Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 111(3): 1144-9, 2014 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-24395787

RESUMO

Parkinsonism-dementia (PD) of Guam is a neurodegenerative disease with parkinsonism and early-onset Alzheimer-like dementia associated with neurofibrillary tangles composed of hyperphosphorylated microtubule-associated protein, tau. ß-N-methylamino-l-alanine (BMAA) has been suspected of being involved in the etiology of PD, but the mechanism by which BMAA leads to tau hyperphosphorylation is not known. We found a decrease in protein phosphatase 2A (PP2A) activity associated with an increase in inhibitory phosphorylation of its catalytic subunit PP2Ac at Tyr(307) and abnormal hyperphosphorylation of tau in brains of patients who had Guam PD. To test the possible involvement of BMAA in the etiopathogenesis of PD, we studied the effect of this environmental neurotoxin on PP2A activity and tau hyperphosphorylation in mouse primary neuronal cultures and metabolically active rat brain slices. BMAA treatment significantly decreased PP2A activity, with a concomitant increase in tau kinase activity resulting in elevated tau hyperphosphorylation at PP2A favorable sites. Moreover, we found an increase in the phosphorylation of PP2Ac at Tyr(307) in BMAA-treated rat brains. Pretreatment with metabotropic glutamate receptor 5 (mGluR5) and Src antagonists blocked the BMAA-induced inhibition of PP2A and the abnormal hyperphosphorylation of tau, indicating the involvement of an Src-dependent PP2A pathway. Coimmunoprecipitation experiments showed that BMAA treatment dissociated PP2Ac from mGluR5, making it available for phosphorylation at Tyr(307). These findings suggest a scenario in which BMAA can lead to tau pathology by inhibiting PP2A through the activation of mGluR5, the consequent release of PP2Ac from the mGluR5-PP2A complex, and its phosphorylation at Tyr(307) by Src.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Regulação Enzimológica da Expressão Gênica , Proteína Fosfatase 2/metabolismo , Proteínas tau/metabolismo , Idoso , Animais , Encéfalo/enzimologia , Feminino , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Neurônios/metabolismo , Neurotoxinas/metabolismo , Fosforilação , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptor de Glutamato Metabotrópico 5/metabolismo , Fatores de Tempo , Tirosina/química , Quinases da Família src/metabolismo
2.
J Biol Chem ; 289(40): 27677-91, 2014 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-25128526

RESUMO

Abnormal hyperphosphorylation of Tau leads to the formation of neurofibrillary tangles, a hallmark of Alzheimer disease (AD), and related tauopathies. The phosphorylation of Tau is regulated by protein phosphatase 2A (PP2A), which in turn is modulated by endogenous inhibitor 2 (I2 (PP2A)). In AD brain, I2 (PP2A) is translocated from neuronal nucleus to cytoplasm, where it inhibits PP2A activity and promotes abnormal phosphorylation of Tau. Here we describe the identification of a potential nuclear localization signal (NLS) in the C-terminal region of I2 (PP2A) containing a conserved basic motif, (179)RKR(181), which is sufficient for directing its nuclear localization. The current study further presents an inducible cell model (Tet-Off system) of AD-type abnormal hyperphosphorylation of Tau by expressing I2 (PP2A) in which the NLS was inactivated by (179)RKR(181) → AAA along with (168)KR(169) → AA mutations. In this model, the mutant NLS (mNLS)-I2 (PP2A) (I2 (PP2A)AA-AAA) was retained in the cell cytoplasm, where it physically interacted with PP2A and inhibited its activity. Inhibition of PP2A was associated with the abnormal hyperphosphorylation of Tau, which resulted in microtubule network instability and neurite outgrowth impairment. Expression of mNLS-I2 (PP2A) activated CAMKII and GSK-3ß, which are Tau kinases regulated by PP2A. The immunoprecipitation experiments showed the direct interaction of I2 (PP2A) with PP2A and GSK-3ß but not with CAMKII. Thus, the cell model provides insights into the nature of the potential NLS and the mechanistic relationship between I2 (PP2A)-induced inhibition of PP2A and hyperphosphorylation of Tau that can be utilized to develop drugs preventing Tau pathology.


Assuntos
Doença de Alzheimer/metabolismo , Citoplasma/metabolismo , Chaperonas de Histonas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Encéfalo/metabolismo , Núcleo Celular/metabolismo , Citoplasma/genética , Proteínas de Ligação a DNA , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Chaperonas de Histonas/genética , Humanos , Sinais de Localização Nuclear , Fosforilação , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Fatores de Transcrição/genética , Proteínas tau/genética
3.
J Neural Transm (Vienna) ; 122(4): 607-17, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25233799

RESUMO

Intraneuronal accumulation of abnormally hyperphosphorylated tau in the brain is a histopathological hallmark of Alzheimer's disease and a family of related neurodegenerative disorders collectively called tauopathies. At present there is no effective treatment available for these progressive neurodegenerative diseases which are clinically characterized by dementia in mid to old-age. Here we report the treatment of 14-17-months-old 3xTg-AD mice with tau antibodies 43D (tau 6-18) and 77E9 (tau 184-195) to the N-terminal projection domain of tau or mouse IgG as a control by intraperitoneal injection once a week for 4 weeks, and the effects of the passive immunization on reduction of hyperphosphorylated tau, Aß accumulation and cognitive performance in these animals. We found that treatment with tau antibodies 43D and 77E9 reduced total tau level, decreased tau hyperphosphorylated at Ser199, Ser202/Thr205 (AT8), Thr205, Ser262/356 (12E8), and Ser396/404 (PHF-1) sites, and a trend to reduce Aß pathology. Most importantly, targeting N-terminal tau especially by 43D (tau 6-18) improved reference memory in the Morris water maze task in 3xTg-AD mice. We did not observe any abnormality in general physical characteristics of the treated animals with either of the two antibodies during the course of this study. Taken together, our studies demonstrate for the first time (1) that passive immunization targeting normal tau can effectively clear the hyperphosphorylated protein and possibly reduce Aß pathology from the brain and (2) that targeting N-terminal projection domain of tau containing amino acid 6-18 is especially beneficial. Thus, targeting selective epitopes of N-terminal domain of tau may present a novel effective therapeutic opportunity for Alzheimer disease and other tauopathies.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Transtornos Cognitivos/patologia , Transtornos Cognitivos/terapia , Imunização Passiva/métodos , Proteínas tau/imunologia , Doença de Alzheimer/complicações , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Anticorpos/administração & dosagem , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Feminino , Humanos , Aprendizagem em Labirinto , Camundongos Transgênicos , Fragmentos de Peptídeos/metabolismo , Fosforilação , Placa Amiloide/etiologia , Placa Amiloide/patologia , Placa Amiloide/terapia , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
4.
Nucleic Acids Res ; 41(5): 3240-56, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23341039

RESUMO

Impaired brain glucose uptake and metabolism precede the appearance of clinical symptoms in Alzheimer disease (AD). Neuronal glucose transporter 3 (GLUT3) is decreased in AD brain and correlates with tau pathology. However, what leads to the decreased GLUT3 is yet unknown. In this study, we found that the promoter of human GLUT3 contains three potential cAMP response element (CRE)-like elements, CRE1, CRE2 and CRE3. Overexpression of CRE-binding protein (CREB) or activation of cAMP-dependent protein kinase significantly increased GLUT3 expression. CREB bound to the CREs and promoted luciferase expression driven by human GLUT3-promoter. Among the CREs, CRE2 and CRE3 were required for the promotion of GLUT3 expression. Full-length CREB was decreased and truncation of CREB was increased in AD brain. This truncation was correlated with calpain I activation in human brain. Further study demonstrated that calpain I proteolysed CREB at Gln28-Ala29 and generated a 41-kDa truncated CREB, which had less activity to promote GLUT3 expression. Importantly, human brain GLUT3 was correlated with full-length CREB positively and with activation of calpain I negatively. These findings suggest that overactivation of calpain I caused by calcium overload proteolyses CREB, resulting in a reduction of GLUT3 expression and consequently impairing glucose uptake and metabolism in AD brain.


Assuntos
Doença de Alzheimer/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/fisiologia , Lobo Frontal/metabolismo , Regulação da Expressão Gênica , Transportador de Glucose Tipo 3/genética , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Calpaína/química , Calpaína/metabolismo , Estudos de Casos e Controles , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/química , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação para Baixo , Feminino , Genes Reporter , Transportador de Glucose Tipo 3/metabolismo , Células HEK293 , Humanos , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Masculino , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/fisiologia , Proteólise , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Elementos de Resposta , Transdução de Sinais
5.
J Biol Chem ; 288(24): 17495-507, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23640887

RESUMO

Neurofibrillary pathology of abnormally hyperphosphorylated Tau is a key lesion of Alzheimer disease and other tauopathies, and its density in the brain directly correlates with dementia. The phosphorylation of Tau is regulated by protein phosphatase 2A, which in turn is regulated by inhibitor 2, I2(PP2A). In acidic conditions such as generated by brain ischemia and hypoxia, especially in association with hyperglycemia as in diabetes, I2(PP2A) is cleaved by asparaginyl endopeptidase at Asn-175 into the N-terminal fragment (I2NTF) and the C-terminal fragment (I2CTF). Both I2NTF and I2CTF are known to bind to the catalytic subunit of protein phosphatase 2A and inhibit its activity. Here we show that the level of activated asparaginyl endopeptidase is significantly increased, and this enzyme and I2(PP2A) translocate, respectively, from neuronal lysosomes and nucleus to the cytoplasm where they interact and are associated with hyperphosphorylated Tau in Alzheimer disease brain. Asparaginyl endopeptidase from Alzheimer disease brain could cleave GST-I2(PP2A), except when I2(PP2A) was mutated at the cleavage site Asn-175 to Gln. Finally, an induction of acidosis by treatment with kainic acid or pH 6.0 medium activated asparaginyl endopeptidase and consequently produced the cleavage of I2(PP2A), inhibition of protein phosphatase 2A, and hyperphosphorylation of Tau, and the knockdown of asparaginyl endopeptidase with siRNA abolished this pathway in SH-SY5Y cells. These findings suggest the involvement of brain acidosis in the etiopathogenesis of Alzheimer disease, and asparaginyl endopeptidase-I2(PP2A)-protein phosphatase 2A-Tau hyperphosphorylation pathway as a therapeutic target.


Assuntos
Doença de Alzheimer/enzimologia , Cisteína Endopeptidases/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Células COS , Estudos de Casos e Controles , Chlorocebus aethiops , Cisteína Endopeptidases/química , Citoplasma/enzimologia , Ativação Enzimática , Feminino , Lobo Frontal/enzimologia , Hipocampo/enzimologia , Humanos , Concentração de Íons de Hidrogênio , Masculino , Fosforilação , Proteína Fosfatase 2/química , Proteína Fosfatase 2/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteólise , Ratos , Ratos Wistar
6.
Acta Neuropathol ; 127(2): 243-56, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24136402

RESUMO

The etiopathogenesis of neither the sporadic form of Alzheimer disease (AD) nor of amyotrophic lateral sclerosis (ALS) is well understood. The activity of protein phosphatase-2A (PP2A), which regulates the phosphorylation of tau and neurofilaments, is negatively regulated by the myeloid leukemia-associated protein SET, also known as inhibitor-2 of PP2A, I2(PP2A). In AD brain, PP2A activity is compromised, probably because I2(PP2A) is overexpressed and is selectively cleaved at asparagine 175 into an N-terminal fragment, I2NTF, and a C-terminal fragment, I2CTF, and both fragments inhibit PP2A. Here, we analyzed the spinal cords from ALS and control cases for I2(PP2A) cleavage and PP2A activity. As observed in AD brain, we found a selective increase in the cleavage of I2(PP2A) into I2NTF and I2CTF and inhibition of the activity and not the expression of PP2A in the spinal cords of ALS cases. To test the hypothesis that both AD and ALS could be triggered by I2CTF, a cleavage product of I2(PP2A), we transduced by intracerebroventricular injections newborn rats with adeno-associated virus serotype 1 (AAV1) containing human I2CTF. AAV1-I2CTF produced reference memory impairment and tau pathology, and intraneuronal accumulation of Aß by 5-8 months, and motor deficit and hyperphosphorylation and proliferation of neurofilaments, tau and TDP-43 pathologies, degeneration and loss of motor neurons and axons in the spinal cord by 10-14 months in rats. These findings suggest a previously undiscovered etiopathogenic relationship between sporadic forms of AD and ALS that is linked to I2(PP2A) and the potential of I2(PP2A)-based therapeutics for these diseases.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/patologia , Esclerose Lateral Amiotrófica/etiologia , Esclerose Lateral Amiotrófica/patologia , Proteína Fosfatase 2/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Proteínas de Ligação a DNA , Dependovirus/genética , Modelos Animais de Doenças , Chaperonas de Histonas/metabolismo , Humanos , Fosforilação , Ratos , Ratos Wistar , Medula Espinal/metabolismo , Medula Espinal/patologia , Fatores de Transcrição/metabolismo , Transdução Genética
7.
J Biol Chem ; 287(36): 30497-506, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22767602

RESUMO

Tau exon 10, which encodes the second microtubule-binding repeat, is regulated by alternative splicing. Its alternative splicing generates Tau isoforms with three- or four-microtubule-binding repeats, named 3R-tau and 4R-tau. Adult human brain expresses equal levels of 3R-tau and 4R-tau. Imbalance of 3R-tau and 4R-tau causes Tau aggregation and neurofibrillary degeneration. In the present study, we found that splicing factor SRp55 (serine/arginine-rich protein 55) promoted Tau exon 10 inclusion. Knockdown of SRp55 significantly promoted Tau exon 10 exclusion. The promotion of Tau exon 10 inclusion by SRp55 required the arginine/serine-rich region, which was responsible for the subnucleic speckle localization. Dyrk1A (dual specificity tyrosine-phosphorylated and regulated kinase 1A) interacted with SRp55 and mainly phosphorylated its proline-rich domain. Phosphorylation of SRp55 by Dyrk1A suppressed its ability to promote Tau exon 10 inclusion. Up-regulation of Dyrk1A as in Down syndrome could lead to neurofibrillary degeneration by shifting the alternative splicing of Tau exon 10 to an increase in the ratio of 3R-tau/4R-tau.


Assuntos
Processamento Alternativo , Núcleo Celular/metabolismo , Íntrons , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Tirosina Quinases/biossíntese , Proteínas de Ligação a RNA/metabolismo , Proteínas tau/metabolismo , Animais , Células COS , Núcleo Celular/genética , Núcleo Celular/patologia , Chlorocebus aethiops , Síndrome de Down/genética , Síndrome de Down/metabolismo , Síndrome de Down/patologia , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Proteínas Nucleares/genética , Fosfoproteínas/genética , Fosforilação/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Proteínas de Ligação a RNA/genética , Ratos , Fatores de Processamento de Serina-Arginina , Regulação para Cima/genética , Proteínas tau/genética , Quinases Dyrk
8.
Nucleic Acids Res ; 39(14): 6161-71, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21470964

RESUMO

Abnormal alternative splicing of tau exon 10 results in imbalance of 3R-tau and 4R-tau expression, which is sufficient to cause neurofibrillary degeneration. Splicing factor SC35, a member of the superfamily of the serine/arginine-rich (SR) proteins, promotes tau exon 10 inclusion. The molecular mechanism by which SC35 participates in tau exon 10 splicing remains elusive. In the present study, we found that tau pre-mRNA was coprecipitated by SC35 tagged with HA. Mutation of the SC35-like exonic splicing enhancer located at exon 10 of tau affected both the binding of SC35 to tau pre-mRNA and promotion of tau exon 10 inclusion, suggesting that SC35 acts on the SC35-like exonic splicing enhancer to promote tau exon 10 inclusion. Dyrk1A (dual-specificity tyrosine-phosphorylated and regulated kinase 1A) phosphorylated SC35 in vitro and interacted with it in cultured cells. Overexpression of Dyrk1A suppressed SC35's ability to promote tau exon 10 inclusion. Downregulation of Dyrk1A promoted 4R-tau expression. Therefore, upregulation of Dyrk1A in Down syndrome brain or Alzheimer's brain may cause dysregulation of tau exon 10 splicing through SC35, and probably together with other splicing factors, leading to the imbalance in 3R-tau and 4R-tau expression, which may initiate or accelerate tau pathology and cause neurofibrillary degeneration in the diseases.


Assuntos
Processamento Alternativo , Éxons , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas tau/genética , Animais , Sítios de Ligação , Linhagem Celular , Humanos , Precursores de RNA/metabolismo , RNA Mensageiro/metabolismo , Fatores de Processamento de Serina-Arginina , Proteínas tau/metabolismo , Quinases Dyrk
9.
J Biol Chem ; 286(16): 14639-48, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21367856

RESUMO

Hyperphosphorylation and deposition of tau into neurofibrillary tangles is a hallmark of Alzheimer disease (AD). Alternative splicing of tau exon 10 generates tau isoforms containing three or four microtubule binding repeats (3R-tau and 4R-tau), which are equally expressed in adult human brain. Dysregulation of exon 10 causes neurofibrillary degeneration. Here, we report that cyclic AMP-dependent protein kinase, PKA, phosphorylates splicing factor SRSF1, modulates its binding to tau pre-mRNA, and promotes tau exon 10 inclusion in cultured cells and in vivo in rat brain. PKA-Cα, but not PKA-Cß, interacts with SRSF1 and elevates SRSF1-mediated tau exon 10 inclusion. In AD brain, the decreased level of PKA-Cα correlates with the increased level of 3R-tau. These findings suggest that a down-regulation of PKA dysregulates the alternative splicing of tau exon 10 and contributes to neurofibrillary degeneration in AD by causing an imbalance in 3R-tau and 4R-tau expression.


Assuntos
Doença de Alzheimer/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação Enzimológica da Expressão Gênica , Proteínas tau/química , Processamento Alternativo , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Proteínas Quinases Dependentes de AMP Cíclico/química , Éxons , Células HeLa , Humanos , Emaranhados Neurofibrilares/química , Proteínas Nucleares/química , Fosforilação , Splicing de RNA , Proteínas de Ligação a RNA/química , Fatores de Processamento de Serina-Arginina , Proteínas tau/genética
10.
Acta Neuropathol ; 123(1): 133-51, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22083255

RESUMO

Alzheimer's disease (AD) is multifactorial and, to date, no single cause of the sporadic form of this disease, which accounts for over 99% of the cases, has been established. In AD brain, protein phosphatase-2A (PP2A) activity is known to be compromised due to the cleavage and translocation of its potent endogenous inhibitor, I2PP2A, from the neuronal nucleus to the cytoplasm. Here, we show that adeno-associated virus vector-induced expression of the N-terminal I2NTF and C-terminal I2CTF halves of I2PP2A , also called SET, in brain reproduced key features of AD in Wistar rats. The I2NTF-CTF rats showed a decrease in brain PP2A activity, abnormal hyperphosphorylation and aggregation of tau, a loss of neuronal plasticity and impairment in spatial reference and working memories. To test whether early pharmacologic intervention with a neurotrophic molecule could rescue neurodegeneration and behavioral deficits, 2.5-month-old I2NTF-CTF rats and control littermates were treated for 40 days with Peptide 6, an 11-mer peptide corresponding to an active region of the ciliary neurotrophic factor. Peripheral administration of Peptide 6 rescued neurodegeneration and cognitive deficit in I2NTF-CTF animals by increasing dentate gyrus neurogenesis and mRNA level of brain derived neurotrophic factor. Moreover, Peptide 6-treated I2NTF-CTF rats showed a significant increase in dendritic and synaptic density as reflected by increased expression of synapsin I, synaptophysin and MAP2, especially in the pyramidal neurons of CA1 and CA3 of the hippocampus.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Fator Neurotrófico Ciliar/uso terapêutico , Transtornos Cognitivos/tratamento farmacológico , Hipocampo/metabolismo , Neurogênese/efeitos dos fármacos , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/uso terapêutico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Masculino , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar , Proteínas tau/metabolismo
11.
J Pathol ; 225(1): 54-62, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21598254

RESUMO

Brain glucose metabolism is impaired in Alzheimer's disease (AD), the most common form of dementia. Type 2 diabetes mellitus (T2DM) is reported to increase the risk for dementia, including AD, but the underlying mechanism is not understood. Here, we investigated the brain insulin-PI3K-AKT signalling pathway in the autopsied frontal cortices from nine AD, 10 T2DM, eight T2DM-AD and seven control cases. We found decreases in the levels and activities of several components of the insulin-PI3K-AKT signalling pathway in AD and T2DM cases. The deficiency of insulin-PI3K-AKT signalling was more severe in individuals with both T2DM and AD (T2DM-AD). This decrease in insulin-PI3K-AKT signalling could lead to activation of glycogen synthase kinase-3ß, the major tau kinase. The levels and the activation of the insulin-PI3K-AKT signalling components correlated negatively with the level of tau phosphorylation and positively with protein O-GlcNAcylation, suggesting that impaired insulin-PI3K-AKT signalling might contribute to neurodegeneration in AD through down-regulation of O-GlcNAcylation and the consequent promotion of abnormal tau hyperphosphorylation and neurodegeneration. The decrease in brain insulin-PI3K-AKT signalling also correlated with the activation of calpain I in the brain, suggesting that the decrease might be caused by calpain over-activation. Our findings provide novel insight into the molecular mechanism by which type 2 diabetes mellitus increases the risk for developing cognitive impairment and dementia in Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Acilação , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/etiologia , Arrestinas/metabolismo , Calpaína/metabolismo , Diabetes Mellitus Tipo 2/complicações , Feminino , Humanos , Masculino , Fosforilação/fisiologia , Transdução de Sinais/fisiologia , beta-Arrestinas , beta-N-Acetil-Hexosaminidases/metabolismo , Proteínas tau/metabolismo
12.
J Biol Chem ; 285(40): 30851-60, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20663882

RESUMO

Abnormal hyperphosphorylation of the microtubule-associated protein Tau is a hallmark of Alzheimer disease and related diseases called tauopathies. As yet, the exact mechanism by which this pathology causes neurodegeneration is not understood. The present study provides direct evidence that Tau abnormal hyperphosphorylation causes its aggregation, breakdown of the microtubule network, and cell death and identifies phosphorylation sites involved in neurotoxicity. We generated pseudophosphorylated Tau proteins by mutating Ser/Thr to Glu and, as controls, to Ala. These mutations involved one, two, or three pathological phosphorylation sites by site-directed mutagenesis using as backbones the wild type or FTDP-17 mutant R406W Tau. Pseudophosphorylated and corresponding control Tau proteins were expressed transiently in PC12 and CHO cells. We found that a single phosphorylation site alone had little influence on the biological activity of Tau, except Thr(212), which, upon mutation to Glu in the R406W background, induced Tau aggregation in cells, suggesting phosphorylation at this site along with a modification on the C-terminal of the protein facilitates self-assembly of Tau. The expression of R406W Tau pseudophosphorylated at Thr(212), Thr(231), and Ser(262) triggered caspase-3 activation in as much as 85% of the transfected cells, whereas the corresponding value for wild type pseudophosphorylated Tau was 30%. Cells transfected with pseudophosphorylated Tau became TUNEL-positive.


Assuntos
Doenças Neurodegenerativas/metabolismo , Proteínas tau/metabolismo , Substituição de Aminoácidos , Animais , Células CHO , Caspase 3/genética , Caspase 3/metabolismo , Cricetinae , Cricetulus , Ativação Enzimática/genética , Humanos , Mutação de Sentido Incorreto , Doenças Neurodegenerativas/genética , Células PC12 , Fosforilação/genética , Estrutura Terciária de Proteína , Ratos , Proteínas tau/genética
13.
Acta Neuropathol ; 122(5): 543-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21959585

RESUMO

Alzheimer disease (AD) is a chronic, progressive disorder with an average disease progression of 7-10 years. However, the histopathological hallmark lesions of this disease, the extracellular Aß plaques and the intraneuronal neurofibrillary tangles, start as early as childhood in the affected individuals. AD is multifactorial and probably involves many different etiopathogenic mechanisms. Thus, while AD offers a wide window of opportunity that practically includes the whole life span of the affected individuals, and numerous therapeutic targets, the multifactorial nature of this disease also makes the selection of the therapeutic targets an immensely challenging task. In addition to ß-amyloidosis and neurofibrillary degeneration, the AD brain also is compromised in its ability to regenerate by enhancing neurogenesis and neuronal plasticity. An increasing number of preclinical studies in transgenic mouse models of AD show that enhancement of neurogenesis and neuronal plasticity can reverse cognitive impairment. Development of both drugs that can inhibit neurodegeneration and drugs that can increase the regenerative capacity of the brain by enhancing neurogenesis and neuronal plasticity are required to control AD.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/terapia , Pesquisa Biomédica/tendências , Doença de Alzheimer/fisiopatologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Degeneração Neural/fisiopatologia , Neurogênese/fisiologia , Plasticidade Neuronal/fisiologia , Placa Amiloide/fisiopatologia
14.
FASEB J ; 24(11): 4420-32, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20651003

RESUMO

Development of rational therapeutic treatments of Alzheimer disease (AD) requires the elucidation of the etiopathogenic mechanisms of neurofibrillary degeneration and ß-amyloidosis, the two hallmarks of this disease. Here we show, employing an adeno-associated virus serotype 1 (AAV1)-induced expression of the C-terminal fragment (I(2CTF)) of I(2)(PP2A), also called SET, in rat brain, decrease in protein phosphatase 2A (PP2A) activity, abnormal hyperphosphorylation of tau, and neurodegeneration; littermates treated identically but with vector only, i.e., AAV1-enhanced green fluorescent protein (GFP), served as a control. Furthermore, there was an increase in the level of activated glycogen synthase kinase-3ß and enhanced expression of intraneuronal Aß in AAV1-I(2CTF) animals. Morris water maze behavioral test revealed that infection with AAV1-I(2CTF) induced spatial reference memory and memory consolidation deficits and a decrease in the brain level of pSer133-CREB. These findings suggest a novel etiopathogenic mechanism of AD, which is initiated by the cleavage of I(2)(PP2A), producing I(2CTF), and describe a novel disease-relevant nontransgenic animal model of AD.


Assuntos
Doença de Alzheimer/patologia , Proteínas de Transporte/metabolismo , Transtornos Cognitivos/patologia , Proteínas Nucleares/metabolismo , Adenoviridae/genética , Animais , Proteínas de Transporte/genética , Linhagem Celular , Dendritos/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Camundongos , Neurônios/patologia , Proteínas Nucleares/genética , Fosforilação , Proteína Fosfatase 2/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/genética , Sinapses/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
15.
Biochem Soc Trans ; 38(4): 962-6, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20658985

RESUMO

Independent of the aetiology, AD (Alzheimer's disease) neurofibrillary degeneration of abnormally hyperphosphorylated tau, a hallmark of AD and related tauopathies, is apparently required for the clinical expression of the disease and hence is a major therapeutic target for drug development. However, AD is multifactorial and heterogeneous and probably involves several different aetiopathogenic mechanisms. On the basis of CSF (cerebrospinal fluid) levels of Abeta(1-42) (where Abeta is amyloid beta-peptide), tau and ubiquitin, five different subgroups, each with its own clinical profile, have been identified. A successful development of rational therapeutic disease-modifying drugs for AD will require understanding of the different aetiopathogenic mechanisms involved and stratification of AD patients by different disease subgroups in clinical trials. We have identified a novel aetiopathogenic mechanism of AD which is initiated by the cleavage of SET, also known as inhibitor-2 (I(2)(PP2A)) of PP2A (protein phosphatase 2A) at Asn(175) into N-terminal (I(2NTF)) and C-terminal (I(2CTF)) halves and their translocation from the neuronal nucleus to the cytoplasm. AAV1 (adeno-associated virus 1)-induced expression of I(2CTF) in rat brain induces inhibition of PP2A activity, abnormal hyperphosphorylation of tau, neurodegeneration and cognitive impairment in rats. Restoration of PP2A activity by inhibition of the cleavage of I(2)(PP2A)/SET offers a promising therapeutic opportunity in AD with this aetiopathogenic mechanism.


Assuntos
Doença de Alzheimer/etiologia , Degeneração Neural/etiologia , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Demência/etiologia , Demência/metabolismo , Demência/patologia , Humanos , Modelos Biológicos , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Emaranhados Neurofibrilares/patologia , Ratos , Tauopatias/etiologia , Tauopatias/metabolismo , Proteínas tau/metabolismo , Proteínas tau/fisiologia
16.
Am J Pathol ; 175(5): 2089-98, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19815707

RESUMO

Recent studies have suggested a possible role of insulin dysfunction in the pathogenesis of sporadic Alzheimer's disease (AD). In AD, brain glucose metabolism is impaired, and this impairment appears to precede the pathology and clinical symptoms of the disease. However, the exact contribution of impaired insulin signaling to AD is not known. In this study, by using a nontransgenic rat model of sporadic AD generated by intracerebroventricular administration of streptozotocin, we investigated insulin signaling, glucose transporters, protein O-GlcNAcylation, and phosphorylation of tau and neurofilaments in the brain. We found impaired insulin signaling, overactivation of glycogen synthase kinase-3beta, decreased levels of major brain glucose transporters, down- regulated protein O-GlcNAcylation, increased phosphorylation of tau and neurofilaments, and decreased microtubule-binding activity of tau in the brains of streptozotocin-treated rats. These results suggest that impaired brain insulin signaling may lead to overactivation of glycogen synthase kinase-3beta and down-regulation of O-GlcNAcylation, which, in turn, facilitate abnormal hyperphosphorylation of tau and neurofilaments and, consequently, neurofibrillary degeneration.


Assuntos
Doença de Alzheimer , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Insulina/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Proteínas de Neurofilamentos/metabolismo , Transdução de Sinais/fisiologia , Proteínas tau/metabolismo , Acetilglucosamina/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Masculino , Microtúbulos/metabolismo , N-Acetilglucosaminiltransferases/genética , Proteínas de Neurofilamentos/genética , Fosforilação , Ratos , Ratos Wistar , Estreptozocina/metabolismo , Proteínas tau/genética
17.
Acta Neuropathol ; 120(5): 605-21, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20697724

RESUMO

In addition to the occurrence of numerous neurofibrillary tangles and Aß plaques, neurogenesis and neuronal plasticity are markedly altered in Alzheimer disease (AD). Although the most popular therapeutic approach has been to inhibit neurodegeneration, another is to promote neurogenesis and neuronal plasticity by utilizing the regenerative capacity of the brain. Here we show that, in a transgenic mouse model of AD, 3xTg-AD mice, there was a marked deficit in neurogenesis and neuroplasticity, which occurred before the formation of any neurofibrillary tangles or Aß plaques and was associated with cognitive impairment. Furthermore, peripheral administration of Peptide 6, an 11-mer, which makes an active region of ciliary neurotrophic factor (CNTF, amino acid residues 146-156), restored cognition by enhancing neurogenesis and neuronal plasticity in these mice. Although this treatment had no detectable effect on Aß and tau pathologies in 9-month animals, it enhanced neurogenesis in dentate gyrus, reduced ectopic birth in the granular cell layer, and increased neuronal plasticity in the hippocampus and cerebral cortex. These findings, for the first time, demonstrate the possibility of therapeutic treatment of AD and related disorders by peripheral administration of a peptide corresponding to a biologically active region of CNTF.


Assuntos
Transtornos Cognitivos/prevenção & controle , Emaranhados Neurofibrilares/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Placa Amiloide/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/metabolismo , Animais , Western Blotting , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Cromatografia Líquida de Alta Pressão , Fator Neurotrófico Ciliar/química , Transtornos Cognitivos/metabolismo , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Imuno-Histoquímica , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Peptídeos/síntese química , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Estrutura Secundária de Proteína , Proteínas tau/metabolismo
18.
Brain ; 132(Pt 7): 1820-32, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19451179

RESUMO

It has been established for a long time that brain glucose metabolism is impaired in Alzheimer's disease. Recent studies have demonstrated that impaired brain glucose metabolism precedes the appearance of clinical symptoms, implying its active role in the development of Alzheimer's disease. However, the molecular mechanism by which this impairment contributes to the disease is not known. In this study, we demonstrated that protein O-GlcNAcylation, a common post-translational modification of nucleocytoplasmic proteins with beta-N-acetyl-glucosamine and a process regulated by glucose metabolism, was markedly decreased in Alzheimer's disease cerebrum. More importantly, the decrease in O-GlcNAc correlated negatively with phosphorylation at most phosphorylation sites of tau protein, which is known to play a crucial role in the neurofibrillary degeneration of Alzheimer's disease. We also found that hyperphosphorylated tau contained 4-fold less O-GlcNAc than non-hyperphosphorylated tau, demonstrating for the first time an inverse relationship between O-GlcNAcylation and phosphorylation of tau in the human brain. Downregulation of O-GlcNAcylation by knockdown of O-GlcNAc transferase with small hairpin RNA led to increased phosphorylation of tau in HEK-293 cells. Inhibition of the hexosamine biosynthesis pathway in rat brain resulted in decreased O-GlcNAcylation and increased phosphorylation of tau, which resembled changes of O-GlcNAcylation and phosphorylation of tau in rodent brains with decreased glucose metabolism induced by fasting, but not those in rat brains when protein phosphatase 2A was inhibited. Comparison of tau phosphorylation patterns under various conditions suggests that abnormal tau hyperphosphorylation in Alzheimer's disease brain may result from downregulation of both O-GlcNAcylation and protein phosphatase 2A. These findings suggest that impaired brain glucose metabolism leads to abnormal hyperphosphorylation of tau and neurofibrillary degeneration via downregulation of tau O-GlcNAcylation in Alzheimer's disease. Thus, restoration of brain tau O-GlcNAcylation and protein phosphatase 2A activity may offer promising therapeutic targets for treating Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Glucose/metabolismo , N-Acetilglucosaminiltransferases/fisiologia , Proteínas tau/metabolismo , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Células Cultivadas , Regulação para Baixo , Feminino , Hexosaminas/biossíntese , Humanos , Masculino , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Fosforilação , Proteína Fosfatase 2/metabolismo
19.
Alzheimers Dement ; 6(5): 420-4, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20813343

RESUMO

Alzheimer's disease (AD) is multifactorial and apparently involves several different etiopathogenic mechanisms. There are at least five subgroups of AD based on cerebrospinal fluid levels of Abeta(1-42), a marker of beta-amyloid (Abeta) plaques, and tau and ubiquitin, two markers of neurofibrillary tangles. These different AD subgroups may respond differently to a given disease-modifying drug, and hence, different therapeutic drugs for different disease subgroups might be required. Stratification of AD patients by disease subgroups in clinical trials is critical to the successful development of potent disease-modifying drugs. Levels of disease markers in the cerebrospinal fluid are promising, both in identifying various subgroups of AD and in monitoring the response to therapeutic drugs.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Antipsicóticos/uso terapêutico , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Antipsicóticos/classificação , Humanos , Fragmentos de Peptídeos/líquido cefalorraquidiano , Placa Amiloide/patologia , Ubiquitina/líquido cefalorraquidiano , Proteínas tau/líquido cefalorraquidiano
20.
J Neurochem ; 111(1): 242-9, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19659459

RESUMO

Type 2 diabetes mellitus (T2DM) increases the risk for Alzheimer's disease (AD), but the underlying mechanism is unknown. In this study, we determined the levels of major brain glucose transporters, O-GlcNAcylation and phosphorylation of tau in the postmortem brain tissue from frontal cortices of 7 controls, 11 T2DM subjects, 10 AD subjects and 8 additional subjects who had both T2DM and AD. We found that the neuronal glucose transporter 3 was decreased to a bigger extent in T2DM brain than in AD brain. The O-GlcNAcylation levels of global proteins and of tau were also decreased in T2DM brain as seen in AD brain. Phosphorylation of tau at some of the AD abnormal hyperphosphorylation sites was increased in T2DM brain. These results suggest that T2DM may contribute to the increased risk for AD by impairing brain glucose uptake/metabolism and, consequently, down-regulation of O-GlcNAcylation, which facilitates abnormal hyperphosphorylation of tau.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/patologia , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , N-Acetilglucosaminiltransferases/fisiologia , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Regulação para Baixo/fisiologia , Feminino , Proteínas Facilitadoras de Transporte de Glucose/classificação , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA