Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
EMBO J ; 41(18): e10242, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35993331

RESUMO

Microtubule-associated protein tau is a central factor in Alzheimer's disease and other tauopathies. However, the physiological functions of tau are unclear. Here, we used proximity-labelling proteomics to chart tau interactomes in primary neurons and mouse brains in vivo. Tau interactors map onto pathways of cytoskeletal, synaptic vesicle and postsynaptic receptor regulation and show significant enrichment for Parkinson's, Alzheimer's and prion disease. We find that tau interacts with and dose-dependently reduces the activity of N-ethylmaleimide sensitive fusion protein (NSF), a vesicular ATPase essential for AMPA-type glutamate receptor (AMPAR) trafficking. Tau-deficient (tau-/- ) neurons showed mislocalised expression of NSF and enhanced synaptic AMPAR surface levels, reversible through the expression of human tau or inhibition of NSF. Consequently, enhanced AMPAR-mediated associative and object recognition memory in tau-/- mice is suppressed by both hippocampal tau and infusion with an NSF-inhibiting peptide. Pathologic mutant tau from mouse models or Alzheimer's disease significantly enhances NSF inhibition. Our results map neuronal tau interactomes and delineate a functional link of tau with NSF in plasticity-associated AMPAR-trafficking and memory.


Assuntos
Doença de Alzheimer , Receptores de AMPA , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Hipocampo/metabolismo , Humanos , Memória , Camundongos , Proteínas Sensíveis a N-Etilmaleimida/genética , Proteínas Sensíveis a N-Etilmaleimida/metabolismo , Neurônios/metabolismo , Transporte Proteico , Receptores de AMPA/genética , Receptores de AMPA/metabolismo
2.
Neurobiol Dis ; 171: 105783, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35675895

RESUMO

Increasing evidence suggests that kynurenine pathway (KP) dyshomeostasis may promote disease progression in dementia. Studies in Alzheimer's disease (AD) patients confirm KP dyshomeostasis in plasma and cerebrospinal fluid (CSF) which correlates with amyloid-ß and tau pathology. Herein, we performed the first comprehensive study assessing baseline levels of KP metabolites in participants enrolling in the Australian Imaging Biomarkers Flagship Study of Aging. Our purpose was to test the hypothesis that changes in KP metabolites may be biomarkers of dementia processes that are largely silent. We used a cross-sectional analytical approach to assess non-progressors (N = 73); cognitively normal (CN) or mild cognitive impairment (MCI) participants at baseline and throughout the study, and progressors (N = 166); CN or MCI at baseline but progressing to either MCI or AD during the study. Significant KP changes in progressors included increased 3-hydroxyanthranilic acid (3-HAA) and 3-hydroxyanthranilic acid/anthranilic acid (3-HAA/AA) ratio, the latter having the largest effect on the odds of an individual being a progressor (OR 35.3; 95% CI between 14 and 104). 3-HAA levels were hence surprisingly bi-phasic, high in progressors but low in non-progressors or participants who had already transitioned to MCI or dementia. This is a new, unexpected and interesting result, as most studies of the KP in neurodegenerative disease show reduced 3-HAA/AA ratio after diagnosis. The neuroprotective metabolite picolinic acid was also significantly decreased while the neurotoxic metabolite 3-hydroxykynurenine increased in progressors. These results were significant even after adjustment for confounders. Considering the magnitude of the OR to predict change in cognition, it is important that these findings are replicated in other populations. Independent validation of our findings may confirm the utility of 3-HAA/AA ratio to predict change in cognition leading to dementia in clinical settings.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Doenças Neurodegenerativas , Ácido 3-Hidroxiantranílico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Austrália , Biomarcadores , Disfunção Cognitiva/líquido cefalorraquidiano , Estudos Transversais , Progressão da Doença , Humanos , Cinurenina , Fragmentos de Peptídeos/líquido cefalorraquidiano , Proteínas tau/líquido cefalorraquidiano
3.
Biochem Cell Biol ; 99(5): 606-616, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33794133

RESUMO

The microtubule-associated protein tau is a key factor in neurodegenerative proteinopathies and is predominantly found in neuronal axons. However, somatodendritic localization of tau occurs in a subset of pathological and physiological tau. Dendritic tau can localize to post-synapses where it interacts with proteins of the post-synaptic density (PSD) protein PSD-95, a membrane-associated guanylate kinase (MAGUK) scaffold factor for organization of protein complexes within the PSD, to mediate downstream signals. However, the molecular details of this interaction remain unclear. Here, we used interaction mapping in cultured cells to demonstrate that tau interacts with the guanylate kinase (GUK) domain in the C-terminal region of PSD-95. The PSD-95 GUK domain is required for a complex with full-length human tau. Mapping the interaction of the MAGUK core with tau revealed that the microtubule binding repeats 2 and 3 and the proline-rich region contributes to this interaction, while the N- and C-terminal regions of tau inhibit interaction. These results reveal the intramolecular determinants of the protein complex of tau and PSD-95 and increase our understanding of tau interactions regulating neurotoxic signaling at the molecular level.


Assuntos
Proteína 4 Homóloga a Disks-Large/metabolismo , Guanilato Quinases/metabolismo , Microtúbulos/metabolismo , Prolina/metabolismo , Proteínas tau/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Ligação Proteica
4.
Brain ; 143(6): 1889-1904, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32375177

RESUMO

Hyperphosphorylation and deposition of tau in the brain characterizes frontotemporal dementia and Alzheimer's disease. Disease-associated mutations in the tau-encoding MAPT gene have enabled the generation of transgenic mouse models that recapitulate aspects of human neurodegenerative diseases, including tau hyperphosphorylation and neurofibrillary tangle formation. Here, we characterized the effects of transgenic P301S mutant human tau expression on neuronal network function in the murine hippocampus. Onset of progressive spatial learning deficits in P301S tau transgenic TAU58/2 mice were paralleled by long-term potentiation deficits and neuronal network aberrations during electrophysiological and EEG recordings. Gene-expression profiling just prior to onset of apparent deficits in TAU58/2 mice revealed a signature of immediate early genes that is consistent with neuronal network hypersynchronicity. We found that the increased immediate early gene activity was confined to neurons harbouring tau pathology, providing a cellular link between aberrant tau and network dysfunction. Taken together, our data suggest that tau pathology drives neuronal network dysfunction through hyperexcitation of individual, pathology-harbouring neurons, thereby contributing to memory deficits.


Assuntos
Tauopatias/genética , Proteínas tau/genética , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Demência Frontotemporal/genética , Hipocampo/metabolismo , Potenciação de Longa Duração/genética , Masculino , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Fosforilação , Tauopatias/fisiopatologia
5.
J Neurosci ; 39(48): 9645-9659, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31641049

RESUMO

Sphingosine 1-phosphate (S1P) is a potent vasculoprotective and neuroprotective signaling lipid, synthesized primarily by sphingosine kinase 2 (SK2) in the brain. We have reported pronounced loss of S1P and SK2 activity early in Alzheimer's disease (AD) pathogenesis, and an inverse correlation between hippocampal S1P levels and age in females, leading us to speculate that loss of S1P is a sensitizing influence for AD. Paradoxically, SK2 was reported to mediate amyloid ß (Aß) formation from amyloid precursor protein (APP) in vitro To determine whether loss of S1P sensitizes to Aß-mediated neurodegeneration, we investigated whether SK2 deficiency worsens pathology and memory in male J20 (PDGFB-APPSwInd) mice. SK2 deficiency greatly reduced Aß content in J20 mice, associated with significant improvements in epileptiform activity and cross-frequency coupling measured by hippocampal electroencephalography. However, several key measures of APPSwInd-dependent neurodegeneration were enhanced on the SK2-null background, despite reduced Aß burden. These included hippocampal volume loss, oligodendrocyte attrition and myelin loss, and impaired performance in Y-maze and social novelty memory tests. Inhibition of the endosomal cholesterol exporter NPC1 greatly reduced sphingosine phosphorylation in glial cells, linking loss of SK2 activity and S1P in AD to perturbed endosomal lipid metabolism. Our findings establish SK2 as an important endogenous regulator of both APP processing to Aß, and oligodendrocyte survival, in vivo These results urge greater consideration of the roles played by oligodendrocyte dysfunction and altered membrane lipid metabolic flux as drivers of neurodegeneration in AD.SIGNIFICANCE STATEMENT Genetic, neuropathological, and functional studies implicate both Aß and altered lipid metabolism and/or signaling as key pathogenic drivers of Alzheimer's disease. In this study, we first demonstrate that the enzyme SK2, which generates the signaling lipid S1P, is required for Aß formation from APP in vivo Second, we establish a new role for SK2 in the protection of oligodendrocytes and myelin. Loss of SK2 sensitizes to Aß-mediated neurodegeneration by attenuating oligodendrocyte survival and promoting hippocampal atrophy, despite reduced Aß burden. Our findings support a model in which Aß-independent sensitizing influences such as loss of neuroprotective S1P are more important drivers of neurodegeneration than gross Aß concentration or plaque density.


Assuntos
Doença de Alzheimer/metabolismo , Doenças Desmielinizantes/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/patologia , Animais , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/prevenção & controle , Feminino , Hipocampo/patologia , Masculino , Camundongos , Camundongos Transgênicos , Neuroproteção/fisiologia , Técnicas de Cultura de Órgãos , Tamanho do Órgão/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Placa Amiloide/patologia
6.
J Biol Chem ; 294(38): 14149-14162, 2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31366728

RESUMO

The microtubule-associated protein tau undergoes aberrant modification resulting in insoluble brain deposits in various neurodegenerative diseases, including frontotemporal dementia (FTD), progressive supranuclear palsy, and corticobasal degeneration. Tau aggregates can form in different cell types of the central nervous system (CNS) but are most prevalent in neurons. We have previously recapitulated aspects of human FTD in mouse models by overexpressing mutant human tau in CNS neurons, including a P301S tau variant in TAU58/2 mice, characterized by early-onset and progressive behavioral deficits and FTD-like neuropathology. The molecular mechanisms underlying the functional deficits of TAU58/2 mice remain mostly elusive. Here, we employed functional genomics (i.e. RNAseq) to determine differentially expressed genes in young and aged TAU58/2 mice to identify alterations in cellular processes that may contribute to neuropathy. We identified genes in cortical brain samples differentially regulated between young and old TAU58/2 mice relative to nontransgenic littermates and by comparative analysis with a dataset of CNS cell type-specific genes expressed in nontransgenic mice. Most differentially-regulated genes had known or putative roles in neurons and included presynaptic and excitatory genes. Specifically, we observed changes in presynaptic factors, glutamatergic signaling, and protein scaffolding. Moreover, in the aged mice, expression levels of several genes whose expression was annotated to occur in other brain cell types were altered. Immunoblotting and immunostaining of brain samples from the TAU58/2 mice confirmed altered expression and localization of identified and network-linked proteins. Our results have revealed genes dysregulated by progressive tau accumulation in an FTD mouse model.


Assuntos
Tauopatias/genética , Tauopatias/metabolismo , Proteínas tau/genética , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Demência Frontotemporal/genética , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Análise de Sequência de RNA/métodos , Tauopatias/fisiopatologia , Proteínas tau/metabolismo
7.
Acta Neuropathol ; 140(3): 279-294, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32725265

RESUMO

Hyperphosphorylation of the neuronal tau protein contributes to Alzheimer's disease (AD) by promoting tau pathology and neuronal and cognitive deficits. In contrast, we have previously shown that site-specific tau phosphorylation can inhibit toxic signals induced by amyloid-ß (Aß) in mouse models. The post-synaptic mitogen-activated protein (MAP) kinase p38γ mediates this site-specific phosphorylation on tau at Threonine-205 (T205). Using a gene therapeutic approach, we draw on this neuroprotective mechanism to improve memory in two Aß-dependent mouse models of AD at stages when advanced memory deficits are present. Increasing activity of post-synaptic kinase p38γ that targets T205 in tau reduced memory deficits in symptomatic Aß-induced AD models. Reconstitution experiments with wildtype human tau or phosphorylation-deficient tauT205A showed that T205 modification is critical for downstream effects of p38γ that prevent memory impairment in APP-transgenic mice. Furthermore, genome editing of the T205 codon in the murine Mapt gene showed that this single side chain in endogenous tau critically modulates memory deficits in APP-transgenic Alzheimer's mice. Ablating the protective effect of p38γ activity by genetic p38γ deletion in a tau transgenic mouse model that expresses non-pathogenic tau rendered tau toxic and resulted in impaired memory function in the absence of human Aß. Thus, we propose that modulating neuronal p38γ activity serves as an intrinsic tau-dependent therapeutic approach to augment compromised cognition in advanced dementia.


Assuntos
Doença de Alzheimer/metabolismo , Transtornos Cognitivos/metabolismo , Transtornos da Memória/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Transtornos Cognitivos/genética , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Memória/fisiologia , Transtornos da Memória/genética , Camundongos , Camundongos Transgênicos
8.
J Biol Chem ; 293(10): 3710-3719, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29382714

RESUMO

Compared with other mammalian species, humans are particularly susceptible to tau-mediated neurodegenerative disorders. Differential interactions of the tau protein with other proteins are critical for mediating tau's physiological functions as well as tau-associated pathological processes. Primate tau harbors an 11-amino acid-long motif in its N-terminal region (residues 18-28), which is not present in non-primate species and whose function is unknown. Here, we used deletion mutagenesis to remove this sequence region from the longest human tau isoform, followed by glutathione S-transferase (GST) pulldown assays paired with isobaric tags for relative and absolute quantitation (iTRAQ) multiplex labeling, a quantitative method to measure protein abundance by mass spectrometry. Using this method, we found that the primate-specific N-terminal tau motif differentially mediates interactions with neuronal proteins. Among these binding partners are proteins involved in synaptic transmission (synapsin-1 and synaptotagmin-1) and signaling proteins of the 14-3-3 family. Furthermore, we identified an interaction of tau with a member of the annexin family (annexin A5) that was linked to the 11-residue motif. These results suggest that primate Tau has evolved specific residues that differentially regulate protein-protein interactions compared with tau proteins from other non-primate mammalian species. Our findings provide in vitro insights into tau's interactions with other proteins that may be relevant to human disease.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Proteínas tau/metabolismo , Sequência de Aminoácidos , Animais , Córtex Cerebral/metabolismo , Biologia Computacional , Sequência Conservada , Deleção de Genes , Ontologia Genética , Células HEK293 , Humanos , Imunoprecipitação , Camundongos Knockout , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/química , Neurônios/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Primatas , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Proteínas tau/química , Proteínas tau/genética
9.
J Neurol Neurosurg Psychiatry ; 87(11): 1234-1241, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27172939

RESUMO

Neurodegeneration refers to a heterogeneous group of brain disorders that progressively evolve. It has been increasingly appreciated that many neurodegenerative conditions overlap at multiple levels and therefore traditional clinicopathological correlation approaches to better classify a disease have met with limited success. Neuronal network disintegration is fundamental to neurodegeneration, and concepts based around such a concept may better explain the overlap between their clinical and pathological phenotypes. In this Review, promoters of overlap in neurodegeneration incorporating behavioural, cognitive, metabolic, motor, and extrapyramidal presentations will be critically appraised. In addition, evidence that may support the existence of large-scale networks that might be contributing to phenotypic differentiation will be considered across a neurodegenerative spectrum. Disintegration of neuronal networks through different pathological processes, such as prion-like spread, may provide a better paradigm of disease and thereby facilitate the identification of novel therapies for neurodegeneration.


Assuntos
Rede Nervosa/patologia , Doenças Neurodegenerativas/patologia , Encéfalo/patologia , Humanos , Doenças Neurodegenerativas/diagnóstico , Fenótipo
10.
J Neurochem ; 132(1): 135-45, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25041093

RESUMO

Immunization is increasingly recognized as a suitable therapeutic avenue for the treatment of neurological diseases such as Alzheimer's disease and other tauopathies. Tau is a key molecular player in these conditions and therefore represents an attractive target for passive immunization approaches. We performed such an approach in two independent tau transgenic mouse models of tauopathy, K369I tau transgenic K3 and P301L tau transgenic pR5 mice. The antibodies we used were either specific for full-length tau or tau phosphorylated at serine 404 (pS404), a residue that forms part of the paired helical filament (PHF)-1 phosphoepitope that characterizes tau neurofibrillary tangles in tauopathies. Although both pS404 antibodies had a similar affinity, they differed in isotype, and only passive immunization with the IgG2a/κ pS404-specific antibody resulted in a lower tangle burden and reduced phosphorylation of tau at the PHF1 epitope in K3 mice. In pR5 mice, the same antibody led to a reduced phosphorylation of the pS422 and PHF1 epitopes of tau. In addition, histological sections of the hippocampal dentate gyrus of the immunized pR5 mice displayed reduced pS422 staining intensities. These results show that passive immunization targeting tau can modulate aspects of tau pathology in tau transgenic mouse models, in an antibody isotype-specific manner. We show that passive immunization targeting the pathological phosphorylation site pS404 on human tau with a monoclonal IgG2a/κ, but not a IgG1/κ antibody, reduced hyperphosphorylation of tau and tangle burden in two independent mouse models of tau pathology. This shows that both specificity and isotype of phospho-tau (p-tau)-specific antibodies are important for therapeutically ameliorating tau pathology.


Assuntos
Imunização Passiva , Imunoterapia/métodos , Tauopatias/terapia , Proteínas tau/imunologia , Animais , Anticorpos/análise , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/patologia , Córtex Cerebral/patologia , Proteínas de Ligação a DNA , Humanos , Corpos de Inclusão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação/genética , Proteínas do Grupo Polycomb , Tauopatias/imunologia , Fatores de Transcrição/genética
11.
Diabetologia ; 57(7): 1410-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24733160

RESUMO

AIMS/HYPOTHESIS: Regulation of insulin secretion along the secretory pathway is incompletely understood. We addressed the expression of SIL1, a nucleotide exchange factor for the endoplasmic reticulum (ER) chaperone glucose-regulated protein 78 kD (GRP78), in pancreatic beta cells and investigated whether or not SIL1 is involved in beta cell function. METHODS: SIL1 expression was analysed by immunoblotting and immunofluorescence. Metabolic and islet variables, including glucose tolerance, beta cell mass, insulin secretion, islet ultrastructure, insulin content and levels of ER stress marker proteins, were addressed in Sil1 knockout (Sil1 (-/-)) mice. Insulin, proinsulin and C-peptide release was addressed in Sil1 (-/-) islets, and SIL1 overexpression or knockdown was explored in MIN6 cells in vitro. Models of type 1 diabetes and insulin resistance were induced in Sil1 (-/-) mice by administration of streptozotocin (STZ) and a high-fat diet (HFD), respectively. RESULTS: We show that SIL1 is expressed in pancreatic beta cells and is required for islet insulin content, islet sizing, glucose tolerance and glucose-stimulated insulin secretion in vivo. Levels of pancreatic ER stress markers are increased in Sil1 (-/-) mice, and Sil1 (-/-) beta cell ER is ultrastructurally compromised. Isolated Sil1 (-/-) islets show lower proinsulin and insulin content and impaired glucose-stimulated insulin secretion. Modulation of SIL1 protein levels in MIN6 cells correlates with changes in insulin content and secreted insulin. Furthermore, Sil1 (-/-) mice are more susceptible to STZ-induced type 1 diabetes with increased apoptosis. Upon HFD feeding, Sil1 (-/-) mice show markedly lower insulin secretion and exacerbated glucose intolerance compared with control mice. Surprisingly, however, HFD-fed Sil1 (-/-) mice display pronounced islet hyperplasia with low amounts of insulin in total pancreas. CONCLUSIONS/INTERPRETATION: These results reveal a novel role for the nucleotide exchange factor SIL1 in pancreatic beta cell function under physiological and disease conditions such as diabetes and the metabolic syndrome.


Assuntos
Retículo Endoplasmático/metabolismo , Glucose/farmacologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animais , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Knockout
12.
EMBO Rep ; 13(11): 1012-20, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-22964757

RESUMO

Adipose tissue is the largest compartment in the mammalian body for storing energy as fat, providing an important reservoir of fuel for maintaining whole body energy homeostasis. Herein, we identify the transcriptional cofactor hairless (HR) to be required for white adipogenesis. Moreover, forced expression of HR in non-adipogenic precursor cells induces adipogenic gene expression and enhances adipocyte formation under permissive conditions. HR exerts its proadipogenic effects by regulating the expression of PPARγ, one of the central adipogenic transcription factors. In conclusion, our data provide a new mechanism required for white adipogenesis.


Assuntos
Adipócitos Brancos/citologia , Adipogenia/genética , Regulação da Expressão Gênica no Desenvolvimento , PPAR gama/metabolismo , Fatores de Transcrição/metabolismo , Células 3T3 , Adipócitos Brancos/metabolismo , Animais , Diferenciação Celular , Camundongos , Camundongos Knockout , Mutação , PPAR gama/genética , Fatores de Transcrição/genética , Transcrição Gênica
13.
EMBO Rep ; 11(11): 834-40, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20930846

RESUMO

Mitogen-activated protein kinase (MAPK) signalling occurs in response to almost any change in the extracellular or intracellular milieu that affects the metabolism of the cell, organ or the entire organism. MAPK-dependent signal transduction is required for physiological metabolic adaptation, but inappropriate MAPK signalling contributes to the development of several interdependent pathological traits, collectively known as metabolic syndrome. Metabolic syndrome leads to life-threatening clinical consequences, such as type 2 diabetes. This Review provides an overview of the MAPK-signalling mechanisms that underly basic cellular metabolism, discussing their link to disease.


Assuntos
Células/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Estresse Fisiológico , Adipócitos/enzimologia , Animais , Células/enzimologia , Humanos , Insulina/metabolismo
14.
Neuroscience ; 484: 53-65, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35031398

RESUMO

Persistent improvement of cognitive deficits in Alzheimer's disease (AD), a common form of dementia, is an unattained therapeutic objective. Gene therapy holds promise for treatment of familial and sporadic forms of AD. p38γ, a member of the p38 mitogen-activated protein (MAP) kinase family, inhibits amyloid-ß toxicity through regulation of tau phosphorylation. We recently showed that a gene delivery approach increasing p38γ resulted in markedly better learning and memory performance in mouse models of AD at advanced stages of amyloid-ß- and tau-mediated cognitive impairment. Notably, low-to-moderate expression of p38γ had beneficial outcomes on cognition. The impact of high levels of p38γ on neuronal function remain unclear. Therefore, we addressed the outcomes of high levels of active p38γ on brain function, by direct injection of p38γ-encoding adeno-associated virus (AAV) into the forebrain of aged mice of an APP transgenic AD mouse model. While motor function in p38γ-expressing APP transgenic mice 2 months post-injection was comparable to control treated APP mice, their activity was markedly reduced in the open field test and included frequent bouts of immobility. Moreover, their learning and memory function was markedly impaired compared to control-treated aged APP mice. These results suggest that high neuronal levels of active p38γ emphasize a stress kinase role of p38γ, perturbing circuit function in motivation, navigation, and spatial learning. Overall, this work shows excessive neuronal p38γ levels can aggravate circuit dysfunction and advises adjustable expression systems will be required for sustainable AD gene therapy based on p38γ activity.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Prosencéfalo/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
15.
Sci Adv ; 8(27): eabl8809, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35857446

RESUMO

Hyperphosphorylation of the neuronal tau protein is a hallmark of neurodegenerative tauopathies such as Alzheimer's disease. A central unanswered question is why tau becomes progressively hyperphosphorylated. Here, we show that tau phosphorylation is governed by interdependence- a mechanistic link between initial site-specific and subsequent multi-site phosphorylation. Systematic assessment of site interdependence identified distinct residues (threonine-50, threonine-69, and threonine-181) as master sites that determine propagation of phosphorylation at multiple epitopes. CRISPR point mutation and expression of human tau in Alzheimer's mice showed that site interdependence governs physiologic and amyloid-associated multi-site phosphorylation and cognitive deficits, respectively. Combined targeting of master sites and p38α, the most central tau kinase linked to interdependence, synergistically ablated hyperphosphorylation. In summary, our work delineates how complex tau phosphorylation arises to inform therapeutic and biomarker design for tauopathies.


Assuntos
Doença de Alzheimer , Tauopatias , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Humanos , Camundongos , Fosforilação , Tauopatias/genética , Tauopatias/metabolismo , Treonina/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
16.
IUBMB Life ; 63(7): 495-502, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21698753

RESUMO

Both Alzheimer's disease (AD) and almost every second case of frontotemporal lobar degeneration (FTLD) are characterized by the deposition of hyperphosphorylated forms of the microtubule-associated protein tau in neurons and/or glia. This unifying pathology led to coining the umbrella term "tauopathies" for these conditions. While the deposition of tau ultimately results in the formation of typical histopathological lesions, such as the neurofibrillary tangles (NFTs) in AD, it is now well accepted that tau interferes with normal functions in neurons already before its deposition. Together with the identification of pathogenic mutations in the tau-encoding gene MAPT in FTLD and evidence from a rising number of in vivo animal models a central role of tau in neurodegeneration has emerged. Here, we review the role of pathological tau in axonal transport, mitochondrial respiration, and in mediating amyloid-ß toxicity in AD. Furthermore, we review recent findings regarding the spreading of tau pathology throughout the brain as disease progresses.


Assuntos
Doenças Neurodegenerativas/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Transporte Axonal/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Humanos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/fisiopatologia , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética
17.
J Neural Transm (Vienna) ; 118(5): 737-45, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21442354

RESUMO

Neuropathological features in Alzheimer's Disease (AD) include the presence of hyperphosphorylated forms of the microtubule-associated tau protein (tau) in hippocampal neurones. Numerous studies indicate a neuroprotective effect of calcium-binding proteins (Ca2+ binding proteins) in neurodegenerative diseases (e.g., AD). Secretagogin is a newly described Ca2+ binding protein that is produced by pyramidal neurones of the human hippocampus. Recently, secretagogin expressing hippocampal neurones were demonstrated to resist tau-induced pathology in AD in contrast to the majority of neighbouring neurones. This suggested a neuroprotective effect of secretagogin in hippocampal neurones. Here, we investigated secretagogin expression in wild type (wt) mice as well as in hemizygous and homozygous P301L tau transgenic (tg) mice, which show pronounced and widespread tau pathology in hippocampal neurones. Secretagogin expression was analyzed at the immunohistochemical and biochemical levels in brains of age-matched wt and hemi- and homozygous tau tg mice. In wt mice hippocampal secretagogin-immunoreactive neurones were invariably detected, while immunoreactivity was much lower (P < 0.001) in tau tg mice. Of note, hippocampal secretagogin immunoreactivity was absent in 62.5% of homozygous tau tg mice. In line with this finding, Western blot analysis demonstrated a significant reduction in protein expression levels of secretagogin in homozygous tau tg compared to wt mice. Our results suggest that increased levels of tau negatively influence secretagogin expression in the hippocampus of tau tg mice.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Regulação da Expressão Gênica/genética , Hipocampo/metabolismo , Leucina/genética , Mutação/genética , Prolina/genética , Proteínas tau/genética , Fatores Etários , Animais , Proteínas de Ligação ao Cálcio/genética , Feminino , Genótipo , Hipocampo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Secretagoginas , Proteínas tau/metabolismo
18.
Behav Brain Res ; 397: 112943, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33017638

RESUMO

Frontotemporal dementia (FTD) and Alzheimer's disease (AD) exhibit intracellular inclusions [neurofibrillary tangles (NFT's)] of microtubule-associated protein tau that contributes to neuronal dysfunction and death. Mutations in the microtubule-associated protein tau (MAPT) gene leads to tau hyperphosphorylation and promotes NFT formation. The TAU58/2 transgenic mouse model expresses mutant human tau (P301S mutation) and exhibits behavioural abnormalities relevant to dementia in early adulthood. Here we comprehensively determined the behavioural phenotype of TAU58/2 transgenic female mice at 14 months of age using test paradigms relevant to FTD and AD. TAU58/2 females showed a significant motor deficit and lower bodyweight compared to WT littermates. Transgenic females failed to habituate to the test arena in the light-dark test. Interestingly, transgenics did not exhibit an anxiolytic-like phenotype and intermediate-term spatial learning in the cheeseboard test was intact. However, a significant learning deficit was detected in the 1st trial across test days indicating impaired long-term spatial memory. In addition, the preference for a previously rewarded location was absent in transgenic females during probe trial testing. Finally, TAU58/2 mice had a defective acoustic startle response and impaired sensorimotor gating. In conclusion TAU58/2 mice exhibit several behavioural deficits that resemble those observed in human FTD and AD. Additionally, we observed a novel startle response deficit in these mice. At 14 months of age, TAU58/2 females represent a later disease stage and are therefore a potentially useful model to test efficacy of therapeutics to reverse or ameliorate behavioural deficits in post-onset tauopapthy-related neurodegenerative disorders.


Assuntos
Comportamento Animal/fisiologia , Demência/fisiopatologia , Modelos Animais de Doenças , Reflexo de Sobressalto/fisiologia , Tauopatias/fisiopatologia , Proteínas tau/genética , Fatores Etários , Doença de Alzheimer/fisiopatologia , Animais , Feminino , Demência Frontotemporal/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Mutantes , Fenótipo
19.
Protein Sci ; 29(5): 1196-1210, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32189389

RESUMO

Mitogen-activated protein (MAP) kinase signaling is central to multiple cellular responses and processes. MAP kinase p38α is the best characterized member of the p38 MAP kinase family. Upstream factors and downstream targets of p38α have been identified in the past by conventional methods such as coimmunoprecipitation. However, a complete picture of its interaction partners and substrates in cells is lacking. Here, we employ a proximity-dependent labeling approach using biotinylation tagging to map the interactome of p38α in cultured 293T cells. Fusing the advanced biotin ligase BioID2 to the N-terminus of p38α, we used mass spectrometry to identify 37 biotin-labeled proteins that putatively interact with p38α. Gene ontology analysis confirms known upstream and downstream factors in the p38 MAP kinase cascade (e.g., MKK3, MAPKAPK2, TAB2, and c-jun). We furthermore identify a cluster of zinc finger (ZnF) domain-containing proteins that is significantly enriched among proximity-labeled interactors and is involved in gene transcription and DNA damage response. Fluorescence imaging and coimmunoprecipitation with overexpressed p38α in cells supports an interaction of p38α with ZnF protein XPA, a key factor in the DNA damage response, that is promoted by UV irradiation. These results define an extensive network of interactions of p38α in cells and new direct molecular targets of MAP kinase p38α in gene regulation and the DNA damage response.


Assuntos
Biotinilação/métodos , Carbono-Nitrogênio Ligases/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/química , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Carbono-Nitrogênio Ligases/análise , Carbono-Nitrogênio Ligases/genética , Proteínas de Escherichia coli/análise , Proteínas de Escherichia coli/genética , Células HEK293 , Humanos , Espectrometria de Massas , Proteínas Repressoras/análise , Proteínas Repressoras/genética , Transdução de Sinais/genética , Raios Ultravioleta , Proteínas Quinases p38 Ativadas por Mitógeno/genética
20.
Front Mol Neurosci ; 13: 570586, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33013322

RESUMO

Mitogen-activated protein (MAP) kinases are a central component in signaling networks in a multitude of mammalian cell types. This review covers recent advances on specific functions of p38 MAP kinases in cells of the central nervous system. Unique and specific functions of the four mammalian p38 kinases are found in all major cell types in the brain. Mechanisms of p38 activation and downstream phosphorylation substrates in these different contexts are outlined and how they contribute to functions of p38 in physiological and under disease conditions. Results in different model organisms demonstrated that p38 kinases are involved in cognitive functions, including functions related to anxiety, addiction behavior, neurotoxicity, neurodegeneration, and decision making. Finally, the role of p38 kinases in psychiatric and neurological conditions and the current progress on therapeutic inhibitors targeting p38 kinases are covered and implicate p38 kinases in a multitude of CNS-related physiological and disease states.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA