Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Int J Mol Sci ; 23(9)2022 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-35563647

RESUMO

Fatigue and other deleterious mood alterations resulting from prolonged efforts such as a long work shift can lead to a decrease in vigilance and cognitive performance, increasing the likelihood of errors during the execution of attention-demanding activities such as piloting an aircraft or performing medical procedures. Thus, a method to rapidly and objectively assess the risk for such cognitive fatigue would be of value. The objective of the study was the identification in saliva-borne exosomes of molecular signals associated with changes in mood and fatigue that may increase the risk of reduced cognitive performance. Using integrated multiomics analysis of exosomes from the saliva of medical residents before and after a 12 h work shift, we observed changes in the abundances of several proteins and miRNAs that were associated with various mood states, and specifically fatigue, as determined by a Profile of Mood States questionnaire. The findings herein point to a promising protein biomarker, phosphoglycerate kinase 1 (PGK1), that was associated with fatigue and displayed changes in abundance in saliva, and we suggest a possible biological mechanism whereby the expression of the PGK1 gene is regulated by miR3185 in response to fatigue. Overall, these data suggest that multiomics analysis of salivary exosomes has merit for identifying novel biomarkers associated with changes in mood states and fatigue. The promising biomarker protein presents an opportunity for the development of a rapid saliva-based test for the assessment of these changes.


Assuntos
Exossomos , MicroRNAs , Biomarcadores/metabolismo , Exossomos/genética , Exossomos/metabolismo , MicroRNAs/metabolismo , Saliva/metabolismo
2.
Anal Biochem ; 596: 113636, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32081619

RESUMO

A procedure is described to measure curcumin (C), demethoxycurcumin (DMC), bisdemethoxycurcumin (BDMC), tetrahydrocurcumim (TC) and their glucuronidated metabolites (CG, DMCG, and BDMCG) in plasma, brain, liver and tumor samples. The procedure involves converting the analytes to their boron difluoride derivatives and analyzing them by combined liquid chromatography coupled to an ion trap mass spectrometer operating in the negative ion MSn scan mode. The method has superb limits of detection of 0.01 nM for all curcuminoids and 0.5 nM for TC and the glucuroniated metabolites, and several representative chromatograms of biological samples containing these analytes are provided. In addition, the pharmacokinetic profile of these compounds in one human who daily consumed an over-the-counter curcuminoid product shows the peak and changes in circulating concentrations achieved by this mode of administration.


Assuntos
Boranos/química , Diarileptanoides/sangue , Animais , Cromatografia Líquida , Diarileptanoides/química , Diarileptanoides/isolamento & purificação , Voluntários Saudáveis , Humanos , Espectrometria de Massas , Camundongos , Estrutura Molecular
3.
J Neurosci ; 34(21): 7124-36, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24849348

RESUMO

Hyperphosphorylation and accumulation of tau aggregates are prominent features in tauopathies, including Alzheimer's disease, but the impact of loss of tau function on synaptic and cognitive deficits remains poorly understood. We report that old (19-20 months; OKO) but not middle-aged (8-9 months; MKO) tau knock-out mice develop Morris Water Maze (MWM) deficits and loss of hippocampal acetylated α-tubulin and excitatory synaptic proteins. Mild motor deficits and reduction in tyrosine hydroxylase (TH) in the substantia nigra were present by middle age, but did not affect MWM performance, whereas OKO mice showed MWM deficits paralleling hippocampal deficits. Deletion of tau, a microtubule-associated protein (MAP), resulted in increased levels of MAP1A, MAP1B, and MAP2 in MKO, followed by loss of MAP2 and MAP1B in OKO. Hippocampal synaptic deficits in OKO mice were partially corrected with dietary supplementation with docosahexaenoic acid (DHA) and both MWM and synaptic deficits were fully corrected by combining DHA with α-lipoic acid (ALA), which also prevented TH loss. DHA or DHA/ALA restored phosphorylated and total GSK3ß and attenuated hyperactivation of the tau C-Jun N-terminal kinases (JNKs) while increasing MAP1B, dephosphorylated (active) MAP2, and acetylated α-tubulin, suggesting improved microtubule stability and maintenance of active compensatory MAPs. Our results implicate the loss of MAP function in age-associated hippocampal deficits and identify a safe dietary intervention, rescuing both MAP function and TH in OKO mice. Therefore, in addition to microtubule-stabilizing therapeutic drugs, preserving or restoring compensatory MAP function may be a useful new prevention strategy.


Assuntos
Envelhecimento/patologia , Hipocampo/patologia , Aprendizagem em Labirinto/fisiologia , Sinapses/metabolismo , Proteínas tau/deficiência , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipocampo/efeitos dos fármacos , Deficiências da Aprendizagem/dietoterapia , Deficiências da Aprendizagem/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtornos dos Movimentos/dietoterapia , Transtornos dos Movimentos/etiologia , Desempenho Psicomotor/fisiologia , Tempo de Reação/efeitos dos fármacos , Substância Negra/metabolismo , Substância Negra/patologia , Sinapses/efeitos dos fármacos , Sinapses/genética , Ácido Tióctico/administração & dosagem
4.
J Biol Chem ; 288(6): 4056-65, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23264626

RESUMO

The mechanisms underlying Tau-related synaptic and cognitive deficits and the interrelationships between Tau species, their clearance pathways, and synaptic impairments remain poorly understood. To gain insight into these mechanisms, we examined these interrelationships in aged non-mutant genomic human Tau mice, with established Tau pathology and neuron loss. We also examined how these interrelationships changed with an intervention by feeding mice either a control diet or one containing the brain permeable beta-amyloid and Tau aggregate binding molecule curcumin. Transgene-dependent elevations in soluble and insoluble phospho-Tau monomer and soluble Tau dimers accompanied deficits in behavior, hippocampal excitatory synaptic markers, and molecular chaperones (heat shock proteins (HSPs)) involved in Tau degradation and microtubule stability. In human Tau mice but not control mice, HSP70, HSP70/HSP72, and HSP90 were reduced in membrane-enriched fractions but not in cytosolic fractions. The synaptic proteins PSD95 and NR2B were reduced in dendritic fields and redistributed into perikarya, corresponding to changes observed by immunoblot. Curcumin selectively suppressed levels of soluble Tau dimers, but not of insoluble and monomeric phospho-Tau, while correcting behavioral, synaptic, and HSP deficits. Treatment increased PSD95 co-immunoprecipitating with NR2B and, independent of transgene, increased HSPs implicated in Tau clearance. It elevated HSP90 and HSC70 without increasing HSP mRNAs; that is, without induction of the heat shock response. Instead curcumin differentially impacted HSP90 client kinases, reducing Fyn without reducing Akt. In summary, curcumin reduced soluble Tau and elevated HSPs involved in Tau clearance, showing that even after tangles have formed, Tau-dependent behavioral and synaptic deficits can be corrected.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Comportamento Animal/efeitos dos fármacos , Curcumina/farmacologia , Proteínas de Choque Térmico/metabolismo , Multimerização Proteica/efeitos dos fármacos , Sinapses/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Proteína 4 Homóloga a Disks-Large , Feminino , Proteínas de Choque Térmico/genética , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Multimerização Proteica/genética , Proteólise/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Solubilidade/efeitos dos fármacos , Sinapses/genética , Sinapses/patologia , Tauopatias/tratamento farmacológico , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética
5.
Dement Geriatr Cogn Disord ; 33(1): 1-5, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22343824

RESUMO

BACKGROUND/AIMS: Biological markers of utility in tracking Alzheimer's disease (AD) during the presymptomatic prodromal phase are important for prevention studies. Changes in cerebrospinal fluid (CSF) levels of 42-amino-acid ß-amyloid (Aß(42)), total tau protein (t-tau) and phosphorylated tau at residue 181 (p-tau(181)) during this state are incompletely characterized. METHODS: We measured CSF markers in 13 carriers of familial AD (FAD) mutations that are fully penetrant for causing AD (PSEN1 and APP) and in 5 non-mutation-carrying family members. RESULTS: Even among the entirely presymptomatic mutation carriers (n = 9), Aß(42) was diminished (388.7 vs. 618.4 pg/ml, p = 0.004), and t-tau (138.5 vs. 50.5 pg/ml, p = 0.002) and p-tau(181) (71.7 vs. 24.6 pg/ml, p = 0.003) were elevated. There was a negative correlation between Aß(42) levels and age relative to the family-specific age of dementia diagnosis. CONCLUSIONS: Our data are consistent with a decline in CSF Aß(42) levels occurring at least 20 years prior to clinical dementia in FAD.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/genética , Fatores Etários , Idoso , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide/genética , Apolipoproteínas E/genética , Biomarcadores , Estudos de Coortes , DNA/genética , Diagnóstico Precoce , Feminino , Genótipo , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Presenilina-1/genética , Proteínas tau/líquido cefalorraquidiano
6.
Neuron ; 51(6): 671-2, 2006 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16982408

RESUMO

The role of cathepsin B, a lysosomal protease implicated in amyloid-beta (Abeta1-42) metabolism, in Alzheimer's disease remains controversial. In this issue of Neuron, Mueller-Steiner et al. manipulate the expression of cathepsin B in aged APP transgenic mice, observing that increased expression degrades preformed oligomeric and fibrillar amyloid, while inactivation accelerates beta-amyloidosis.


Assuntos
Doença de Alzheimer/metabolismo , Amiloide/metabolismo , Catepsina B/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Catepsina B/genética , Humanos , Hidrólise , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Mutação/genética , Neuritos/metabolismo , Neuritos/patologia , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patologia
7.
J Neurosci ; 29(28): 9078-89, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19605645

RESUMO

Both insulin resistance (type II diabetes) and beta-amyloid (Abeta) oligomers are implicated in Alzheimer's disease (AD). Here, we investigate the role of Abeta oligomer-induced c-Jun N-terminal kinase (JNK) activation leading to phosphorylation and degradation of the adaptor protein insulin receptor substrate-1 (IRS-1). IRS-1 couples insulin and other trophic factor receptors to downstream kinases and neuroprotective signaling. Increased phospho-IRS-1 is found in AD brain and insulin-resistant tissues from diabetics. Here, we report Abeta oligomers significantly increased active JNK and phosphorylation of IRS-1 (Ser616) and tau (Ser422) in cultured hippocampal neurons, whereas JNK inhibition blocked these responses. The omega-3 fatty acid docosahexaenoic acid (DHA) similarly inhibited JNK and the phosphorylation of IRS-1 and tau in cultured hippocampal neurons. Feeding 3xTg-AD transgenic mice a diet high in saturated and omega-6 fat increased active JNK and phosphorylated IRS-1 and tau. Treatment of the 3xTg-AD mice on high-fat diet with fish oil or curcumin or a combination of both for 4 months reduced phosphorylated JNK, IRS-1, and tau and prevented the degradation of total IRS-1. This was accompanied by improvement in Y-maze performance. Mice fed with fish oil and curcumin for 1 month had more significant effects on Y-maze, and the combination showed more significant inhibition of JNK, IRS-1, and tau phosphorylation. These data indicate JNK mediates Abeta oligomer inactivation of IRS-1 and phospho-tau pathology and that dietary treatment with fish oil/DHA, curcumin, or a combination of both has the potential to improve insulin/trophic signaling and cognitive deficits in AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/farmacologia , Curcumina/farmacologia , Inibidores Enzimáticos/farmacologia , Ácidos Graxos Ômega-3/farmacologia , Proteínas Substratos do Receptor de Insulina/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/dietoterapia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Comportamento Animal , Células Cultivadas , Curcumina/uso terapêutico , Modelos Animais de Doenças , Embrião de Mamíferos , Inibidores Enzimáticos/uso terapêutico , Ácidos Graxos Ômega-3/uso terapêutico , Hipocampo/citologia , Humanos , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Fosforilação/efeitos dos fármacos , Mudanças Depois da Morte , Presenilina-1/genética , Ratos , Ratos Sprague-Dawley , Serina/metabolismo
8.
J Nutr ; 140(4): 869-74, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20181786

RESUMO

The risk for dementia, a major contributor to incapacitation and institutionalization, rises rapidly as we age, doubling every 5 y after age 65. Tens of millions of new Alzheimer's disease (AD) and other dementia cases are projected as elderly populations increase around the world, creating a projected dementia epidemic for which most nations are not prepared. Thus, there is an urgent need for prevention approaches that are safe, effective, and affordable. This review addresses the potential of one promising candidate, the (n-3) fatty acid docosahexaenoic acid (DHA), which appears to slow pathogenesis of AD and possibly vascular dementia. DHA is pleiotropic, acting at multiple steps to reduce the production of the beta-amyloid peptide, widely believed to initiate AD. DHA moderates some of the kinases that hyperphosphorylate the tau-protein, a component of the neurofibrillary tangle. DHA may help suppress insulin/neurotrophic factor signaling deficits, neuroinflammation, and oxidative damage that contribute to synaptic loss and neuronal dysfunction in dementia. Finally, DHA increases brain levels of neuroprotective brain-derived neurotrophic factor and reduces the (n-6) fatty acid arachidonate and its prostaglandin metabolites that have been implicated in promoting AD. Clinical trials suggest that DHA or fish oil alone can slow early stages of progression, but these effects may be apolipoprotein E genotype specific, and larger trials with very early stages are required to prove efficacy. We advocate early intervention in a prodromal period with nutrigenomically defined subjects with an appropriately designed nutritional supplement, including DHA and antioxidants.


Assuntos
Envelhecimento , Demência/prevenção & controle , Ácidos Docosa-Hexaenoicos/administração & dosagem , Idoso , Demência/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Humanos
9.
Nat Neurosci ; 9(2): 234-42, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16415866

RESUMO

Defects in dendritic spines are common to several forms of cognitive deficits, including mental retardation and Alzheimer disease. Because mutation of p21-activated kinase (PAK) can lead to mental retardation and because PAK-cofilin signaling is critical in dendritic spine morphogenesis and actin dynamics, we hypothesized that the PAK pathway is involved in synaptic and cognitive deficits in Alzheimer disease. Here, we show that PAK and its activity are markedly reduced in Alzheimer disease and that this is accompanied by reduced and redistributed phosphoPAK, prominent cofilin pathology and downstream loss of the spine actin-regulatory protein drebrin, which cofilin removes from actin. We found that beta-amyloid (Abeta) was directly involved in PAK signaling deficits and drebrin loss in Abeta oligomer-treated hippocampal neurons and in the Appswe transgenic mouse model bearing a double mutation leading to higher Abeta production. In addition, pharmacological PAK inhibition in adult mice was sufficient to cause similar cofilin pathology, drebrin loss and memory impairment, consistent with a potential causal role of PAK defects in cognitive deficits in Alzheimer disease.


Assuntos
Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Animais , Células Cultivadas , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/patologia , Espinhas Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neuropeptídeos/metabolismo , Ratos , Quinases Ativadas por p21
10.
Neurobiol Dis ; 33(2): 193-206, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19038340

RESUMO

The dysregulation of glycogen synthase kinase-3 (GSK3) has been implicated in Alzheimer disease (AD) pathogenesis and in Abeta-induced neurotoxicity, leading us to investigate it as a therapeutic target in an intracerebroventricular Abeta infusion model. Infusion of a specific GSK3 inhibitor SB216763 (SB) reduced a downstream target, phospho-glycogen synthase 39%, and increased glycogen levels 44%, suggesting effective inhibition of enzyme activity. Compared to vehicle, Abeta increased GSK3 activity, and was associated with elevations in levels of ptau, caspase-3, the tau kinase phospho-c-jun N-terminal kinase (pJNK), neuronal DNA fragmentation, and gliosis. Co-infusion of SB corrected all responses to Abeta infusion except the induction of gliosis and behavioral deficits in the Morris water maze. Nevertheless, SB alone was associated with induction of neurodegenerative markers and behavioral deficits. These data support a role for GSK3 hyperactivation in AD pathogenesis, but emphasize the importance of developing inhibitors that do not suppress constitutive activity.


Assuntos
Doença de Alzheimer/terapia , Inibidores Enzimáticos/uso terapêutico , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Indóis/uso terapêutico , Maleimidas/uso terapêutico , Doença de Alzheimer/induzido quimicamente , Peptídeos beta-Amiloides/farmacologia , Animais , Caspase 3/metabolismo , Células Cultivadas , Fragmentação do DNA , Modelos Animais de Doenças , Inibidores Enzimáticos/efeitos adversos , Gliose/induzido quimicamente , Glicogênio/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Indóis/efeitos adversos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Maleimidas/efeitos adversos , Aprendizagem em Labirinto , Degeneração Neural/tratamento farmacológico , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Proteínas tau/metabolismo
11.
N Engl J Med ; 355(25): 2652-63, 2006 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-17182990

RESUMO

BACKGROUND: Amyloid senile plaques and tau neurofibrillary tangles are neuropathological hallmarks of Alzheimer's disease that accumulate in the cortical regions of the brain in persons with mild cognitive impairment who are at risk for Alzheimer's disease. Noninvasive methods to detect these abnormal proteins are potentially useful in developing surrogate markers for drug discovery and diagnostics. METHODS: We enrolled 83 volunteers with self-reported memory problems who had undergone neurologic and psychiatric evaluation and positron-emission tomography (PET). On the basis of cognitive testing, 25 volunteers were classified as having Alzheimer's disease, 28 as having mild cognitive impairment, and 30 as having no cognitive impairment (healthy controls). PET was performed after injection of 2-(1-{6-[(2-[F-18]fluoroethyl)(methyl)amino]-2-naphthyl}ethylidene)malononitrile (FDDNP), a molecule that binds to plaques and tangles in vitro. All subjects also underwent 2-deoxy-2-[F-18]fluoro-D-glucose (FDG) PET, and 72 underwent magnetic resonance imaging (MRI). RESULTS: Global values for FDDNP-PET binding (average of the values for the temporal, parietal, posterior cingulate, and frontal regions) were lower in the control group than in the group with mild cognitive impairment (P<0.001), and the values for binding in the group with mild cognitive impairment were lower than in the group with Alzheimer's disease (P<0.001). FDDNP-PET binding differentiated among the diagnostic groups better than did metabolism on FDG-PET or volume on MRI. CONCLUSIONS: FDDNP-PET scanning can differentiate persons with mild cognitive impairment from those with Alzheimer's disease and those with no cognitive impairment. This technique is potentially useful as a noninvasive method to determine regional cerebral patterns of amyloid plaques and tau neurofibrillary tangles.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Transtornos Cognitivos/diagnóstico por imagem , Emaranhados Neurofibrilares/diagnóstico por imagem , Placa Amiloide/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Transtornos Cognitivos/diagnóstico , Transtornos Cognitivos/metabolismo , Diagnóstico Diferencial , Feminino , Fluordesoxiglucose F18/metabolismo , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Nitrilas/metabolismo , Proteínas tau
12.
Neuron ; 37(6): 889-90, 2003 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-12670416

RESUMO

In this issue of Neuron, Kaur et al. demonstrate that iron chelation by ferritin transgene or the metal chelator clioquinol prevent oxidative damage and MPTP toxicity in mice. This raises the issue of specific iron chelators or clioquinol for control of oxidative damage in Parkinson's, Alzheimer's, and other neurodegenerative diseases, but not without safety concerns.


Assuntos
Clioquinol/uso terapêutico , Ferro/metabolismo , Legislação de Medicamentos , Doenças Neurodegenerativas/prevenção & controle , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Doença de Alzheimer/prevenção & controle , Animais , Ferritinas/genética , Expressão Gênica , Humanos , Quelantes de Ferro/uso terapêutico , Camundongos , Camundongos Transgênicos , Oxirredução , Doença de Parkinson/prevenção & controle , Substância Negra/metabolismo
13.
Neuron ; 43(5): 633-45, 2004 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-15339646

RESUMO

Learning and memory depend on dendritic spine actin assembly and docosahexaenoic acid (DHA), an essential n-3 (omega-3) polyunsaturated fatty acid (PFA). High DHA consumption is associated with reduced Alzheimer's disease (AD) risk, yet mechanisms and therapeutic potential remain elusive. Here, we report that reduction of dietary n-3 PFA in an AD mouse model resulted in 80%-90% losses of the p85alpha subunit of phosphatidylinositol 3-kinase and the postsynaptic actin-regulating protein drebrin, as in AD brain. The loss of postsynaptic proteins was associated with increased oxidation, without concomitant neuron or presynaptic protein loss. n-3 PFA depletion increased caspase-cleaved actin, which was localized in dendrites ultrastructurally. Treatment of n-3 PFA-restricted mice with DHA protected against these effects and behavioral deficits and increased antiapoptotic BAD phosphorylation. Since n-3 PFAs are essential for p85-mediated CNS insulin signaling and selective protection of postsynaptic proteins, these findings have implications for neurodegenerative diseases where synaptic loss is critical, especially AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Encéfalo/metabolismo , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Actinas/metabolismo , Doença de Alzheimer/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Dendritos/patologia , Gorduras Insaturadas na Dieta/metabolismo , Gorduras Insaturadas na Dieta/farmacologia , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/metabolismo , Ácidos Docosa-Hexaenoicos/uso terapêutico , Regulação para Baixo/genética , Feminino , Alimentos Formulados , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Sinapses/patologia , Proteína de Morte Celular Associada a bcl
14.
J Neurosci ; 27(52): 14299-307, 2007 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18160637

RESUMO

Environmental and genetic factors, notably ApoE4, contribute to the etiology of late-onset Alzheimer's disease (LOAD). Reduced mRNA and protein for an apolipoprotein E (ApoE) receptor family member, SorLA (LR11) has been found in LOAD but not early-onset AD, suggesting that LR11 loss is not secondary to pathology. LR11 is a neuronal sorting protein that reduces amyloid precursor protein (APP) trafficking to secretases that generate beta-amyloid (Abeta). Genetic polymorphisms that reduce LR11 expression are associated with increased AD risk. However these polymorphisms account for only a fraction of cases with LR11 deficits, suggesting involvement of environmental factors. Because lipoprotein receptors are typically lipid-regulated, we postulated that LR11 is regulated by docosahexaenoic acid (DHA), an essential omega-3 fatty acid related to reduced AD risk and reduced Abeta accumulation. In this study, we report that DHA significantly increases LR11 in multiple systems, including primary rat neurons, aged non-Tg mice and an aged DHA-depleted APPsw AD mouse model. DHA also increased LR11 in a human neuronal line. In vivo elevation of LR11 was also observed with dietary fish oil in young rats with insulin resistance, a model for type II diabetes, another AD risk factor. These data argue that DHA induction of LR11 does not require DHA-depleting diets and is not age dependent. Because reduced LR11 is known to increase Abeta production and may be a significant genetic cause of LOAD, our results indicate that DHA increases in SorLA/LR11 levels may play an important role in preventing LOAD.


Assuntos
Doença de Alzheimer/dietoterapia , Doença de Alzheimer/metabolismo , Ácidos Docosa-Hexaenoicos/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Receptores de LDL/metabolismo , Fatores Etários , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Humanos , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Neuroblastoma , Ratos , Ratos Sprague-Dawley , Receptores de LDL/genética , Fatores de Tempo
15.
Lancet Neurol ; 7(2): 161-72, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18207114

RESUMO

Technological advances have led to greater use of both structural and functional brain imaging to assist with the diagnosis of dementia for the increasing numbers of people with cognitive decline as they age. In current clinical practice, structural imaging (CT or MRI) is used to identify space-occupying lesions and stroke. Functional methods, such as PET scanning of glucose metabolism, could be used to differentiate Alzheimer's disease from frontotemporal dementia, which helps to guide clinicians in symptomatic treatment strategies. New neuroimaging methods that are currently being developed can measure specific neurotransmitter systems, amyloid plaque and tau tangle concentrations, and neuronal integrity and connectivity. Successful co-development of neuroimaging surrogate markers and preventive treatments might eventually lead to so-called brain-check scans for determining risk of cognitive decline, so that physicians can administer disease-modifying medications, vaccines, or other interventions to avoid future cognitive losses and to delay onset of disease.


Assuntos
Transtornos Cognitivos/diagnóstico por imagem , Idoso , Envelhecimento/fisiologia , Biomarcadores , Demência/diagnóstico por imagem , Diagnóstico por Imagem , Imagem de Difusão por Ressonância Magnética , Humanos , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/prevenção & controle , Tomografia por Emissão de Pósitrons , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
16.
J Pharmacol Exp Ther ; 326(1): 196-208, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18417733

RESUMO

Curcumin can reduce inflammation and neurodegeneration, but its chemical instability and metabolism raise concerns, including whether the more stable metabolite tetrahydrocurcumin (TC) may mediate efficacy. We examined the antioxidant, anti-inflammatory, or anti-amyloidogenic effects of dietary curcumin and TC, either administered chronically to aged Tg2576 APPsw mice or acutely to lipopolysaccharide (LPS)-injected wild-type mice. Despite dramatically higher drug plasma levels after TC compared with curcumin gavage, resulting brain levels of parent compounds were similar, correlating with reduction in LPS-stimulated inducible nitric-oxide synthase, nitrotyrosine, F2 isoprostanes, and carbonyls. In both the acute (LPS) and chronic inflammation (Tg2576), TC and curcumin similarly reduced interleukin-1beta. Despite these similarities, only curcumin was effective in reducing amyloid plaque burden, insoluble beta-amyloid peptide (Abeta), and carbonyls. TC had no impact on plaques or insoluble Abeta, but both reduced Tris-buffered saline-soluble Abeta and phospho-c-Jun NH(2)-terminal kinase (JNK). Curcumin but not TC prevented Abeta aggregation. The TC metabolite was detected in brain and plasma from mice chronically fed the parent compound. These data indicate that the dienone bridge present in curcumin, but not in TC, is necessary to reduce plaque deposition and protein oxidation in an Alzheimer's model. Nevertheless, TC did reduce neuroinflammation and soluble Abeta, effects that may be attributable to limiting JNK-mediated transcription. Because of its favorable safety profile and the involvement of misfolded proteins, oxidative damage, and inflammation in multiple chronic degenerative diseases, these data relating curcumin dosing to the blood and tissue levels required for efficacy should help translation efforts from multiple successful preclinical models.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Curcumina/metabolismo , Curcumina/uso terapêutico , Modelos Animais de Doenças , Doença de Alzheimer/patologia , Animais , Disponibilidade Biológica , Curcumina/química , Feminino , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Relação Estrutura-Atividade
17.
J Alzheimers Dis ; 15(4): 625-40, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19096161

RESUMO

The rat amyloid-beta (Abeta) intracerebroventricular infusion can model aspects of Alzheimer's disease (AD) and has predicted efficacy of therapies such as ibuprofen and curcumin in transgenic mouse models. High density lipoprotein (HDL), a normal plasma carrier of Abeta, is used to attenuate Abeta aggregation within the pump, causing Abeta-dependent toxicity and cognitive deficits within 3 months. Our goal was to identify factors that might accelerate onset of Abeta-dependent deficits to improve efficiency and cost-effectiveness of model. We focused on: 1) optimizing HDL-Abeta preparation for maximal toxicity; 2) evaluating the role of copper, a factor typically in water that can impact oligomer stability; and 3) determining impact of insulin resistance (type II diabetes), a risk factor for AD. In vitro studies were performed to determine doses of copper and methods of Abeta-HDL preparation that maximized toxicity. These preparations when infused resulted in earlier onset of cognitive deficits within 6 weeks post-infusion. Induction of insulin resistance did not exacerbate Abeta-dependent cognitive deficits, but did exacerbate synaptic protein loss. In summary, the newly described in vivo infusion model may be useful cost-effective method for screening for new therapeutic drugs for AD.


Assuntos
Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/genética , Transtornos Cognitivos/psicologia , Cobre/toxicidade , Resistência à Insulina/genética , Proteínas do Tecido Nervoso/metabolismo , Sinapses/metabolismo , Doença de Alzheimer/patologia , Animais , Western Blotting , Células Cultivadas , Transtornos Cognitivos/patologia , Dieta , Frutose/farmacologia , Resistência à Insulina/fisiologia , Aprendizagem em Labirinto/fisiologia , Osmose , Ratos , Ratos Sprague-Dawley
18.
J Neurochem ; 103(4): 1594-607, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17760871

RESUMO

Extracellular-signal regulated kinase (ERK) signaling is critical for memory and tightly regulated by acute environmental stimuli. In Alzheimer disease transgenic models, active ERK is shown to first be increased, then later reduced, but whether these baseline changes reflect disruptions in ERK signaling is less clear. We investigated the influence of the familial Alzheimer's disease transgene APPsw and beta-amyloid peptide (Abeta) immunoneutralization on cannulation injury-associated (i.c.v. infusion) ERK activation. At both 12 and 22 months of age, the trauma-associated activation of ERK observed in Tg(-) mice was dramatically attenuated in Tg(+). In cortices of 22-month-old non-infused mice, a reduction in ERK activation was observed in Tg(+), relative to Tg(-) mice. Intracerebroventricular (i.c.v.) anti-Abeta infusion significantly increased phosphorylated ERK, its substrate cAMP-response element-binding protein (CREB) and a downstream target, the NMDA receptor subunit. We also demonstrated that Abeta oligomer decreased active ERK and subsequently active CREB in human neuroblastoma cells, which could be prevented by oligomer immunoneutralization. Abeta oligomers also inhibited active ERK and CREB in primary neurons, in addition to reducing the downstream post-synaptic protein NMDA receptor subunit. These effects were reversed by anti-oligomer. Our data strongly support the existence of an APPsw transgene-dependent and Abeta oligomer-mediated defect in regulation of ERK activation.


Assuntos
Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/fisiologia , Proteína de Ligação a CREB/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Sistema de Sinalização das MAP Quinases/genética , Transgenes/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Proteína de Ligação a CREB/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ativação Enzimática/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Humanos , Camundongos , Camundongos Transgênicos
19.
Trends Mol Med ; 12(9): 395-6, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16880006

RESUMO

Beta-amyloid peptides (Abeta) have been genetically implicated as the cause of Alzheimer's disease, but the causality of amyloid deposited as plaques has been challenged. The controversial role of amyloid peptides in Alzheimer's disease has been highlighted in a recent paper from Lesne and colleagues, who applied Koch's postulates to cast a specific memory-deficit-inducing oligomer species as a central player causing memory loss. These authors used a transgenic mouse model to identify a specific type of aggregate that emerges with cognitive deficits and is capable of transmitting a spatial memory defect to unimpaired animals.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transtornos da Memória/metabolismo , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/química , Animais , Modelos Animais de Doenças , Humanos , Transtornos da Memória/psicologia , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/química
20.
Exp Gerontol ; 42(1-2): 10-21, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17049785

RESUMO

Although increased lifespan is associated with reduced insulin signaling, insulin signaling is essential for neuronal development and survival. Insulin resistance is central to Type II diabetes and is also implicated in the pathogenesis of Alzheimer's Disease (AD). This has prompted ongoing clinical trials in AD patients to test the efficacy of improving insulin - like signaling with dietary omega-3 fatty acids or insulin - sensitizing drugs as well as exercise regimens. Here we review the role of insulin signaling in brain aging and AD, concluding that the signaling pathways downstream to neurotrophic and insulin signaling are defective and coincident with aberrant phosphorylation and translocation of key components, notably AKT and GSK3beta, but also rac> PAK signaling. These responses are likely to contribute to defects in synaptic plasticity, learning and memory. Both oligomers of beta-amyloid (which are elevated in the AD brain) and pro-inflammatory cytokines (which are elevated in the aged or AD brain) can be used to mimic the trophic factor/insulin resistance observed in AD, but details on other factors and mechanisms contributing to this resistance remain elusive. A better understanding of the precise mechanisms underlying alterations in the insulin/neurotrophic factor signal transduction pathways should aid the search for better AD therapeutic and prevention strategies.


Assuntos
Envelhecimento/fisiologia , Doença de Alzheimer/metabolismo , Encéfalo/fisiologia , Insulina/fisiologia , Neurotrofina 3/fisiologia , Transdução de Sinais/fisiologia , Idoso , Humanos , Resistência à Insulina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA