Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 418
Filtrar
1.
Mol Neurobiol ; 59(6): 3617-3634, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35359226

RESUMO

Tau is a microtubule-associated protein widely distributed in the central nervous system (CNS). The main function of tau is to promote the assembly of microtubules and stabilize their structure. After a long period of research on neurodegenerative diseases, the function and dysfunction of the microtubule-associated protein tau in neurodegenerative diseases and tau neurotoxicity have attracted increasing attention. Tauopathies are a series of progressive neurodegenerative diseases caused by pathological changes in tau, such as abnormal phosphorylation. The pathological features of tauopathies are the deposition of abnormally phosphorylated tau proteins and the aggregation of tau proteins in neurons. This article first describes the normal physiological function and dysfunction of tau proteins and then discusses the enzymes and proteins involved in tau phosphorylation and dephosphorylation, the role of tau in cell dysfunction, and the relationships between tau and several neurodegenerative diseases. The study of tau neurotoxicity provides new directions for the treatment of tauopathies.


Assuntos
Tauopatias , Proteínas tau , Humanos , Microtúbulos/metabolismo , Neurônios/metabolismo , Fosforilação , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/metabolismo , Proteínas tau/fisiologia
2.
DNA Cell Biol ; 40(9): 1185-1199, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34379990

RESUMO

Clinical progression of tauopathies may result from transcellular propagation of pathogenic Tau seeds with the possible involvement of extracellular vesicles (EVs) as transport vectors. We established a cell model for investigating EV delivery of proteins, since the mechanism regulating EV cargo delivery to recipient cells is poorly understood. In our cell model, EVs are readily internalized and accumulate in degradative organelles (DOs). We then show for the first time that in this acidic compartment, profibrillogenic Tau delivered by EVs interacts with Tau expressed by the recipient cells and cause its accumulation by a process that involves the participation of autophagy. Thus, the degradative compartment of cells may represent the subcellular site initiating a cascade of events resulting in early hallmarks of tauopathies. These are characterized by seeded Tau accumulation, pathology-associated epitopes, DO stress, and cytotoxicity. The involvement of autophagy to this process and the relative accessibility of the degradative pathway for extracellular agents, support possible modes of intervention to slow down the progression of neurodegeneration.


Assuntos
Vesículas Extracelulares/metabolismo , Organelas/metabolismo , Tauopatias , Proteínas tau/fisiologia , Animais , Linhagem Celular , Camundongos , Células-Tronco Multipotentes , Tauopatias/metabolismo , Tauopatias/patologia
3.
Biochemistry ; 60(21): 1658-1669, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34009955

RESUMO

The microtubule-associated protein tau promotes the stabilization of the axonal cytoskeleton in neurons. In several neurodegenerative diseases, such as Alzheimer's disease, tau has been found to dissociate from microtubules, leading to the formation of pathological aggregates that display an amyloid fibril-like structure. Recent structural studies have shown that the tau filaments isolated from different neurodegenerative disorders have structurally distinct fibril cores that are specific to the disease. These "strains" of tau fibrils appear to propagate between neurons in a prion-like fashion that maintains their initial template structure. In addition, the strains isolated from diseased tissue appear to have structures that are different from those made by the most commonly used in vitro modeling inducer molecule, heparin. The structural differences among strains in different diseases and in vitro-induced tau fibrils may contribute to recent failures in clinical trials of compounds designed to target tau pathology. This study identifies an isoquinoline compound (ANTC-15) isolated from the fungus Aspergillus nidulans that can both inhibit filaments induced by arachidonic acid (ARA) and disassemble preformed ARA fibrils. When compared to a tau aggregation inhibitor currently in clinical trials (LMTX, LMTM, or TRx0237), ANTC-15 and LMTX were found to have opposing inducer-specific activities against ARA and heparin in vitro-induced tau filaments. These findings may help explain the disappointing results in translating potent preclinical inhibitor candidates to successful clinical treatments.


Assuntos
Isoquinolinas/farmacologia , Tauopatias/fisiopatologia , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Amiloide/química , Aspergillus nidulans/metabolismo , Fungos/metabolismo , Humanos , Isoquinolinas/metabolismo , Microtúbulos/metabolismo , Neurônios/metabolismo , Príons/metabolismo , Agregados Proteicos/fisiologia , Agregação Patológica de Proteínas/metabolismo , Relação Estrutura-Atividade , Tauopatias/metabolismo , Proteínas tau/fisiologia
4.
Commun Biol ; 4(1): 560, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980987

RESUMO

Accumulation and spread of tau in Alzheimer's disease and other tauopathies occur in a prion-like manner. However, the mechanisms and downstream consequences of tau trafficking remain largely unknown. We hypothesized that tau traffics from neurons to microglia via extracellular vesicles (EVs), leading to IL-6 generation and cognitive impairment. We assessed mice and neurons treated with anesthetics sevoflurane and desflurane, and applied nanobeam-sensor technology, an ultrasensitive method, to measure tau/p-tau amounts. Sevoflurane, but not desflurane, increased tau or p-tau amounts in blood, neuron culture medium, or EVs. Sevoflurane increased p-tau amounts in brain interstitial fluid. Microglia from tau knockout mice took up tau and p-tau when treated with sevoflurane-conditioned neuron culture medium, leading to IL-6 generation. Tau phosphorylation inhibitor lithium and EVs generation inhibitor GW4869 attenuated tau trafficking. GW4869 mitigated sevoflurane-induced cognitive impairment in mice. Thus, tau trafficking could occur from neurons to microglia to generate IL-6, leading to cognitive impairment.


Assuntos
Transporte Proteico/efeitos dos fármacos , Sevoflurano/farmacologia , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Anestésicos Inalatórios/farmacologia , Animais , Encéfalo/metabolismo , Disfunção Cognitiva/induzido quimicamente , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/fisiologia , Feminino , Hipocampo/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação , Transporte Proteico/fisiologia , Sevoflurano/metabolismo , Tauopatias/metabolismo , Tauopatias/fisiopatologia , Proteínas tau/fisiologia
5.
Pharmacol Res ; 168: 105585, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33798735

RESUMO

Cancer is the second leading cause of death worldwide, and the World Health Organization estimates that one in six deaths globally is due to cancer. Chemotherapy is one of the major modalities used to treat advanced cancers and their metastasis. However, the existence of acquired and intrinsic resistance to anti-cancer drugs often diminishes their therapeutic effect. In order to pre-select patients who could benefit the most from these treatments, the efforts of many research groups have been focused on identification of biomarkers of therapy response. Taxanes paclitaxel (Taxol) and docetaxel (Taxotere) have been introduced as chemotherapy for treatment of cancers of ovary in 1992 and breast in 1996, respectively. Since then, clinical use of taxanes has expanded to include lung, prostate, gastric, head and neck, esophageal, pancreatic, and cervical cancers, as well as Kaposi sarcoma. Several independent molecular mechanisms have been shown to support taxane chemoresistance. One such mechanism is dependent on microtubule associated protein tau. Tau binds to the same site on the inner side of the microtubules that is also occupied by paclitaxel or docetaxel, and several studies have demonstrated that low/no tau expression significantly correlated with better response to the taxane treatment, suggesting that levels of tau expression could have a predictive value in pre-selecting patient cohorts that are likely to benefit from the treatment. However, several other studies have found no correlation between tau expression and taxane response, introducing a controversy and precluding its wide use as a predictive biomarker. Based on the knowledge of tau biology accumulated thus far, in this review we attempt to critically analyze the studies that evaluated tau as a biomarker of taxane response. Further, we identify yet unknown aspects of tau biology understanding of which is necessary for improvement of development of tau as a biomarker of response and a target for increasing response to taxane treatment.


Assuntos
Neoplasias/tratamento farmacológico , Taxoides/uso terapêutico , Proteínas tau/fisiologia , Biomarcadores , Humanos , Microtúbulos/química , Microtúbulos/metabolismo , Paclitaxel/uso terapêutico , Tubulina (Proteína)/fisiologia , Proteínas tau/química
6.
J Neurochem ; 158(2): 94-104, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33569813

RESUMO

The enteric nervous system plays a critical role in the regulation of gastrointestinal tract functions and is often referred to as the 'second brain' because it shares many features with the central nervous system. These similarities include among others a large panel of neurotransmitters, a large population of glial cells and a susceptibility to neurodegeneration. This close homology between the central and enteric nervous systems suggests that a disease process affecting the central nervous system could also involve its enteric counterpart. This was already documented in Parkinson's disease, the most common synucleinopathy, in which alpha-synuclein deposits are reported in the enteric nervous system in the vast majority of patients. Tau is another key protein involved in neurodegenerative disorders of the brain. Whether changes in tau also occur in the enteric nervous system during gut or brain disorders has just begun to be explored. The scope of the present article is therefore to review existing studies on the expression and phosphorylation pattern of tau in the enteric nervous system under physiological and pathological conditions and to discuss the possible occurrence of 'enteric tauopathies'.


Assuntos
Sistema Nervoso Entérico/metabolismo , Tauopatias/genética , Proteínas tau/genética , Animais , Microbioma Gastrointestinal , Humanos , Tauopatias/metabolismo , Proteínas tau/metabolismo , Proteínas tau/fisiologia
7.
Cell Death Differ ; 28(3): 884-899, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33473181

RESUMO

Tauopathies define a broad range of neurodegenerative diseases that encompass pathological aggregation of the microtubule-associated protein tau. Although tau aggregation is a central feature of these diseases, their underlying pathobiology is remarkably heterogeneous at the molecular level. In this review, we summarize critical differences that account for this heterogeneity and contrast the physiological and pathological functions of tau. We focus on the recent understanding of its prion-like behavior that accounts for its spread in the brain. Moreover, we acknowledge the limited appreciation about how upstream cellular changes influence tauopathy. Dysfunction of the highly conserved endosomal trafficking complex retromer is found in numerous tauopathies such as Alzheimer's disease, Pick's disease, and progressive supranuclear palsy, and we discuss how this has emerged as a major contributor to various aspects of neurodegenerative diseases. In particular, we highlight recent investigations that have elucidated the contribution of retromer dysfunction to distinct measures of tauopathy such as tau hyperphosphorylation, aggregation, and impaired cognition and behavior. Finally, we discuss the potential benefit of targeting retromer for modifying disease burden and identify important considerations with such an approach moving toward clinical translation.


Assuntos
Doenças Neurodegenerativas/metabolismo , Tauopatias/metabolismo , Proteínas tau/fisiologia , Animais , Humanos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética
8.
Neuron ; 109(5): 767-777.e5, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33472038

RESUMO

Tau is a major driver of neurodegeneration and is implicated in over 20 diseases. Tauopathies are characterized by synaptic loss and neuroinflammation, but it is unclear if these pathological events are causally linked. Tau binds to Synaptogyrin-3 on synaptic vesicles. Here, we interfered with this function to determine the role of pathogenic Tau at pre-synaptic terminals. We show that heterozygous knockout of synaptogyrin-3 is benign in mice but strongly rescues mutant Tau-induced defects in long-term synaptic plasticity and working memory. It also significantly rescues the pre- and post-synaptic loss caused by mutant Tau. However, Tau-induced neuroinflammation remains clearly upregulated when we remove the expression of one allele of synaptogyrin-3. Hence neuroinflammation is not sufficient to cause synaptic loss, and these processes are separately induced in response to mutant Tau. In addition, the pre-synaptic defects caused by mutant Tau are enough to drive defects in cognitive tasks.


Assuntos
Transtornos da Memória/fisiopatologia , Microglia/fisiologia , Terminações Pré-Sinápticas/fisiologia , Sinaptogirinas/fisiologia , Proteínas tau/fisiologia , Animais , Encefalite/fisiopatologia , Feminino , Hipocampo/fisiopatologia , Hipocampo/ultraestrutura , Masculino , Camundongos Knockout , Plasticidade Neuronal , Terminações Pré-Sinápticas/ultraestrutura , Sinaptogirinas/genética
9.
Alzheimers Dement ; 17(4): 618-628, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33196147

RESUMO

INTRODUCTION: A biphasic model for brain structural changes in preclinical Alzheimer's disease (AD) could reconcile some conflicting and paradoxical findings in observational studies and anti-amyloid clinical trials. METHODS: In this study we tested this model fitting linear versus quadratic trajectories and computed the timing of the inflection points vertexwise of cortical thickness and cortical diffusivity-a novel marker of cortical microstructure-changes in 389 participants from the Dominantly Inherited Alzheimer Network. RESULTS: In early preclinical AD, between 20 and 15 years before estimated symptom onset, we found increases in cortical thickness and decreases in cortical diffusivity followed by cortical thinning and cortical diffusivity increases in later preclinical and symptomatic stages. The inflection points 16 to 19 years before estimated symptom onset are in agreement with the start of tau biomarker alterations. DISCUSSION: These findings confirm a biphasic trajectory for brain structural changes and have direct implications when interpreting magnetic resonance imaging measures in preventive AD clinical trials.


Assuntos
Doença de Alzheimer/patologia , Córtex Cerebral/patologia , Sintomas Prodrômicos , Adulto , Doença de Alzheimer/genética , Biomarcadores/líquido cefalorraquidiano , Encéfalo , Imagem de Difusão por Ressonância Magnética , Humanos , Estudos Longitudinais , Mutação/genética , Proteínas tau/fisiologia
10.
Brain Res ; 1751: 147207, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33212022

RESUMO

Tauopathies is a class of neurodegenerative disorders which involves the transformation of physiological tau into pathogenic tau. One of the prime causes reported to drive this conversion is tau hyperphosphorylation and the subsequent propagation of pathogenic protein aggregates across the nervous system. Although past attempts have been made to deduce the details of tau propagation, yet not much is known about its mechanism. A better understanding of this aspect of disease pathology can prove to be beneficial for the development of diagnostic and therapeutic approaches. For the first time, we demonstrate that the human tau possesses an intrinsic property to spread trans-cellularly in the fly nervous system irrespective of the tau allele or the neuronal tissue type. Aggregate migration restricted by targeted down-regulation of a specific kinase, elucidates the role of hyper-phosphorylation in its movement. On the contrary to the previous models, our study delivers an easy and rapid in-vivo model for comprehensive examination of tau migration pathology. Henceforth, the developed model would not only be immensely helpful in uncovering the mechanistic in-depths of tau propagation pathology but also aid in modifier and/or drug screening for amelioration of tauopathies.


Assuntos
Agregados Proteicos/fisiologia , Transcitose/fisiologia , Proteínas tau/metabolismo , Fatores Etários , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Humanos , Movimento (Física) , Neurônios/metabolismo , Fosforilação , Tauopatias/metabolismo , Tauopatias/fisiopatologia , Proteínas tau/fisiologia
11.
Cell Rep ; 33(7): 108398, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33207193

RESUMO

To understand how neural-immune-associated genes and pathways contribute to neurodegenerative disease pathophysiology, we performed a systematic functional genomic analysis in purified microglia and bulk tissue from mouse and human AD, FTD, and PSP. We uncover a complex temporal trajectory of microglial-immune pathways involving the type 1 interferon response associated with tau pathology in the early stages, followed by later signatures of partial immune suppression and, subsequently, the type 2 interferon response. We find that genetic risk for dementias shows disease-specific patterns of pathway enrichment. We identify drivers of two gene co-expression modules conserved from mouse to human, representing competing arms of microglial-immune activation (NAct) and suppression (NSupp) in neurodegeneration. We validate our findings by using chemogenetics, experimental perturbation data, and single-cell sequencing in post-mortem brains. Our results refine the understanding of stage- and disease-specific microglial responses, implicate microglial viral defense pathways in dementia pathophysiology, and highlight therapeutic windows.


Assuntos
Demência/genética , Tauopatias/genética , Proteínas tau/metabolismo , Idoso , Animais , Encéfalo/metabolismo , Feminino , Demência Frontotemporal/genética , Redes Reguladoras de Genes/genética , Predisposição Genética para Doença , Genômica/métodos , Humanos , Terapia de Imunossupressão , Inflamação/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Cultura Primária de Células , Fatores de Risco , Tauopatias/metabolismo , Tauopatias/fisiopatologia , Proteínas tau/genética , Proteínas tau/fisiologia
12.
FASEB J ; 34(9): 12239-12254, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-33000527

RESUMO

α-Synuclein (α-syn)-induced neurotoxicity has been generally accepted as a key step in the pathogenesis of Parkinson's disease (PD). Microtubule-associated protein tau, which is considered second only to α-syn, has been repeatedly linked with PD in association studies. However, the underlying interaction between these two PD-related proteins in vivo remains unclear. To investigate how the expression of tau affects α-syn-induced neurodegeneration in vivo, we generated triple transgenic mice that overexpressed α-syn A53T mutation in the midbrain dopaminergic neurons (mDANs) with different expression levels of tau. Here, we found that tau had no significant effect on the A53T α-syn-mediated mDANs degeneration. However, tau knockout could modestly promote the formation of α-syn aggregates, accelerate the severe and progressive degeneration of parvalbumin-positive (PV+) neurons in substantia nigra pars reticulata (SNR), accompanied with anxiety-like behavior in aged PD-related α-syn A53T mice. The mechanisms may be associated with A53T α-syn-mediated specifically successive impairment of N-methyl-d-aspartate receptor subunit 2B (NR2B), postsynaptic density-95 (PSD-95) and microtubule-associated protein 1A (MAP1A) in PV+ neurons. Our study indicates that MAP1A may play a beneficial role in preserving the survival of PV+ neurons, and that inhibition of the impairment of NR2B/PSD-95/MAP1A pathway, may be a novel and preferential option to ameliorate α-syn-induced neurodegeneration.


Assuntos
Mutação , Degeneração Neural , Doença de Parkinson/etiologia , Parvalbuminas/análise , Substância Negra/patologia , alfa-Sinucleína/genética , Proteínas tau/fisiologia , Animais , Proteína 4 Homóloga a Disks-Large/fisiologia , Proteínas de Homeodomínio/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/fisiologia , Doença de Parkinson/patologia , Fragmentos de Peptídeos/fisiologia , Agregados Proteicos , Receptores de N-Metil-D-Aspartato/fisiologia , Fatores de Transcrição/fisiologia , alfa-Sinucleína/fisiologia , Proteínas tau/química , Proteínas tau/genética
13.
Neurobiol Aging ; 95: 214-224, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32858248

RESUMO

Tau is a microtubule-associated protein involved in Alzheimer's disease. However, little is known on its physiological function in the healthy central nervous system. Here, we observed that the expression of Tau isoforms was modulated by neuronal maturation and visual experience in the mouse retina and in the visual cortex. The visual function of wild-type (WT) and Tau knockout (KO) mice was evaluated using the optokinetic reflex (OKR), an innate visuomotor behavior, and by electroretinography. Visual tests did not reveal functional impairments in young adult and old Tau KO animals. Moreover, monocular deprivation (MD) was used to increase OKR sensitivity, a plasticity phenomenon depending on the visual cortex. MD-induced OKR sensitivity enhancement was significantly stronger in Tau KO than in WT mice suggesting that Tau restricts visual plasticity. In addition, human Tau expression did not affect visual function and plasticity in a mouse tauopathy model, relative to WT controls. Our results unveil a novel function for Tau in the adaptive mechanisms of plasticity operating in the adult brain subjected to sensory experience changes.


Assuntos
Envelhecimento/metabolismo , Envelhecimento/fisiologia , Plasticidade Neuronal/genética , Córtex Visual/fisiologia , Proteínas tau/metabolismo , Proteínas tau/fisiologia , Adaptação Fisiológica/genética , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos Knockout , Retina/metabolismo , Tauopatias/fisiopatologia , Córtex Visual/metabolismo
14.
Acta Neuropathol ; 140(4): 417-447, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32728795

RESUMO

Tau and amyloid beta (Aß) are the prime suspects for driving pathology in Alzheimer's disease (AD) and, as such, have become the focus of therapeutic development. Recent research, however, shows that these proteins have been highly conserved throughout evolution and may have crucial, physiological roles. Such functions may be lost during AD progression or be unintentionally disrupted by tau- or Aß-targeting therapies. Tau has been revealed to be more than a simple stabiliser of microtubules, reported to play a role in a range of biological processes including myelination, glucose metabolism, axonal transport, microtubule dynamics, iron homeostasis, neurogenesis, motor function, learning and memory, neuronal excitability, and DNA protection. Aß is similarly multifunctional, and is proposed to regulate learning and memory, angiogenesis, neurogenesis, repair leaks in the blood-brain barrier, promote recovery from injury, and act as an antimicrobial peptide and tumour suppressor. This review will discuss potential physiological roles of tau and Aß, highlighting how changes to these functions may contribute to pathology, as well as the implications for therapeutic development. We propose that a balanced consideration of both the physiological and pathological roles of tau and Aß will be essential for the design of safe and effective therapeutics.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/fisiologia , Proteínas tau/fisiologia , Animais , Humanos
15.
Brain Pathol ; 30(5): 913-925, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32500646

RESUMO

Chronic traumatic encephalopathy (CTE) is a neurodegenerative tauopathy characterized by accumulation of hyperphosphorylated tau (p-tau) in perivascular aggregates in neurons and glia at the depths of neocortical sulci and progresses to diffuse neocortical, allocortical and brainstem structures. The strongest risk factor is exposure to repetitive head impacts acquired most commonly through contact sports and military service. Given that CTE can only be definitively diagnosed after death, a better understanding of the cellular and molecular changes in CTE brains may lead to identification of mechanisms that could be used for novel biomarkers, monitoring progression or therapeutic development. Disruption of alternative pre-mRNA splicing of tau mRNA plays a pathogenic role in tauopathy, with multiple characteristic patterns of isoform accumulation varying among tauopathies. Limited data are available on CTE, particularly at early stages. Using biochemical and histological approaches, we performed a detailed characterization of tau isoform signatures in post-mortem human brain tissue from individuals with a range of CTE stages (n = 99). In immunoblot analyses, severity was associated with decreased total monomeric tau and increased total oligomeric tau. Immunoblot with isoform-specific antisera revealed that oligomeric tau with three and four microtubule binding domain repeats (3R and 4R) also increased with CTE severity. Similarly, immunohistochemical studies revealed p-tau accumulation consisting of both 3R and 4R in perivascular lesions. When the ratio of 4R:3R was analyzed, there was mixed expression throughout CTE stages, although 4R predominated in early CTE stages (I-II), a 3R shift was observed in later stages (III-IV). While neurons were found to contain both 3R and 4R, astrocytes only contained 4R. These 4R-positive cells were exclusively neuronal at early stages. Overall, these findings demonstrate that CTE is a mixed 4R/3R tauopathy. Furthermore, histologic analysis reveals a progressive shift in tau isoforms that correlates with CTE stage and extent of neuronal pathology.


Assuntos
Encefalopatia Traumática Crônica/patologia , Tauopatias/patologia , Proteínas tau/metabolismo , Adulto , Doença de Alzheimer/patologia , Astrócitos/patologia , Autopsia , Encéfalo/patologia , Encefalopatia Traumática Crônica/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Neuroglia/metabolismo , Neurônios/metabolismo , Isoformas de Proteínas/metabolismo , Tauopatias/metabolismo , Proteínas tau/fisiologia
16.
Urol Oncol ; 38(6): 605.e9-605.e17, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32139291

RESUMO

INTRODUCTION: Microtubule-associated protein tau (MAPT) overexpression has been linked to poor prognosis in several cancers. MAPT-AS1 is a long noncoding RNA existing at the antisense strand of the MAPT promoter region. The clinical significance of MAPT and MAPT-AS-1 in clear cell renal cell carcinoma (ccRCC) is unknown. This study aimed to assess the expression and function of MAPT and MAPT-AS1 in ccRCC. METHODS: The expression of MAPT was determined using immunohistochemistry in ccRCC. The effects of MAPT knockdown on cell growth and invasion were evaluated and the interaction between MAPT and microtubule-associated protein tau antisense (MAPT-AS1) were analyzed. The expression of MAPT-AS1 was determined using quantitative reverse transcription polymerase chain reaction in ccRCC tissues. We investigated the effect of MAPT-AS1 knockdown on cell growth and invasion. We analyzed the regulation of MAPT and MAPT-AS1. RESULTS: Immunohistochemistry in 135 ccRCC cases showed that 61% of the cases were positive for MAPT. Kaplan-Meier analysis showed that the low expression of MAPT was associated with poor overall survival after nephrectomy. Knockdown of MAPT enhanced cell growth and invasion. quantitative reverse transcription polymerase chain reaction revealed a positive correlation between MAPT and MAPT-AS1. The expression of MAPT-AS1 was higher in ccRCC tissue than in nonneoplastic kidney tissue. Kaplan-Meier analysis showed that the low expression of MAPT-AS1 was associated with poor overall survival after nephrectomy by in silico analysis. MAPT-AS1 knockdown promoted cell growth and invasion activity. P53 knockout suppressed the expression of MAPT and MAPT-AS1. CONCLUSION: These results suggest that MAPT and MAPT-AS1 may be promising predictive biomarkers for survival and play a tumor-suppressive role in ccRCC.


Assuntos
Biomarcadores Tumorais/fisiologia , Carcinoma de Células Renais/etiologia , Neoplasias Renais/etiologia , Proteínas Supressoras de Tumor/fisiologia , Proteínas tau/fisiologia , Idoso , Biomarcadores Tumorais/biossíntese , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/mortalidade , Feminino , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/mortalidade , Masculino , Pessoa de Meia-Idade , Prognóstico , RNA Longo não Codificante , Taxa de Sobrevida , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/genética , Proteínas tau/biossíntese , Proteínas tau/genética
17.
Brain Res Bull ; 156: 76-85, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31931119

RESUMO

OBJECTIVE: Diabetes mellitus (DM) can increase the risk of cognitive dysfunction, but its exact mechanisms remain unclear. The involvement of aberrant O-GlcNAcylation has been identified in hyperglycemia and DM, as well as the pathogenesis of Alzheimer's disease via competition with tau phosphorylation. This study was designed to investigate the role of O-GlcNAcylation in diabetes-associated cognitive dysfunction (DACD). METHODS: Fifteen-week old male KK-Ay mice were used as DACD models, and advanced glycation end product (AGE)-treated HT22 cells were used as a model of high glucose toxicity. Morris water maze tests, histological staining, real-time quantitative PCR, and Western blot were also applied. RESULTS: Mice with DACD exhibited evident obesity, hyperinsulinemia, hyperglycemia, and impaired learning and memory function. O-GlcNAcylation levels decreased and tau phosphorylation levels at Ser396, Ser404, Thr212, and Thr231 increased in the hippocampus of mice with DACD, as well as in AGE-treated HT22 cells. Hypoglycemic therapy improved these anomalies and elevated O-GlcNAc transferase (OGT) levels in mice with DACD. OGT plasmid transfection in HT22 cells partially reversed AGE-induced decreases in O-GlcNAcylation levels and increased tau phosphorylation levels. CONCLUSIONS: Chronic hyperglycemia can induce tau hyperphosphorylation by downregulating OGT-involved O-GlcNAcylation in vivo and in vitro, which mediates DACD.


Assuntos
Disfunção Cognitiva/fisiopatologia , Hiperglicemia/fisiopatologia , Proteínas tau/metabolismo , Acilação , Animais , Encéfalo/metabolismo , Disfunção Cognitiva/metabolismo , Diabetes Mellitus/fisiopatologia , Modelos Animais de Doenças , Hipocampo/metabolismo , Hiperglicemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , N-Acetilglucosaminiltransferases/metabolismo , N-Acetilglucosaminiltransferases/fisiologia , Fosforilação , beta-N-Acetil-Hexosaminidases/metabolismo , beta-N-Acetil-Hexosaminidases/fisiologia , Proteínas tau/fisiologia
18.
PLoS One ; 14(12): e0226380, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31821364

RESUMO

Tauopathies are a class of neurodegenerative diseases characterized by the abnormal phosphorylation and accumulation of the microtubule-associated protein, tau, in both neuronal and glial cells. Though tau pathology in glial cells is a prominent feature of many of these disorders, the pathological contribution of these lesions to tauopathy pathogenesis remains largely unknown. Moreover, while tau pathology is predominantly found in the central nervous system, a role for tau in the cells of the peripheral nervous system has been described, though not well characterized. To investigate the effects of glial tau expression on the development and maintenance of the peripheral nervous system, we utilized a Drosophila melanogaster model of tauopathy that expresses human wild-type tau in glial cells during development. We found that glial tau expression during development results in larval locomotor deficits and organismal lethality at the pupal stage, without affecting larval neuromuscular junction synapse development or post-synaptic amplitude. There was, however, a significant decrease in the decay time of synaptic potentials upon repeated stimulation of the motoneuron. Behavioral abnormalities were accompanied by glial cell death, disrupted maintenance of glial-axonal integrity, and the abnormal accumulation of the presynaptic protein, Bruchpilot, in peripheral nerve axons. Together, these data demonstrate that human tau expression in Drosophila glial cells does not affect neuromuscular junction synapse formation during development, but is deleterious to the maintenance of glial-axonal interactions in the peripheral nervous system.


Assuntos
Neurônios Motores/fisiologia , Neuroglia/fisiologia , Sistema Nervoso Periférico/fisiopatologia , Tauopatias/fisiopatologia , Proteínas tau/metabolismo , Animais , Axônios/fisiologia , Modelos Animais de Doenças , Drosophila melanogaster , Humanos , Neuroglia/metabolismo , Proteínas tau/fisiologia
19.
Neuroscience ; 422: 65-74, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31689387

RESUMO

Accumulation of microtubule associated protein tau in the substantia nigra is associated with several tauopathies including progressive supranuclear palsy (PSP). A number of studies have used mutant tau transgenic mouse model to mimic the neuropathology of tauopathies and disease phenotypes. However, tau expression in these transgenic mouse models is not specific to brain subregions, and may not recapitulate subcortical disease phenotypes of PSP. It is necessary to develop a new disease modeling system for cell and region-specific expression of pathogenic tau for modeling PSP in mouse brain. In this study, we developed a novel strategy to express P301L mutant tau to the dopaminergic neurons of substantia nigra by coupling tyrosine hydroxylase promoter Cre-driver mice with a Cre-inducible adeno-associated virus (iAAV). The results showed that P301L mutant tau was successfully transduced in the dopaminergic neurons of the substantia nigra at the presence of Cre recombinase and iAAV. Furthermore, the iAAV-tau-injected mice displayed severe motor deficits including impaired movement ability, motor balance, and motor coordination compared to the control groups over a short time-course. Immunochemical analysis revealed that tau gene transfer significantly resulted in loss of tyrosine hydroxylase-positive dopaminergic neurons and elevated phosphorylated tau in the substantia nigra. Our development of dopaminergic neuron-specific neurodegenerative mouse model with tauopathy will be helpful for studying the underlying mechanism of pathological protein propagation as well as development of new therapies.


Assuntos
Dependovirus , Transtornos Motores/fisiopatologia , Degeneração Neural/patologia , Substância Negra/patologia , Proteínas tau/fisiologia , Animais , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Vetores Genéticos , Integrases , Camundongos Transgênicos , Transtornos Motores/genética , Mutação , Fosforilação , Substância Negra/metabolismo , Paralisia Supranuclear Progressiva/genética , Transdução Genética , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas tau/biossíntese , Proteínas tau/genética , Proteínas tau/metabolismo
20.
J Alzheimers Dis ; 71(4): 1125-1138, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31524157

RESUMO

Abnormal folding and aggregation of the microtubule-associated protein, tau, is a hallmark of several neurodegenerative disorders, including Alzheimer's disease (AD). Although normal tau is an intrinsically disordered protein, it does exhibit tertiary structure whereby the N- and C-termini are often in close proximity to each other and to the contiguous microtubule-binding repeat domains that extend C-terminally from the middle of the protein. Unfolding of this paperclip-like conformation might precede formation of toxic tau oligomers and filaments, like those found in AD brain. While there are many ways to monitor tau aggregation, methods to monitor changes in tau folding are not well established. Using full length human 2N4R tau doubly labeled with the Förster resonance energy transfer (FRET) compatible fluorescent proteins, Venus and Teal, on the N- and C-termini, respectively (Venus-Tau-Teal), intensity and lifetime FRET measurements were able to distinguish folded from unfolded tau in living cells independently of tau-tau intermolecular interactions. When expression was restricted to low levels in which tau-tau aggregation was minimized, Venus-Tau-Teal was sensitive to microtubule binding, phosphorylation, and pathogenic oligomers. Of particular interest is our finding that amyloid-ß oligomers (AßOs) trigger Venus-Tau-Teal unfolding in cultured mouse neurons. We thus provide direct experimental evidence that AßOs convert normally folded tau into a conformation thought to predominate in toxic tau aggregates. This finding provides further evidence for a mechanistic connection between Aß and tau at seminal stages of AD pathogenesis.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Agregação Patológica de Proteínas , Proteínas tau/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Sítios de Ligação , Células Cultivadas , Humanos , Proteínas Intrinsicamente Desordenadas/metabolismo , Camundongos , Microtúbulos/fisiologia , Neurônios/fisiologia , Dobramento de Proteína , Resposta a Proteínas não Dobradas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...