Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Clin Immunol ; 34 Suppl 1: S64-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24711006

RESUMO

The top-down, reductionist approach of the past three decades has resulted in remarkable progress in identifying genes and proteins involved in Alzheimer's disease (AD), including ß-amyloid (Aß) peptides and tau protein. Recently, a number of genes of the innate immune pathway have been identified as AD risk factors and several microglial proteins have been shown to be chronically activated in AD brains. Together, these observations suggest a crucial role for neuroinflammation in AD pathogenesis and emerging evidence suggests that neuroinflammation is both a cause and a consequence of AD. Epidemiological studies show that long-term users of anti-inflammatory drugs are protected from AD but anti-inflammatory treatment in mild AD patients has not been successful. These observations suggest that anti-inflammatory treatment is likely to be successful if initiated prior to the onset of neurological symptoms. Finally, after the remarkable success of the reductionist approach, a complimentary bottom-up systems approach is necessary to gain a better understanding of the highly complex, multifactorial nature of AD pathogenesis.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Imunoterapia/métodos , Inflamação Neurogênica , Proteínas tau/imunologia , Doença de Alzheimer/terapia , Animais , Anti-Inflamatórios/uso terapêutico , Humanos , Imunidade Inata , Terapia de Alvo Molecular
2.
Neurodegener Dis ; 12(1): 51-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22965147

RESUMO

BACKGROUND: Amyloid-ß (Aß) peptides derive from the amyloid precursor protein (APP) and play a pivotal role in Alzheimer's disease (AD) pathogenesis. Our previous work showed that the APP intracellular domain (AICD), which is produced simultaneously with Aß, also contributes to the development of AD-like features. Studies show that administration of apolipoprotein E (apoE) and apoE-derived small peptide mimetics protect AD mouse models against these AD-like features. However, the effects of apoE-mimetic treatment on AICD-mediated AD-like pathologies remain to be elucidated. OBJECTIVE: To study the effects of an apoE mimetic (COG112) on neuroinflammation, hyperphosphorylation of tau and defects in adult neurogenesis in AICD- overexpressing transgenic mice (FeCγ25 line). METHODS: Beginning at 1 month of age, animals were administered subcutaneous COG112 3 times per week for 3 months, followed by immunohistochemical analysis for neuroinflammation, neurogenesis and phosphorylated tau. RESULTS: Treatment with COG112 significantly reduced neuroinflammation in AICD mice and protected against impaired adult hippocampal neurogenesis. We also found that COG112 treatment reduced hyperphosphorylation and somatodendritic accumulation of tau in the hippocampus and cerebral cortex of AICD mice. CONCLUSIONS: Reduction of neuroinflammation by the apoE-mimetic COG112 protects against impaired neurogenesis and tau pathology in AICD transgenic mice. These data suggest that neuroinflammation plays an important role in AICD-induced AD-like pathologies.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide/genética , Gliose/tratamento farmacológico , Neurogênese/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Apolipoproteínas E/metabolismo , Feminino , Gliose/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação/tratamento farmacológico , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Peptídeos/uso terapêutico , Fosforilação/efeitos dos fármacos
4.
Proc Natl Acad Sci U S A ; 106(43): 18367-72, 2009 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-19837693

RESUMO

The hypothesis that amyloid-beta (Abeta) peptides are the primary cause of Alzheimer's disease (AD) remains the best supported theory of AD pathogenesis. Yet, many observations are inconsistent with the hypothesis. Abeta peptides are generated when amyloid precursor protein (APP) is cleaved by presenilins, a process that also produces APP intracellular domain (AICD). We previously generated AICD-overexpressing transgenic mice that showed abnormal activation of GSK-3beta, a pathological feature of AD. We now report that these mice exhibit additional AD-like characteristics, including hyperphosphorylation and aggregation of tau, neurodegeneration and working memory deficits that are prevented by treatment with lithium, a GSK-3beta inhibitor. Consistent with its potential role in AD pathogenesis, we find AICD levels to be elevated in brains from AD patients. The in vivo findings that AICD can contribute to AD pathology independently of Abeta have important therapeutic implications and may explain some observations that are discordant with the amyloid hypothesis.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Envelhecimento , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/patologia , Humanos , Espaço Intracelular/metabolismo , Compostos de Lítio/uso terapêutico , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Fosforilação , Estrutura Terciária de Proteína , Proteínas tau/metabolismo
5.
J Neurosci ; 30(45): 14946-54, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21068297

RESUMO

Despite the progress of the past two decades, the cause of Alzheimer's disease (AD) and effective treatments against it remain elusive. The hypothesis that amyloid-ß (Aß) peptides are the primary causative agents of AD retains significant support among researchers. Nonetheless, a growing body of evidence shows that Aß peptides are unlikely to be the sole factor in AD etiology. Evidence that Aß/amyloid-independent factors, including the actions of AD-related genes, also contribute significantly to AD pathogenesis was presented in a symposium at the 2010 Annual Meeting of the Society for Neuroscience. Here we summarize the studies showing how amyloid-independent mechanisms cause defective endo-lysosomal trafficking, altered intracellular signaling cascades, or impaired neurotransmitter release and contribute to synaptic dysfunction and/or neurodegeneration, leading to dementia in AD. A view of AD pathogenesis that encompasses both the amyloid-dependent and -independent mechanisms will help fill the gaps in our knowledge and reconcile the findings that cannot be explained solely by the amyloid hypothesis.


Assuntos
Doença de Alzheimer/etiologia , Lisossomos/patologia , Neurônios/patologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides , Humanos , Transporte Proteico , Transdução de Sinais
6.
Dev Biol ; 335(1): 1-11, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19664615

RESUMO

Amyloid precursor protein (APP) has been a focus of intense investigation because of its role in Alzheimer's disease (AD), however, its biological function remains uncertain. Loss of APP and APP-like proteins results in postnatal lethality in mice, suggesting a role during embryogenesis. Here we show that in a zebrafish model system, knock down of APP results in the generation of fish with dramatically reduced body length and a short, curly tail. In situ examination of gene expression suggests that the APP morphant embryos have defective convergent-extension movements. We also show that wild-type human APP rescues the morphant phenotype, but the Swedish mutant APP, which causes familial AD (fAD), does not rescue the developmental defects. Collectively, this work demonstrates that the zebrafish model is a powerful system to define the role of APP during embryonic development and to evaluate the functional activity of fAD mutant APP.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/classificação , Precursor de Proteína beta-Amiloide/genética , Animais , Técnicas de Silenciamento de Genes , Humanos , Hibridização In Situ , Camundongos , Mutação , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Fenótipo , Filogenia , Peixe-Zebra/anatomia & histologia , Peixe-Zebra/fisiologia , Proteínas de Peixe-Zebra/genética
8.
J Cell Biol ; 171(2): 327-35, 2005 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-16230462

RESUMO

Amyloid precursor protein (APP), implicated in Alzheimer's disease, is a trans-membrane protein of undetermined function. APP is cleaved by gamma-secretase that releases the APP intracellular domain (AICD) in the cytoplasm. In vitro studies have implicated AICD in cell signaling and transcriptional regulation, but its biologic relevance has been uncertain and its in vivo function has not been examined. To investigate its functional role, we generated AICD transgenic mice, and found that AICD causes significant biologic changes in vivo. AICD transgenic mice show activation of glycogen synthase kinase-3beta (GSK-3beta) and phosphorylation of CRMP2 protein, a GSK-3beta substrate that plays a crucial role in Semaphorin3a-mediated axonal guidance. Our data suggest that AICD is biologically relevant, causes significant alterations in cell signaling, and may play a role in axonal elongation or pathfinding.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Ativação Enzimática , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Transgênicos , Fosforilação , Estrutura Terciária de Proteína/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
J Neurosci ; 28(43): 10786-93, 2008 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-18945886

RESUMO

Neurons subject to degeneration in Alzheimer's disease (AD) exhibit evidence of re-entry into a mitotic cell cycle even before the development of substantial AD brain pathology. In efforts to identify the initiating factors underlying these cell cycle events (CCEs), we have characterized the appearance of the neuronal CCEs in the genomic-based R1.40 transgenic mouse model of AD. Notably, R1.40 mice exhibit neuronal CCEs in a reproducible temporal and spatial pattern that recapitulates the neuronal vulnerability seen in human AD. Neuronal CCEs first appear at 6 months in the frontal cortex layers II/III. This is 6-8 months before detectable amyloid beta (Abeta) deposition, suggesting that specific amyloid precursor protein (APP) processing products are responsible for the induction of neuronal CCEs. Furthermore, a reduction in the levels of Abeta (achieved by shifting the genetic background from C57BL/6 to the DBA/2 mouse strain) dramatically delays the appearance of neuronal CCEs. More significantly, elimination of beta-secretase activity blocks the appearance of CCEs, providing direct genetic evidence that the amyloidogenic processing of APP is required for the induction of CCEs. Finally, in vitro preparations of oligomeric, but not monomeric, Abeta induce DNA synthesis in dissociated cortical neurons, and this response is blocked by antioligomer specific antibodies. Together, our data suggest that low molecular weight aggregates of Abeta induce neuronal cell cycle re-entry in mouse models of Alzheimer's disease.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/farmacologia , Ciclo Celular/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/farmacologia , Fatores Etários , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/deficiência , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/deficiência , Bromodesoxiuridina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Córtex Cerebral/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação/genética
10.
Biol Psychiatry ; 83(5): 416-427, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28587718

RESUMO

BACKGROUND: Mitophagy and mitochondrial dynamics alterations are two major hallmarks of neurodegenerative diseases. Dysfunctional mitochondria accumulate in Alzheimer's disease-affected brains by yet unexplained mechanisms. METHODS: We combined cell biology, molecular biology, and pharmacological approaches to unravel a novel molecular pathway by which presenilins control phosphatase and tensin homolog-induced kinase 1 (Pink-1) expression and transcription. In vivo approaches were carried out on various transgenic and knockout animals as well as in adeno-associated virus-infected mice. Functional readout and mitochondrial physiology (mitochondrial potential) were assessed by combined procedures including flow cytometry, live imaging analysis, and immunohistochemistry. RESULTS: We show that presenilins 1 and 2 trigger opposite effects on promoter transactivation, messenger RNA, and protein expression of Pink-1. This control is linked to γ-secretase activity and ß-amyloid precursor protein but is independent of phosphatase and tensin homolog. We show that amyloid precursor protein intracellular domain (AICD) accounts for presenilin-dependent phenotype and upregulates Pink-1 transactivation in cells as well as in vivo in a Forkhead box O3a-dependent manner. Interestingly, the modulation of γ-secretase activity or AICD expression affects Pink-1-related control of mitophagy and mitochondrial dynamics. Finally, we show that parkin acts upstream of presenilins to control Pink-1 promoter transactivation and protein expression. CONCLUSIONS: Overall, we delineate a molecular cascade presenilins-AICD-Forkhead box O3a linking parkin to Pink-1. Our study demonstrates AICD-mediated Pink-1-dependent control of mitochondrial physiology by presenilins. Furthermore, it unravels a parkin-Pink-1 feedback loop controlling mitochondrial physiology that could be disrupted in neurodegenerative conditions.


Assuntos
Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteína Forkhead Box O3/metabolismo , Hipocampo/metabolismo , Mitocôndrias/metabolismo , Presenilinas/metabolismo , Proteínas Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Embrião de Mamíferos , Fibroblastos , Células HEK293 , Humanos , Espaço Intracelular/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
11.
Mol Endocrinol ; 19(2): 350-61, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15514032

RESUMO

Early reports showed that androgen receptor (AR) NH2- and COOH-terminal (N-C) interaction was important for full AR function. However, the influence of these interactions on the AR in vivo effects remains unclear. Here we tested some AR-associated peptides and coregulators to determine their influences on AR N-C interaction, AR transactivation, and AR coregulator function. The results showed that AR coactivators such as ARA70N, gelsolin, ARA54, and SRC-1 can enhance AR transactivation but showed differential influences on the N-C interaction. In contrast, AR corepressors ARA67 and Rad9 can suppress AR transactivation, with ARA67 enhancing and Rad9 suppressing AR N-C interaction. Furthermore, liganded AR C terminus-associated peptides can block AR N-C interaction, but only selective peptides can block AR transactivation and coregulator function. We found all the tested peptides can suppress prostate cancer LNCaP cell growth at different levels in the presence of 5alpha-dihydrotestosterone, but only the tested FXXLF-containing peptides, not FXXMF-containing peptides, can suppress prostate cancer CWR22R cell growth. Together, these results suggest that the effects of AR N-C interactions may not always correlate with similar effects on AR-mediated transactivation and/or AR-mediated cell growth. Therefore, drugs designed by targeting AR N-C interaction as a therapeutic intervention for prostate cancer treatment may face unpredictable in vivo effects.


Assuntos
Receptores Androgênicos/química , Motivos de Aminoácidos , Animais , Western Blotting , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Di-Hidrotestosterona/química , Gelsolina/metabolismo , Genes Reporter , Histona Acetiltransferases , Humanos , Ligantes , Masculino , Microscopia de Fluorescência , Coativador 1 de Receptor Nuclear , Coativadores de Receptor Nuclear , Proteínas Oncogênicas/metabolismo , Peptídeos/química , Plasmídeos/metabolismo , Neoplasias da Próstata/patologia , Ligação Proteica , Estrutura Terciária de Proteína , Fatores de Tempo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Transfecção , Técnicas do Sistema de Duplo-Híbrido
12.
PLoS One ; 11(7): e0159435, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27459671

RESUMO

Amyloid precursor protein (APP) is cleaved by gamma-secretase to simultaneously generate amyloid beta (Aß) and APP Intracellular Domain (AICD) peptides. Aß plays a pivotal role in Alzheimer's disease (AD) pathogenesis but recent studies suggest that amyloid-independent mechanisms also contribute to the disease. We previously showed that AICD transgenic mice (AICD-Tg) exhibit AD-like features such as tau pathology, aberrant neuronal activity, memory deficits and neurodegeneration in an age-dependent manner. Since AD is a tauopathy and tau has been shown to mediate Aß-induced toxicity, we examined the role of tau in AICD-induced pathological features. We report that ablating endogenous tau protects AICD-Tg mice from deficits in adult neurogenesis, seizure severity, short-term memory deficits and neurodegeneration. Deletion of tau restored abnormal phosphorylation of NMDA receptors, which is likely to underlie hyperexcitability and associated excitotoxicity in AICD-Tg mice. Conversely, overexpression of wild-type human tau aggravated receptor phosphorylation, impaired adult neurogenesis, memory deficits and neurodegeneration. Our findings show that tau is essential for mediating the deleterious effects of AICD. Since tau also mediates Aß-induced toxic effects, our findings suggest that tau is a common downstream factor in both amyloid-dependent and-independent pathogenic mechanisms and therefore could be a more effective drug target for therapeutic intervention in AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas tau/metabolismo , Doença de Alzheimer/psicologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Feminino , Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Ácido Caínico/efeitos adversos , Lítio/farmacologia , Masculino , Aprendizagem em Labirinto , Memória de Curto Prazo , Camundongos , Camundongos Transgênicos , Neurogênese/efeitos dos fármacos , Neurogênese/genética , Neurônios/metabolismo , Neurônios/patologia , Fenótipo , Fosforilação/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/induzido quimicamente , Convulsões/metabolismo , Proteínas tau/genética
13.
Neurobiol Aging ; 36(8): 2370-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26022769

RESUMO

Amyloid precursor protein, which generates amyloid beta peptides, is intimately associated with Alzheimer's disease (AD) pathogenesis. We previously showed that transgenic mice overexpressing amyloid precursor protein intracellular domain (AICD), a peptide generated simultaneously with amyloid beta, develop AD-like pathologies, including hyperphosphorylated tau, loss of synapses, and memory impairments. AICD is known to bind c-Jun N-terminal kinase (JNK)-interacting protein 1 (JIP1), a scaffold protein that associates with and activates JNK. The aim of this study was to examine the role of JIP1 in AICD-induced AD-like pathologies in vivo, since the JNK pathway is aberrantly activated in AD brains and contributes to AD pathologies. We generated AICD-Tg mice lacking the JIP1 gene (AICD; JIP1(-/-)) and found that although AICD; JIP1(-/-) mice exhibit increased AICD, the absence of JIP1 results in decreased levels of hyperphosphorylated tau and activated JNK. AICD; JIP1(-/-) mice are also protected from synaptic loss and show improved performance in behavioral tests. These results suggest that JIP1 mediates AD-like pathologies in AICD-Tg mice and that JNK signaling may contribute to amyloid-independent mechanisms of AD pathogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/patologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Feminino , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ligação Proteica
14.
Invert Neurosci ; 15(3): 4, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26159098

RESUMO

Oligodendrocytes produce multi-lamellar myelin membranes that surround axons in the central nervous system (CNS). Preservation and generation of myelin are potential therapeutic targets for dysmyelinating and demyelinating diseases. MicroRNAs (miRNAs) play a vital role in oligodendrocyte differentiation and overall CNS development. miR-124 is a well-conserved neuronal miRNA with important roles in neuronal differentiation and function. miR-124 levels increase following loss of myelin in both human and rodent brains. While the role of neuronal miR-124 in neurogenesis has been established, its effects on axonal outgrowth and oligodendrocytes are not currently known. We therefore explored the possible effect of selective knockdown of miR-124 in Danio rerio using a morpholino-based knockdown approach. No morphological abnormalities or loss of motor neurons were detected despite loss of axonal outgrowth. Morpholino-based knockdown of miR-124 led to reciprocal increases in mRNA levels of target genes that inhibit axonal and dendritic projections. Importantly, loss of miR-124 led to decreased oligodendrocyte cell numbers and myelination of axonal projections in the ventral hindbrain. Taken together, our results add a new dimension to the existing complexity of neuron-glial relationships and highlight the utility of Danio rerio as a model system to investigate such interactions.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , MicroRNAs/metabolismo , Oligodendroglia/metabolismo , Peixe-Zebra/anatomia & histologia , Animais , Animais Geneticamente Modificados , Relação Dose-Resposta a Droga , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Larva , MicroRNAs/genética , Morfolinos/farmacologia , Oligodendroglia/efeitos dos fármacos
15.
J Exp Med ; 212(3): 287-95, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25732305

RESUMO

Variants in triggering receptor expressed on myeloid cells 2 (TREM2) confer high risk for Alzheimer's disease (AD) and other neurodegenerative diseases. However, the cell types and mechanisms underlying TREM2's involvement in neurodegeneration remain to be established. Here, we report that TREM2 is up-regulated on myeloid cells surrounding amyloid deposits in AD mouse models and human AD tissue. TREM2 was detected on CD45(hi)Ly6C(+) myeloid cells, but not on P2RY12(+) parenchymal microglia. In AD mice deficient for TREM2, the CD45(hi)Ly6C(+) macrophages are virtually eliminated, resulting in reduced inflammation and ameliorated amyloid and tau pathologies. These data suggest a functionally important role for TREM2(+) macrophages in AD pathogenesis and an unexpected, detrimental role of TREM2 in AD pathology. These findings have direct implications for future development of TREM2-targeted therapeutics.


Assuntos
Doença de Alzheimer/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Fatores Etários , Idoso , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Antígenos Comuns de Leucócito/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Camundongos Transgênicos , Receptores Imunológicos/genética , Regulação para Cima , Proteínas tau/metabolismo
16.
PLoS One ; 7(4): e34209, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22545081

RESUMO

Amyloid precursor protein (APP) plays a pivotal role in Alzheimer's disease (AD) pathogenesis, but its normal physiological functions are less clear. Combined deletion of the APP and APP-like protein 2 (APLP2) genes in mice results in post-natal lethality, suggesting that APP performs an essential, if redundant, function during embryogenesis. We previously showed that injection of antisense morpholino to reduce APP levels in zebrafish embryos caused convergent-extension defects. Here we report that a reduction in APP levels causes defective axonal outgrowth of facial branchiomotor and spinal motor neurons, which involves disorganized axonal cytoskeletal elements. The defective outgrowth is caused in a cell-autonomous manner and both extracellular and intracellular domains of human APP are required to rescue the defective phenotype. Interestingly, wild-type human APP rescues the defective phenotype but APPswe mutation, which causes familial AD, does not. Our results show that the zebrafish model provides a powerful system to delineate APP functions in vivo and to study the biological effects of APP mutations.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Axônios/patologia , Técnicas de Silenciamento de Genes , Neurônios Motores/patologia , Peixe-Zebra/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/uso terapêutico , Animais , Axônios/metabolismo , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Neurônios Motores/metabolismo , Mutação , Fenótipo , Peixe-Zebra/embriologia
17.
Methods Mol Biol ; 670: 85-91, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20967585

RESUMO

The cleavage of amyloid precursor protein (APP) by γ-secretase produces Aß peptides, which are prominent features in Alzheimer's disease and have been extensively studied. By contrast, APP intracellular domain (AICD), also a product of this cleavage event, has received little or no investigative attention. A major reason for this is that AICD is generally not detected in tissue lysates and, therefore, is neglected as a non-relevant product of APP metabolism. However, recent studies have shown that AICD regulates a number of important cellular events. Furthermore, we found that contrary to previous assertions, AICD can be detected in brain lysates using Western blotting if an antigen retrieval protocol is employed. Here we describe the protocol for AICD detection and note the biological relevance of AICD in physiological and pathological conditions.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Camundongos , Camundongos Transgênicos , Estrutura Terciária de Proteína
18.
Neurobiol Aging ; 32(12): 2320.e1-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20493588

RESUMO

The cleavage of amyloid precursor protein (APP) by presenilins simultaneously generates amyloid-ß (Aß) and APP intracellular Domain (AICD) peptides. Aß plays a pivotal role in Alzheimer's disease (AD) pathology and recently AICD was also shown to contribute to AD. Transgenic mice overexpressing AICD show age-dependent tau phosphorylation and aggregation, memory deficits, and neurodegeneration. Moreover, these mice show aberrant electrical activity and silent seizures beginning at 3-4 months of age. Here we show that AICD mice also displayed abnormal mossy fiber sprouting beginning about the same time and that this sprouting intensified as the animals aged. Expression of neuropeptide Y was increased in mossy fiber terminals in aged but not young AICD mice. Importantly, young AICD mice injected with kainic acid showed similar pathology to that observed in aged AICD mice. These data show that elevated levels of AICD render neurons hypersensitive to stress and induce hippocampal circuit reorganization, which can further exacerbate hyperexcitability. These results further demonstrate that AICD, in addition to Aß, can play a significant role in AD pathogenesis.


Assuntos
Envelhecimento/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Líquido Intracelular/metabolismo , Rede Nervosa/metabolismo , Estresse Oxidativo , Envelhecimento/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Agonistas de Aminoácidos Excitatórios/toxicidade , Líquido Intracelular/fisiologia , Camundongos , Camundongos Transgênicos , Rede Nervosa/patologia , Estresse Oxidativo/genética , Estrutura Terciária de Proteína/genética
19.
Stem Cell Res Ther ; 2(5): 39, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-22000643

RESUMO

Amyloid precursor protein (APP) fascinates cell biologists because it is proteolytically processed to generate multiple peptides, including amyloid-ß, which is implicated in Alzheimer's disease. However, a large body of data also shows that the extracellular soluble fragment of APP produced by α-secretase (sAPPα) is neuroprotective and promotes neuronal outgrowth. A study by Demars and colleagues appearing in the previous issue provides data showing that sAPPα is a general growth factor for stem cells of multiple lineages. Thus, APP seems to play complex and disparate roles in neurodegeneration and neuroprotection.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Doenças Neurodegenerativas/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Humanos , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
20.
Neurobiol Aging ; 32(6): 1099-113, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19570594

RESUMO

The cleavage of amyloid precursor protein (APP) by caspases unmasks a domain extending from membrane to caspase cleavage site. This domain induces apoptosis in vitro and in vivo when overexpressed in neurons through the help of an internalization vector. In this model, we previously showed that SET rapidly binds to the internalized domain and is involved in downstream deleterious effects. Under these conditions SET mislocalizes from the nucleus to the cytoplasm, as in Alzheimer's disease (AD). In this report using the same model, we show that PAT1 attaches to the internalized domain earlier than SET and that this binding causes an increase in the levels of APP and APLP2 at the cell surface. Down regulation experiments of PAT1 and of APP and APLP2 show that the increase of the levels of APP and APLP2 at the cell surface triggers the cell death signal and SET mislocalization into the cytoplasm. In the context of AD these data suggest that mislocalization of SET into the cytoplasm may occur downstream of first cell death signal events involving PAT1 protein.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/metabolismo , Citoplasma/metabolismo , Chaperonas de Histonas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Simportadores/metabolismo , Fatores de Transcrição/metabolismo , Sistemas de Transporte de Aminoácidos/química , Sistemas de Transporte de Aminoácidos/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Apoptose/fisiologia , Biotinilação/métodos , Caspases/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Proteínas de Ligação a DNA , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Embrião de Mamíferos , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/fisiologia , RNA Interferente Pequeno/farmacologia , Simportadores/química , Simportadores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA