Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 421
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Neurosci ; 37: 79-100, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24821312

RESUMEN

The vast majority of Alzheimer's disease (AD) cases are late onset (LOAD), which is genetically complex with heritability estimates up to 80%. Apolipoprotein E (APOE) has been irrefutably recognized as the major genetic risk factor, with semidominant inheritance, for LOAD. Although the mechanisms that underlie the pathogenic nature of APOE in AD are still not completely understood, emerging data suggest that APOE contributes to AD pathogenesis through both amyloid-ß (Aß)-dependent and Aß-independent pathways. Given the central role for APOE in the modulation of AD pathogenesis, many therapeutic strategies have emerged, including converting APOE conformation, regulating APOE expression, mimicking APOE peptides, blocking the APOE/Aß interaction, modulating APOE lipidation state, and gene therapy. Accumulating evidence also suggests the utility of APOE genotyping in AD diagnosis, risk assessment, prevention, and treatment response.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Apolipoproteínas E/fisiología , Terapia Molecular Dirigida/métodos , Proteínas tau/fisiología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/fisiología , Apolipoproteínas E/efectos de los fármacos , Apolipoproteínas E/genética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Modelos Biológicos , Transducción de Señal/fisiología , Proteínas tau/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(12): 5765-5774, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30842285

RESUMEN

Mutations in the vacuolar protein sorting 35 ortholog (VPS35) gene represent a cause of late-onset, autosomal dominant familial Parkinson's disease (PD). A single missense mutation, D620N, is considered pathogenic based upon its segregation with disease in multiple families with PD. At present, the mechanism(s) by which familial VPS35 mutations precipitate neurodegeneration in PD are poorly understood. Here, we employ a germline D620N VPS35 knockin (KI) mouse model of PD to formally establish the age-related pathogenic effects of the D620N mutation at physiological expression levels. Our data demonstrate that a heterozygous or homozygous D620N mutation is sufficient to reproduce key neuropathological hallmarks of PD as indicated by the progressive degeneration of nigrostriatal pathway dopaminergic neurons and widespread axonal pathology. Unexpectedly, endogenous D620N VPS35 expression induces robust tau-positive somatodendritic pathology throughout the brain as indicated by abnormal hyperphosphorylated and conformation-specific tau, which may represent an important and early feature of mutant VPS35-induced neurodegeneration in PD. In contrast, we find no evidence for α-synuclein-positive neuropathology in aged VPS35 KI mice, a hallmark of Lewy body pathology in PD. D620N VPS35 expression also fails to modify the lethal neurodegenerative phenotype of human A53T-α-synuclein transgenic mice. Finally, by crossing VPS35 KI and null mice, our data demonstrate that a single D620N VPS35 allele is sufficient for survival and early maintenance of dopaminergic neurons, indicating that the D620N VPS35 protein is fully functional. Our data raise the tantalizing possibility of a pathogenic interplay between mutant VPS35 and tau for inducing neurodegeneration in PD.


Asunto(s)
Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/fisiología , Proteínas tau/metabolismo , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/fisiología , Técnicas de Sustitución del Gen , Masculino , Ratones , Mutación , Enfermedades del Sistema Nervioso/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología , Neuropatología , Enfermedad de Parkinson/genética , Transporte de Proteínas , alfa-Sinucleína/metabolismo , Proteínas tau/fisiología
3.
Biochemistry ; 60(21): 1658-1669, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34009955

RESUMEN

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.


Asunto(s)
Isoquinolinas/farmacología , Tauopatías/fisiopatología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Amiloide/química , Aspergillus nidulans/metabolismo , Hongos/metabolismo , Humanos , Isoquinolinas/metabolismo , Microtúbulos/metabolismo , Neuronas/metabolismo , Priones/metabolismo , Agregado de Proteínas/fisiología , Agregación Patológica de Proteínas/metabolismo , Relación Estructura-Actividad , Tauopatías/metabolismo , Proteínas tau/fisiología
4.
J Neurochem ; 158(2): 94-104, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33569813

RESUMEN

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'.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Tauopatías/genética , Proteínas tau/genética , Animales , Microbioma Gastrointestinal , Humanos , Tauopatías/metabolismo , Proteínas tau/metabolismo , Proteínas tau/fisiología
5.
Annu Rev Genet ; 47: 601-23, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24274755

RESUMEN

Prions are proteins that acquire alternative conformations that become self-propagating. Transformation of proteins into prions is generally accompanied by an increase in ß-sheet structure and a propensity to aggregate into oligomers. Some prions are beneficial and perform cellular functions, whereas others cause neurodegeneration. In mammals, more than a dozen proteins that become prions have been identified, and a similar number has been found in fungi. In both mammals and fungi, variations in the prion conformation encipher the biological properties of distinct prion strains. Increasing evidence argues that prions cause many neurodegenerative diseases (NDs), including Alzheimer's, Parkinson's, Creutzfeldt-Jakob, and Lou Gehrig's diseases, as well as the tauopathies. The majority of NDs are sporadic, and 10% to 20% are inherited. The late onset of heritable NDs, like their sporadic counterparts, may reflect the stochastic nature of prion formation; the pathogenesis of such illnesses seems to require prion accumulation to exceed some critical threshold before neurological dysfunction manifests.


Asunto(s)
Enfermedades Neurodegenerativas/etiología , Priones/fisiología , Edad de Inicio , Proteínas Amiloidogénicas/química , Proteínas Amiloidogénicas/clasificación , Proteínas Amiloidogénicas/fisiología , Animales , Proteínas Fúngicas/química , Proteínas Fúngicas/clasificación , Proteínas Fúngicas/fisiología , Humanos , Cuerpos de Inclusión , Mamíferos , Modelos Moleculares , Enfermedades Neurodegenerativas/epidemiología , Enfermedades Neurodegenerativas/genética , Ovillos Neurofibrilares , Factores de Terminación de Péptidos/química , Factores de Terminación de Péptidos/clasificación , Factores de Terminación de Péptidos/fisiología , Placa Amiloide , Enfermedades por Prión/etiología , Enfermedades por Prión/genética , Priones/genética , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/clasificación , Proteínas de Saccharomyces cerevisiae/fisiología , Sinucleínas/fisiología , Tauopatías/etiología , Tauopatías/genética , Factores de Transcripción/química , Factores de Transcripción/clasificación , Virulencia , Factores de Escisión y Poliadenilación de ARNm/química , Factores de Escisión y Poliadenilación de ARNm/clasificación , Proteínas tau/genética , Proteínas tau/fisiología
6.
FASEB J ; 34(9): 12239-12254, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-33000527

RESUMEN

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.


Asunto(s)
Mutación , Degeneración Nerviosa , Enfermedad de Parkinson/etiología , Parvalbúminas/análisis , Sustancia Negra/patología , alfa-Sinucleína/genética , Proteínas tau/fisiología , Animales , Homólogo 4 de la Proteína Discs Large/fisiología , Proteínas de Homeodominio/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/fisiología , Enfermedad de Parkinson/patología , Fragmentos de Péptidos/fisiología , Agregado de Proteínas , Receptores de N-Metil-D-Aspartato/fisiología , Factores de Transcripción/fisiología , alfa-Sinucleína/fisiología , Proteínas tau/química , Proteínas tau/genética
7.
Pharmacol Res ; 168: 105585, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33798735

RESUMEN

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.


Asunto(s)
Neoplasias/tratamiento farmacológico , Taxoides/uso terapéutico , Proteínas tau/fisiología , Biomarcadores , Humanos , Microtúbulos/química , Microtúbulos/metabolismo , Paclitaxel/uso terapéutico , Tubulina (Proteína)/fisiología , Proteínas tau/química
8.
Alzheimers Dement ; 17(4): 618-628, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33196147

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/patología , Corteza Cerebral/patología , Síntomas Prodrómicos , Adulto , Enfermedad de Alzheimer/genética , Biomarcadores/líquido cefalorraquídeo , Encéfalo , Imagen de Difusión por Resonancia Magnética , Humanos , Estudios Longitudinales , Mutación/genética , Proteínas tau/fisiología
9.
Acta Neuropathol ; 140(4): 417-447, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32728795

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/fisiología , Proteínas tau/fisiología , Animales , Humanos
10.
J Biol Chem ; 293(22): 8462-8472, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29632073

RESUMEN

Tau hyperphosphorylation at several sites, including those close to the microtubule domain region (MDr), is considered a key pathological event in the development of Alzheimer's disease (AD). Recent studies indicate that at the very early stage of this disease, increased phosphorylation in Tau's MDr domain correlates with reduced levels of neuronal excitability. Mechanistically, we show that pyramidal neurons and some parvalbumin-positive interneurons in 1-month-old triple-transgenic AD mice accumulate hyperphosphorylated Tau protein and that this accumulation correlates with changes in theta oscillations in hippocampal neurons. Pyramidal neurons from young triple-transgenic AD mice exhibited less spike accommodation and power increase in subthreshold membrane oscillations. Furthermore, triple-transgenic AD mice challenged with the potassium channel blocker 4-aminopyridine had reduced theta amplitude compared with 4-aminopyridine-treated control mice and, unlike these controls, displayed no seizure-like activity after this challenge. Collectively, our results provide new insights into AD pathogenesis and suggest that increases in Tau phosphorylation at the initial stages of the disease represent neuronal responses that compensate for brain circuit overexcitation.


Asunto(s)
Potenciales de Acción , Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Hipocampo/patología , Células Piramidales/patología , Ritmo Teta/fisiología , Proteínas tau/fisiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Células Cultivadas , Hipocampo/metabolismo , Masculino , Ratones , Ratones Transgénicos , Fosforilación , Células Piramidales/metabolismo
11.
Mol Psychiatry ; 23(12): 2363-2374, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30171212

RESUMEN

The typical abnormalities observed in the brain of Alzheimer's disease (AD) patients include synaptic alterations, neuronal death, brain inflammation, and the accumulation of protein aggregates in the form of amyloid plaques and neurofibrillary tangles. Despite the development of many animal and in vitro models for AD, there is a lack of an experimental approach that fully recapitulates essential aspects of the disease in human cells. Here, we report the generation of a new model to study AD, consisting of cerebral organoids (COs) produced from human-induced pluripotent stem cells (iPSCs). Under our experimental conditions, COs grow to form three-dimensional (3D) structures containing neural areas with cortical-like organization. Analysis of COs by histological and biochemical methods revealed that organoids produced from iPSCs derived from patients affected by familial AD or Down syndrome (DS) spontaneously develop over time pathological features of AD, including accumulation of structures highly reminiscent to amyloid plaques and neurofibrillary tangles. These pathological abnormalities were not observed in COs generated from various controls, including human iPSCs from healthy individuals, human iPSCs from patients affected by Creutzfeldt-Jakob disease, mouse embryonic stem cells (ESCs), or mouse iPSCs. These findings enable modeling genetic AD in a human cellular context in a 3D cortical-like tissue developed in vitro from patient-specific stem cells. This system provides a more relevant disease model compared to pre-existing methods and offers a new platform for discovery of novel targets and screening of drugs for therapeutic intervention.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Organoides/metabolismo , Proteínas tau/metabolismo , Anciano , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/fisiología , Encéfalo/metabolismo , Técnicas de Cultivo de Célula/métodos , Corteza Cerebral , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lactante , Masculino , Persona de Mediana Edad , Ovillos Neurofibrilares/patología , Neuronas/metabolismo , Fosforilación , Placa Amiloide/metabolismo , Proteínas tau/genética , Proteínas tau/fisiología
12.
Mol Psychiatry ; 23(6): 1530-1540, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28696431

RESUMEN

Changes in synaptic excitability and reduced brain metabolism are among the earliest detectable alterations associated with the development of Alzheimer's disease (AD). Stimulation of synaptic activity has been shown to be protective in models of AD beta-amyloidosis. Remarkably, deep brain stimulation (DBS) provides beneficial effects in AD patients, and represents an important therapeutic approach against AD and other forms of dementia. While several studies have explored the effect of synaptic activation on beta-amyloid, little is known about Tau protein. In this study, we investigated the effect of synaptic stimulation on Tau pathology and synapses in in vivo and in vitro models of AD and frontotemporal dementia (FTD). We found that chronic DBS or chemically induced synaptic stimulation reduced accumulation of pathological forms of Tau and protected synapses, while chronic inhibition of synaptic activity worsened Tau pathology and caused detrimental effects on pre- and post-synaptic markers, suggesting that synapses are affected. Interestingly, degradation via the proteasomal system was not involved in the reduction of pathological Tau during stimulation. In contrast, chronic synaptic activation promoted clearance of Tau oligomers by autophagosomes and lysosomes. Chronic inhibition of synaptic activity resulted in opposite outcomes, with build-up of Tau oligomers in enlarged auto-lysosomes. Our data indicate that synaptic activity counteracts the negative effects of Tau in AD and FTD by acting on autophagy, providing a rationale for therapeutic use of DBS and synaptic stimulation in tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Sinapsis/metabolismo , Tauopatías/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Autofagia/fisiología , Encéfalo/metabolismo , Estimulación Encefálica Profunda/métodos , Modelos Animales de Enfermedad , Femenino , Demencia Frontotemporal/metabolismo , Hipocampo/patología , Humanos , Lisosomas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Fármacos Neuroprotectores/metabolismo , Proteínas tau/fisiología
13.
Amino Acids ; 51(3): 513-528, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30604097

RESUMEN

Tauopathies consist of intracellular accumulation of hyperphosphorylated and aggregated microtubule protein tau, which remains a histopathological feature of Alzheimer's disease (AD) and frontotemporal dementia. L-Arginine is a semi-essential amino acid with a number of bioactive molecules. Its downstream metabolites putrescine, spermidine, and spermine (polyamines) are critically involved in microtubule assembly and stabilization. Recent evidence implicates altered arginine metabolism in the pathogenesis of AD. Using high-performance liquid chromatographic and mass spectrometric assays, the present study systematically determined the tissue concentrations of L-arginine and its nine downstream metabolites in the frontal cortex, hippocampus, parahippocampal region, striatum, thalamus, and cerebellum in male PS19 mice-bearing human tau P301S mutation at 4, 8, and 12-14 months of age. As compared to their wild-type littermates, PS19 mice displayed early and/or prolonged increases in L-ornithine and altered polyamine levels with age. There were also genotype- and age-related changes in L-arginine, L-citrulline, glutamine, glutamate, and γ-aminobutyric acid in a region- and/or chemical-specific manner. The results demonstrate altered brain arginine metabolism in PS19 mice with the most striking changes in L-ornithine, polyamines, and glutamate, indicating a shift of L-arginine metabolism to favor the arginase-polyamine pathway. Given the role of polyamines in maintaining microtubule stability, the functional significance of these changes remains to be explored in future research.


Asunto(s)
Arginina/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Tauopatías/metabolismo , Proteínas tau/fisiología , Animales , Encéfalo/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Tauopatías/patología
14.
Curr HIV/AIDS Rep ; 16(1): 66-75, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30778853

RESUMEN

PURPOSE OF REVIEW: The purpose of this review is to summarize recent developments in PET imaging of neuropathologies underlying HIV-associated neurocognitive dysfunction (HAND). We concentrate on the recent post antiretroviral era (ART), highlighting clinical and preclinical brain PET imaging studies. RECENT FINDINGS: In the post ART era, PET imaging has been used to better understand perturbations of glucose metabolism, neuroinflammation, the function of neurotransmitter systems, and amyloid/tau protein deposition in the brains of HIV-infected patients and HIV animal models. Preclinical and translational findings from those studies shed a new light on the complex pathophysiology underlying HAND. The molecular imaging capabilities of PET in neuro-HIV are great complements for structural imaging modalities. Recent and future PET imaging studies can improve our understanding of neuro-HIV and provide biomarkers of disease progress that could be used as surrogate endpoints in the evaluation of the effectiveness of potential neuroprotective therapies.


Asunto(s)
Complejo SIDA Demencia/diagnóstico por imagen , Complejo SIDA Demencia/fisiopatología , Encéfalo/diagnóstico por imagen , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/fisiopatología , Tomografía de Emisión de Positrones/métodos , Amiloide/fisiología , Animales , Antirretrovirales/uso terapéutico , Biomarcadores , Glucosa/metabolismo , Infecciones por VIH/tratamiento farmacológico , Humanos , Proteínas tau/fisiología
15.
Brain ; 141(9): 2685-2699, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30084913

RESUMEN

Traumatic brain injury is a risk factor for subsequent neurodegenerative disease, including chronic traumatic encephalopathy, a tauopathy mostly associated with repetitive concussion and blast, but not well recognized as a consequence of severe traumatic brain injury. Here we show that a single severe brain trauma is associated with the emergence of widespread hyperphosphorylated tau pathology in a proportion of humans surviving late after injury. In parallel experimental studies, in a model of severe traumatic brain injury in wild-type mice, we found progressive and widespread tau pathology, replicating the findings in humans. Brain homogenates from these mice, when inoculated into the hippocampus and overlying cerebral cortex of naïve mice, induced widespread tau pathology, synaptic loss, and persistent memory deficits. These data provide evidence that experimental brain trauma induces a self-propagating tau pathology, which can be transmitted between mice, and call for future studies aimed at investigating the potential transmissibility of trauma associated tau pathology in humans.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Tauopatías/etiología , Tauopatías/fisiopatología , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/patología , Conmoción Encefálica/patología , Lesiones Traumáticas del Encéfalo/fisiopatología , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Enfermedades Neurodegenerativas/patología , Fosforilación , Proteínas tau/metabolismo , Proteínas tau/fisiología
16.
Brain ; 141(9): 2740-2754, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30052812

RESUMEN

Alzheimer's disease is characterized by the histopathological presence of amyloid-ß plaques and tau-containing neurofibrillary tangles. Microglial activation is also a recognized pathological component. The relationship between microglial activation and protein aggregation is still debated. We investigated the relationship between amyloid plaques, tau tangles and activated microglia using PET imaging. Fifty-one subjects (19 healthy controls, 16 mild cognitive impairment and 16 Alzheimer's disease subjects) participated in the study. All subjects had neuropsychometric testing, MRI, amyloid (18F-flutemetamol), and microglial (11C-PBR28) PET. All subjects with mild cognitive impairment and Alzheimer's disease and eight of the controls had tau (18F-AV1451) PET. 11C-PBR28 PET was analysed using Logan graphical analysis with an arterial plasma input function, while 18F-flutemetamol and 18F-AV1451 PET were analysed as target:cerebellar ratios to create parametric standardized uptake value ratio maps. Biological parametric mapping in the Statistical Parametric Mapping platform was used to examine correlations between uptake of tracers at a voxel-level. There were significant widespread clusters of positive correlation between levels of microglial activation and tau aggregation in both the mild cognitive impairment (amyloid-positive and amyloid-negative) and Alzheimer's disease subjects. The correlations were stronger in Alzheimer's disease than in mild cognitive impairment, suggesting that these pathologies increase together as disease progresses. Levels of microglial activation and amyloid deposition were also correlated, although in a different spatial distribution; correlations were stronger in mild cognitive impairment than Alzheimer's subjects, in line with a plateauing of amyloid load with disease progression. Clusters of positive correlations between microglial activation and protein aggregation often targeted similar areas of association cortex, indicating that all three processes are present in specific vulnerable brain areas. For the first time using PET imaging, we show that microglial activation can correlate with both tau aggregation and amyloid deposition. This confirms the complex relationship between these processes. These results suggest that preventative treatment for Alzheimer's disease should target all three processes.


Asunto(s)
Enfermedad de Alzheimer/patología , Disfunción Cognitiva/patología , Microglía/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/fisiología , Amiloidosis/patología , Encéfalo/patología , Mapeo Encefálico/métodos , Disfunción Cognitiva/metabolismo , Progresión de la Enfermedad , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Microglía/fisiología , Persona de Mediana Edad , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/fisiología , Pruebas Neuropsicológicas , Placa Amiloide/patología , Tomografía de Emisión de Positrones/métodos , Proteínas tau/metabolismo , Proteínas tau/fisiología
17.
Alzheimers Dement ; 15(9): 1195-1207, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31420203

RESUMEN

INTRODUCTION: We classified individuals based on their baseline performance on cognitive measures and investigated the association between cognitive classifications and neuropathological findings ∼7 years later, as an external validator. METHODS: Brain autopsies of 779 decedents were examined. Baseline latent class analysis on 10 neuropsychological measures was previously assigned: mixed-domains impairment (n = 39, 5%), memory-specific impairment (n = 210, 27%), frontal impairment (n = 113, 14.5%), average cognition (n = 360, 46.2%), and superior cognition (n = 57, 7.3%). Linear regressions and risks ratios were used to examine the relation of latent class assignment at enrollment with neuropathological indices. RESULTS: Amyloid ß, tau, and transactive response DNA-binding protein 43 were associated with mixed-domains impairment and memory-specific impairment classes ∼7 years before death. Moderate arteriolosclerosis was associated with membership in the frontal impairment class. DISCUSSION: Our findings support the use of latent class models that incorporate more comprehensive neuropsychological measures to classify cognitive impairment.


Asunto(s)
Autopsia , Cognición/fisiología , Disfunción Cognitiva/patología , Trastornos de la Memoria/patología , Neuropatología/estadística & datos numéricos , Pruebas Neuropsicológicas/estadística & datos numéricos , Anciano de 80 o más Años , Péptidos beta-Amiloides/fisiología , Encéfalo/patología , Femenino , Humanos , Masculino , Proteínas tau/fisiología
18.
Nat Rev Neurosci ; 14(9): 626-36, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23900411

RESUMEN

Dementia is increasingly being recognized in cases of Parkinson's disease (PD); such cases are termed PD dementia (PDD). The spread of fibrillar α-synuclein (α-syn) pathology from the brainstem to limbic and neocortical structures seems to be the strongest neuropathological correlate of emerging dementia in PD. In addition, up to 50% of patients with PDD also develop sufficient numbers of amyloid-ß plaques and tau-containing neurofibrillary tangles for a secondary diagnosis of Alzheimer's disease, and these pathologies may act synergistically with α-syn pathology to confer a worse prognosis. An understanding of the relationships between these three distinct pathologies and their resultant clinical phenotypes is crucial for the development of effective disease-modifying treatments for PD and PDD.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/fisiología , Enfermedad de Parkinson/patología , alfa-Sinucleína/fisiología , Proteínas tau/fisiología , Enfermedad de Alzheimer/fisiopatología , Encéfalo/patología , Encéfalo/fisiología , Humanos , Neuronas/patología , Neuronas/fisiología , Enfermedad de Parkinson/fisiopatología
19.
Hum Mol Genet ; 24(11): 3058-81, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25687137

RESUMEN

Disarrangement in functions and quality control of mitochondria at synapses are early events in Alzheimer's disease (AD) pathobiology. We reported that a 20-22 kDa NH2-tau fragment mapping between 26 and 230 amino acids of the longest human tau isoform (aka NH2htau): (i) is detectable in cellular and animal AD models, as well in synaptic mitochondria and cerebrospinal fluids (CSF) from human AD subjects; (ii) is neurotoxic in primary hippocampal neurons; (iii) compromises the mitochondrial biology both directly, by inhibiting the ANT-1-dependent ADP/ATP exchange, and indirectly, by impairing their selective autophagic clearance (mitophagy). Here, we show that the extensive Parkin-dependent turnover of mitochondria occurring in NH2htau-expressing post-mitotic neurons plays a pro-death role and that UCHL-1, the cytosolic Ubiquitin-C-terminal hydrolase L1 which directs the physiological remodeling of synapses by controlling ubiquitin homeostasis, critically contributes to mitochondrial and synaptic failure in this in vitro AD model. Pharmacological or genetic suppression of improper mitophagy, either by inhibition of mitochondrial targeting to autophagosomes or by shRNA-mediated silencing of Parkin or UCHL-1 gene expression, restores synaptic and mitochondrial content providing partial but significant protection against the NH2htau-induced neuronal death. Moreover, in mitochondria from human AD synapses, the endogenous NH2htau is stably associated with Parkin and with UCHL-1. Taken together, our studies show a causative link between the excessive mitochondrial turnover and the NH2htau-induced in vitro neuronal death, suggesting that pathogenetic tau truncation may contribute to synaptic deterioration in AD by aberrant recruitment of Parkin and UCHL-1 to mitochondria making them more prone to detrimental autophagic clearance.


Asunto(s)
Enfermedad de Alzheimer/genética , Neuronas/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas tau/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Células HeLa , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Mitocondriales/metabolismo , Mitofagia , Neuronas/fisiología , Transporte de Proteínas , Ratas Wistar , Proteínas tau/fisiología
20.
EMBO J ; 32(22): 2920-37, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24065130

RESUMEN

Mislocalization and aggregation of Aß and Tau combined with loss of synapses and microtubules (MTs) are hallmarks of Alzheimer disease. We exposed mature primary neurons to Aß oligomers and analysed changes in the Tau/MT system. MT breakdown occurs in dendrites invaded by Tau (Tau missorting) and is mediated by spastin, an MT-severing enzyme. Spastin is recruited by MT polyglutamylation, induced by Tau missorting triggered translocalization of TTLL6 (Tubulin-Tyrosine-Ligase-Like-6) into dendrites. Consequences are spine loss and mitochondria and neurofilament mislocalization. Missorted Tau is not axonally derived, as shown by axonal retention of photoconvertible Dendra2-Tau, but newly synthesized. Recovery from Aß insult occurs after Aß oligomers lose their toxicity and requires the kinase MARK (Microtubule-Affinity-Regulating-Kinase). In neurons derived from Tau-knockout mice, MTs and synapses are resistant to Aß toxicity because TTLL6 mislocalization and MT polyglutamylation are prevented; hence no spastin recruitment and no MT breakdown occur, enabling faster recovery. Reintroduction of Tau re-establishes Aß-induced toxicity in TauKO neurons, which requires phosphorylation of Tau's KXGS motifs. Transgenic mice overexpressing Tau show TTLL6 translocalization into dendrites and decreased MT stability. The results provide a rationale for MT stabilization as a therapeutic approach.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Péptidos beta-Amiloides/fisiología , Microtúbulos/fisiología , Péptido Sintasas/fisiología , Sinapsis/patología , Proteínas tau/fisiología , Péptidos beta-Amiloides/química , Animales , Células Cultivadas , Ácido Glutámico/metabolismo , Ratones , Ratones Noqueados , Ratas , Espastina , Proteínas tau/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA