AßpE3 antibodies were almost exclusively of the IgG1 isotype, suggesting an anti-inflammatory Th2 response bias to the AßpE3:CRM197 vaccine. To the best of our knowledge, this study shows for the first time that CRM197 has potential as a safe and suitable vaccine carrier for active and selective immunization against specific protein sequence modifications or conformations, such as AßpE3.

7.
J Cell Sci ; 128(13): 2330-8, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25999473

RESUMO

Alzheimer's disease is characterized by amyloid-ß (Aß) peptide accumulation in the brain. CALHM1, a cell-surface Ca(2+) channel expressed in brain neurons, has anti-amyloidogenic properties in cell cultures. Here, we show that CALHM1 controls Aß levels in vivo in the mouse brain through a previously unrecognized mechanism of regulation of Aß clearance. Using pharmacological and genetic approaches in cell lines, we found that CALHM1 ion permeability and extracellular Ca(2+) were required for the Aß-lowering effect of CALHM1. Aß level reduction by CALHM1 could be explained by an increase in extracellular Aß degradation by insulin-degrading enzyme (IDE), extracellular secretion of which was strongly potentiated by CALHM1 activation. Importantly, Calhm1 knockout in mice reduced IDE enzymatic activity in the brain, and increased endogenous Aß concentrations by up to ∼50% in both the whole brain and primary neurons. Thus, CALHM1 controls Aß levels in cell lines and in vivo by facilitating neuronal and Ca(2+)-dependent degradation of extracellular Aß by IDE. This work identifies CALHM1 ion channel as a potential target for promoting amyloid clearance in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Canais de Cálcio/metabolismo , Insulisina/metabolismo , Animais , Cálcio/farmacologia , Canais de Cálcio/deficiência , Linhagem Celular , Citidina Desaminase/metabolismo , Espaço Extracelular/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos Knockout , Proteólise/efeitos dos fármacos , Solubilidade
8.
J Neurosci ; 34(36): 12230-8, 2014 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-25186765

RESUMO

The AMP-activated protein kinase (AMPK) is a Ser/Thr kinase that is activated in response to low-energy states to coordinate multiple signaling pathways to maintain cellular energy homeostasis. Dysregulation of AMPK signaling has been observed in Alzheimer's disease (AD), which is associated with abnormal neuronal energy metabolism. In the current study we tested the hypothesis that aberrant AMPK signaling underlies AD-associated synaptic plasticity impairments by using pharmacological and genetic approaches. We found that amyloid ß (Aß)-induced inhibition of long-term potentiation (LTP) and enhancement of long-term depression were corrected by the AMPK inhibitor compound C (CC). Similarly, LTP impairments in APP/PS1 transgenic mice that model AD were improved by CC treatment. In addition, Aß-induced LTP failure was prevented in mice with genetic deletion of the AMPK α2-subunit, the predominant AMPK catalytic subunit in the brain. Furthermore, we found that eukaryotic elongation factor 2 (eEF2) and its kinase eEF2K are key downstream effectors that mediate the detrimental effects of hyperactive AMPK in AD pathophysiology. Our findings describe a previously unrecognized role of aberrant AMPK signaling in AD-related synaptic pathophysiology and reveal a potential therapeutic target for AD.


Assuntos
Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Hipocampo/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Animais , Células Cultivadas , Quinase do Fator 2 de Elongação/metabolismo , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator 2 de Elongação de Peptídeos/metabolismo , Presenilina-1/genética , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais
9.
J Cell Sci ; 126(Pt 5): 1199-206, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23345406

RESUMO

Calcium homeostasis modulator 1 (CALHM1) is a Ca(2+) channel controlling neuronal excitability and potentially involved in the pathogenesis of Alzheimer's disease (AD). Although strong evidence indicates that CALHM1 is required for neuronal electrical activity, its role in intracellular Ca(2+) signaling remains unknown. In the present study, we show that in hippocampal HT-22 cells, CALHM1 expression led to a robust and relatively selective activation of the Ca(2+)-sensing kinases ERK1/2. CALHM1 also triggered activation of MEK1/2, the upstream ERK1/2-activating kinases, and of RSK1/2/3 and MSK1, two downstream effectors of ERK1/2 signaling. CALHM1-mediated activation of ERK1/2 signaling was controlled by the small GTPase Ras. Pharmacological inhibition of CALHM1 permeability using Ruthenium Red, Zn(2+), and Gd(3+), or expression of the CALHM1 N140A and W114A mutants, which are deficient in mediating Ca(2+) influx, prevented the effect of CALHM1 on the MEK, ERK, RSK and MSK signaling cascade, demonstrating that CALHM1 controlled this pathway via its channel properties. Importantly, expression of CALHM1 bearing the natural P86L polymorphism, which leads to a partial loss of CALHM1 function and is associated with an earlier age at onset in AD patients, showed reduced activation of ERK1/2, RSK1/2/3, and MSK1. In line with these results obtained in transfected cells, primary cerebral neurons isolated from Calhm1 knockout mice showed significant impairments in the activation of MEK, ERK, RSK and MSK signaling. The present study identifies a previously uncharacterized mechanism of control of Ca(2+)-dependent ERK1/2 signaling in neurons, and further establishes CALHM1 as a critical ion channel for neuronal signaling and function.


Assuntos
Canais de Cálcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurônios/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Cálcio , Canais de Cálcio/genética , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Neurônios/efeitos dos fármacos , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Rutênio Vermelho/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
10.
Proc Natl Acad Sci U S A ; 109(28): E1963-71, 2012 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-22711817

RESUMO

Extracellular Ca(2+) (Ca(2+)(o)) plays important roles in physiology. Changes of Ca(2+)(o) concentration ([Ca(2+)](o)) have been observed to modulate neuronal excitability in various physiological and pathophysiological settings, but the mechanisms by which neurons detect [Ca(2+)](o) are not fully understood. Calcium homeostasis modulator 1 (CALHM1) expression was shown to induce cation currents in cells and elevate cytoplasmic Ca(2+) concentration ([Ca(2+)](i)) in response to removal of Ca(2+)(o) and its subsequent addback. However, it is unknown whether CALHM1 is a pore-forming ion channel or modulates endogenous ion channels. Here we identify CALHM1 as the pore-forming subunit of a plasma membrane Ca(2+)-permeable ion channel with distinct ion permeability properties and unique coupled allosteric gating regulation by voltage and [Ca(2+)](o). Furthermore, we show that CALHM1 is expressed in mouse cortical neurons that respond to reducing [Ca(2+)](o) with enhanced conductance and action potential firing and strongly elevated [Ca(2+)](i) upon Ca(2+)(o) removal and its addback. In contrast, these responses are strongly muted in neurons from mice with CALHM1 genetically deleted. These results demonstrate that CALHM1 is an evolutionarily conserved ion channel family that detects membrane voltage and extracellular Ca(2+) levels and plays a role in cortical neuronal excitability and Ca(2+) homeostasis, particularly in response to lowering [Ca(2+)](o) and its restoration to normal levels.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Membrana Celular/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/genética , Animais , Sítios de Ligação , Cálcio/química , Canais de Cálcio/genética , Eletrofisiologia/métodos , Feminino , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Íons , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Mutagênese , Doenças Neurodegenerativas/genética , Oócitos/citologia , Oócitos/metabolismo , Polimorfismo Genético , Fatores de Tempo , Xenopus
11.
Mol Med ; 20: 29-36, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24722782

RESUMO

The endocannabinoid CB2 receptor system has been implicated in the neuropathology of Alzheimer's disease (AD). In order to investigate the impact of the CB2 receptor system on AD pathology, a colony of mice with a deleted CB2 receptor gene, CNR2, was established on a transgenic human mutant APP background for pathological comparison with CB2 receptor-sufficient transgenic mice. J20 APP (PDGFB-APPSwInd) mice were bred over two generations with CNR2(-/-) (Cnr2(tm1Dgen)/J) mice to produce a colony of J20 CNR2(+/+) and J20 CNR2(-/-) mice. Seventeen J20 CNR2(+/+) mice (12 females, 5 males) and 16 J20 CNR2(-/-) mice (11 females, 5 males) were killed at 12 months, and their brains were interrogated for AD-related pathology with both biochemistry and immunocytochemistry (ICC). In addition to amyloid-dependent endpoints such as soluble Aß production and plaque deposition quantified with 6E10 staining, the effect of CB2 receptor deletion on total soluble mouse tau production was assayed by using a recently developed high-sensitivity assay. Results revealed that soluble Aß42 and plaque deposition were significantly increased in J20 CNR2(-/-) mice relative to CNR2(+/+) mice. Microgliosis, quantified with ionized calcium-binding adapter molecule 1 (Iba-1) staining, did not differ between groups, whereas plaque associated microglia was more abundant in J20 CNR2(-/-) mice. Total tau was significantly suppressed in J20 CNR2(-/-) mice relative to J20 CNR2(+/+) mice. The results confirm the constitutive role of the CB2 receptor system both in reducing amyloid plaque pathology in AD and also support tehpotential of cannabinoid therapies targeting CB2 to reduce Aß; however, the results suggest that interventions may have a divergent effect on tau pathology.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Agonistas de Receptores de Canabinoides/farmacologia , Canabinoides/farmacologia , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Microglia/metabolismo , Microglia/patologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Proteínas do Grupo Polycomb , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/genética , Fatores de Transcrição/metabolismo
12.
Proteome Sci ; 12: 24, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24944524

RESUMO

BACKGROUND: Lipid lowering agent such as agonists of peroxisome proliferator-activated receptors (PPAR) are suggested as neuroprotective agents and may protect from the sequelae of brain ischemic stroke. Although the demonstration is not clearly established in human, the underlying molecular mechanism may be of interest for future therapeutic purposes. To this end, we have used our well established rodent model of ischemia-reperfusion pre-treated or not with fenofibrate or atorvastatin and performed a differential proteomics analyses of the brain and analysed the protein markers which levels returned to "normal" following pre-treatments with PPARα agonists. RESULTS: In order to identify potential therapeutic targets positively modulated by pre-treatment with the PPARα agonists, two-dimensional gel electrophoresis proteome profiles between control, ischemia-reperfusion and pre-treated or not, were compared. The polypeptide which expression was altered following ischemia - reperfusion but whose levels remain unchanged after pre-treatment were characterized by mass spectrometry and further investigated by Western-blotting and immunohistochemistry. A series of 28 polypeptides were characterized among which the protein disulfide isomerase reduction - a protein instrumental to the unfolded protein response system - was shown to be reduced following PPARα agonists treatment while it was strongly increased in ischemia-reperfusion. CONCLUSIONS: Pre-treatment with PPARα agonist or atorvastatin show potential neuroprotective effects by inhibiting the PDI overexpression in conjunction with the preservation of other neuronal markers, several of which are associated with the regulation of protein homeostasis, signal transduction and maintenance of synaptic plasticity. This proteomic study therefore suggests that neuroprotective effect of PPARα agonists supposes the preservation of the expression of several proteins essential for the maintenance of protein homeostasis not necessarily directly linked to PPARα known-regulated targets.

13.
Hum Mol Genet ; 20(10): 2026-36, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21357679

RESUMO

Alzheimer's disease (AD), the most common neurodegenerative disorder, is characterized by cerebral deposition of amyloid-ß (Aß), a series of peptides derived from the processing of the amyloid-ß precursor protein (APP). To identify new candidate genes for AD, we recently performed a transcriptome analysis to screen for genes preferentially expressed in the hippocampus and located in AD linkage regions. This strategy identified CALHM1 (calcium homeostasis modulator 1), a gene modulating AD age at onset and Aß metabolism. Here, we focused our attention on another candidate identified using this screen, growth arrest-specific 1 (Gas1), a gene involved in the central nervous system development. We found that Gas1 formed a complex with APP and controlled APP maturation and processing. Gas1 expression inhibited APP full glycosylation and routing to the cell surface by leading to a trafficking blockade of APP between the endoplasmic reticulum and the Golgi. Gas1 expression also resulted in a robust inhibition of APP transport into multivesicular bodies, further demonstrating that Gas1 negatively regulated APP intracellular trafficking. Consequently, Gas1 overexpression led to a reduction in Aß production, and conversely, Gas1 silencing in cells expressing endogenously Gas1 increased Aß levels. These results suggest that Gas1 is a novel APP-interacting protein involved in the control of APP maturation and processing.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Ciclo Celular/metabolismo , Processamento de Proteína Pós-Traducional , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular , Retículo Endoplasmático/metabolismo , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Ordem dos Genes , Predisposição Genética para Doença/genética , Glicosilação , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Polimorfismo de Nucleotídeo Único/genética , Ligação Proteica , Transporte Proteico
14.
Mol Med ; 19: 357-64, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24408112

RESUMO

The endocannabinoid CB2 receptor system has been implicated in the neuropathology of Alzheimer's disease (AD). In order to investigate the impact of the CB2 receptor system on AD pathology, a colony of mice with a deleted CB2 receptor gene, CNR2, was established on a transgenic human mutant APP background for pathological comparison with CB2 receptor-sufficient transgenic mice. J20 APP (PDGFB-APPSwInd) mice were bred over two generations with CNR2⁻/⁻ (Cnr2(tm1Dgen)/J) mice to produce a colony of J20 CNR2⁺/⁺ and J20 CNR2⁻/⁻ mice. Seventeen J20 CNR2⁺/⁺ mice (12 females, 5 males) and 16 J20 CNR2⁻/⁻ mice (11 females, 5 males) were killed at 12 months, and their brains were interrogated for AD-related pathology with both biochemistry and immunocytochemistry (ICC). In addition to amyloid-dependent endpoints such as soluble Aß production and plaque deposition quantified with 6E10 staining, the effect of CB2 receptor deletion on total soluble mouse tau production was assayed by using a recently developed high-sensitivity assay. Results revealed that soluble Aß42 and plaque deposition were significantly increased in J20 CNR2⁻/⁻ mice relative to CNR2⁺/⁺ mice. Microgliosis, quantified with ionized calcium-binding adapter molecule 1 (Iba-1) staining, did not differ between groups, whereas plaque associated microglia was more abundant in J20 CNR2⁻/⁻ mice. Total tau was significantly suppressed in J20 CNR2⁻/⁻ mice relative to J20 CNR2⁺/⁺ mice. The results confirm the constitutive role of the CB2 receptor system both in reducing amyloid plaque pathology in AD and also support tehpotential of cannabinoid therapies targeting CB2 to reduce Aß; however, the results suggest that interventions may have a divergent effect on tau pathology.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Microglia/patologia , Placa Amiloide/metabolismo , Receptor CB2 de Canabinoide/genética , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microglia/metabolismo , Receptor CB2 de Canabinoide/metabolismo
15.
J Neurochem ; 120 Suppl 1: 34-45, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22121879

RESUMO

'Secretase' is a generic term coined more than 20 years ago to refer to a group of proteases responsible for the cleavage of a vast number of membrane proteins. These endoproteolytic events result in the extracellular or intracellular release of soluble metabolites associated with a broad range of intrinsic physiological functions. α-Secretase refers to the activity targeting the amyloid precursor protein (APP) and generating sAPPα, a soluble extracellular fragment potentially associated with neurotrophic and neuroprotective functions. Several proteases from the a disintegrin and metalloproteinase (ADAM) family, including ADAM10 and ADAM17, have been directly or indirectly associated with the constitutive and regulated α-secretase activities. Recent evidence in primary neuronal cultures indicates that ADAM10 may represent the genuine constitutive α-secretase. Mainly because α-secretase cleaves APP within the sequence of Aß, the core component of the cerebral amyloid plaques in Alzheimer's disease, α-secretase activation is considered to be of therapeutic value. In this article, we will provide a historical perspective on the characterization of α-secretase and review the recent literature on the identification and biology of the current α-secretase candidates.


Assuntos
Doença de Alzheimer/enzimologia , Secretases da Proteína Precursora do Amiloide/isolamento & purificação , Secretases da Proteína Precursora do Amiloide/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/química , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Humanos , Placa Amiloide/química , Placa Amiloide/enzimologia , Placa Amiloide/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia
16.
J Neurochem ; 120(3): 461-72, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22118570

RESUMO

Activation of microglia, the resident macrophages of the brain, around the amyloid plaques is a key hallmark of Alzheimer's disease (AD). Recent evidence in mouse models indicates that microglia are required for the neurodegenerative process of AD. Amyloid-ß (Aß) peptides, the core components of the amyloid plaques, can trigger microglial activation by interacting with several Toll-like receptors (TLRs), including TLR4. In this study, we show that resveratrol, a natural polyphenol associated with anti-inflammatory effects and currently in clinical trials for AD, prevented the activation of murine RAW 264.7 macrophages and microglial BV-2 cells treated with the TLR4 ligand, lipopolysaccharide (LPS). Resveratrol preferentially inhibited nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) activation upon LPS stimulation by interfering with IKK and IκB phosphorylation, an effect that potently reduced the transcriptional stimulation of several NF-κB target genes, including tumor necrosis factor-α and interleukin-6. Consequently, downstream phosphorylation of signal transducer and activator of transcription (STAT)1 and STAT3 upon LPS stimulation was also inhibited by resveratrol. We found that resveratrol acted upstream in the activation cascade by interfering with TLR4 oligomerization upon receptor stimulation. Resveratrol treatment also prevented the pro-inflammatory effect of fibrillar Aß on macrophages by potently inhibiting the effect of Aß on IκB phosphorylation, activation of STAT1 and STAT3, and on tumor necrosis factor-α and interleukin-6 secretion. Importantly, orally administered resveratrol in a mouse model of cerebral amyloid deposition lowered microglial activation associated with cortical amyloid plaque formation. Together this work provides strong evidence that resveratrol has in vitro and in vivo anti-inflammatory effects against Aß-triggered microglial activation. Further studies in cell culture systems showed that resveratrol acted via a mechanism involving the TLR4/NF-κB/STAT signaling cascade.


Assuntos
Doença de Alzheimer/patologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Mediadores da Inflamação/farmacologia , Microglia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Estilbenos/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/genética , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular Transformada , Citocinas/metabolismo , Modelos Animais de Doenças , Interações Medicamentosas , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Transgênicos , Inibidor de NF-kappaB alfa , Presenilina-1/genética , Resveratrol , Estatísticas não Paramétricas , Estilbenos/farmacologia , Fatores de Tempo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
17.
FASEB J ; 25(1): 219-31, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20852062

RESUMO

AMP-activated protein kinase (AMPK) is a metabolic sensor involved in intracellular energy metabolism through the control of several homeostatic mechanisms, which include autophagy and protein degradation. Recently, we reported that AMPK activation by resveratrol promotes autophagy-dependent degradation of the amyloid-ß (Aß) peptides, the core components of the cerebral senile plaques in Alzheimer's disease. To identify more potent enhancers of Aß degradation, we screened a library of synthetic small molecules selected for their structural similarities with resveratrol. Here, we report the identification of a series of structurally related molecules, the RSVA series, which inhibited Aß accumulation in cell lines nearly 40 times more potently than did resveratrol. Two of these molecules, RSVA314 and RSVA405, were further characterized and were found to facilitate CaMKKß-dependent activation of AMPK, to inhibit mTOR (mammalian target of rapamycin), and to promote autophagy to increase Aß degradation by the lysosomal system (apparent EC(50) ∼ 1 µM). This work identifies the RSVA compounds as promising lead molecules for the development of a new class of AMPK activating drugs controlling mTOR signaling, autophagy, and Aß clearance.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeos beta-Amiloides/metabolismo , Autofagia/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Proteínas Quinases Ativadas por AMP/genética , Peptídeos beta-Amiloides/genética , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/síntese química , Ativadores de Enzimas/química , Feminino , Células HEK293 , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Resveratrol , Estilbenos/química , Estilbenos/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Transfecção
18.
J Biol Chem ; 285(12): 9100-13, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20080969

RESUMO

Alzheimer disease is an age-related neurodegenerative disorder characterized by amyloid-beta (Abeta) peptide deposition into cerebral amyloid plaques. The natural polyphenol resveratrol promotes anti-aging pathways via the activation of several metabolic sensors, including the AMP-activated protein kinase (AMPK). Resveratrol also lowers Abeta levels in cell lines; however, the underlying mechanism responsible for this effect is largely unknown. Moreover, the bioavailability of resveratrol in the brain remains uncertain. Here we show that AMPK signaling controls Abeta metabolism and mediates the anti-amyloidogenic effect of resveratrol in non-neuronal and neuronal cells, including in mouse primary neurons. Resveratrol increased cytosolic calcium levels and promoted AMPK activation by the calcium/calmodulin-dependent protein kinase kinase-beta. Direct pharmacological and genetic activation of AMPK lowered extracellular Abeta accumulation, whereas AMPK inhibition reduced the effect of resveratrol on Abeta levels. Furthermore, resveratrol inhibited the AMPK target mTOR (mammalian target of rapamycin) to trigger autophagy and lysosomal degradation of Abeta. Finally, orally administered resveratrol in mice was detected in the brain where it activated AMPK and reduced cerebral Abeta levels and deposition in the cortex. These data suggest that resveratrol and pharmacological activation of AMPK have therapeutic potential against Alzheimer disease.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeos beta-Amiloides/química , Doença de Alzheimer/metabolismo , Animais , Autofagia , Cálcio/metabolismo , Citosol/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Resveratrol , Transdução de Sinais , Estilbenos/farmacologia
19.
Mol Med ; 17(9-10): 1022-30, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21647536

RESUMO

AMP-activated protein kinase (AMPK) is a sensor and regulator of cellular energy metabolism potentially implicated in a broad range of conditions, including obesity and Alzheimer's disease. Its role in the control of key metabolic enzymes makes this kinase a central player in glucose and lipid homeostasis. Recently, by screening a library of synthetic small molecules selected for their structural similarity with the natural polyphenol resveratrol, we identified RSVA314 and RSVA405 as potent indirect activators of AMPK (half-maximal effective concentration [EC50] = 1 µmol/L in cell-based assays). Here we show that RSVA314 and RSVA405 can significantly activate AMPK and inhibit acetyl-CoA carboxylase (ACC), one target of AMPK and a key regulator of fatty acid biogenesis, in nondifferentiated and proliferating 3T3-L1 adipocytes. We found that RSVA314 and RSVA405 treatments inhibited 3T3-L1 adipocyte differentiation by interfering with mitotic clonal expansion during preadipocyte proliferation (half-maximal inhibitory concentration [IC50] = 0.5 µmol/L). RSVA314 and RSVA405 prevented the adipogenesis-dependent transcriptional changes of multiple gene products involved in the adipogenic process, including peroxisome proliferator-activated receptor (PPAR)-γ, CCAAT/enhancer-binding protein α (C/EBPα), fatty acid synthase, fatty acid binding protein 4 (aP2), RANTES or resistin. Furthermore, orally administered RSVA405 at 20 and 100 mg/kg/d significantly reduced the body weight gain of mice fed a high-fat diet. This work shows that the novel small-molecule activators of AMPK (RSVA314 and RSVA405) are potent inhibitors of adipogenesis and thus may have therapeutic potential against obesity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Adipogenia/efeitos dos fármacos , Aminofenóis/farmacologia , Ativadores de Enzimas/farmacologia , Hidrazonas/farmacologia , Estilbenos/farmacologia , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/enzimologia , Adipócitos/metabolismo , Adipogenia/genética , Aminofenóis/química , Animais , Antioxidantes/química , Antioxidantes/farmacologia , Western Blotting , Peso Corporal/efeitos dos fármacos , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proliferação de Células/efeitos dos fármacos , Gorduras na Dieta/efeitos adversos , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/química , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Expressão Gênica/efeitos dos fármacos , Hidrazonas/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Obesidade/etiologia , Obesidade/prevenção & controle , PPAR gama/genética , PPAR gama/metabolismo , Resveratrol , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estilbenos/química , Aumento de Peso/efeitos dos fármacos
20.
Mol Med ; 17(9-10): 974-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21629967

RESUMO

The calcium homeostasis modulator 1 (CALHM1) gene codes for a novel cerebral calcium channel controlling intracellular calcium homeostasis and amyloid-ß (Aß) peptide metabolism, a key event in the etiology of Alzheimer's disease (AD). The P86L polymorphism in CALHM1 (rs2986017) initially was proposed to impair CALHM1 functionally and to lead to an increase in Aß accumulation in vitro in cell lines. Recently, it was reported that CALHM1 P86L also may influence Aß metabolism in vivo by increasing Aß levels in human cerebrospinal fluid (CSF). Although the role of CALHM1 in AD risk remains uncertain, concordant data have now emerged showing that CALHM1 P86L is associated with an earlier age at onset of AD. Here, we have analyzed the association of CALHM1 P86L with CSF Aß in samples from 203 AD cases and 46 young cognitively healthy individuals with a positive family history of AD. We failed to detect an association between the CALHM1 polymorphism and CSF Aß levels in AD patients. Our data, however, revealed a significant association of CALHM1 P86L with elevated CSF Aß42 and Aß40 in the normal cohort at risk for AD. This work shows that CALHM1 modulates CSF Aß levels in presymptomatic individuals, strengthening the notion that CALHM1 is involved in AD pathogenesis. These data further demonstrate the utility of endophenotype-based approaches focusing on CSF biomarkers for the identification or validation of risk factors for AD.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Canais de Cálcio/genética , Glicoproteínas de Membrana/genética , Polimorfismo de Nucleotídeo Único , Idoso , Doença de Alzheimer/fisiopatologia , Secretases da Proteína Precursora do Amiloide/líquido cefalorraquidiano , Apolipoproteínas E/genética , Ácido Aspártico Endopeptidases/líquido cefalorraquidiano , Biomarcadores/líquido cefalorraquidiano , Cognição/fisiologia , Estudos de Coortes , Feminino , Frequência do Gene , Predisposição Genética para Doença/genética , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/líquido cefalorraquidiano , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA