Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Mol Psychiatry ; 27(7): 3010-3023, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35393558

RESUMO

The microtubule-associated protein tau plays a central role in tauopathies such as Alzheimer's disease (AD). The exact molecular mechanisms underlying tau toxicity are unclear, but aging is irrefutably the biggest risk factor. This raises the question of how cellular senescence affects the function of tau as a microtubule regulator. Here we report that the proportion of tau that is proteolytically cleaved at the caspase-3 site (TauC3) doubles in the hippocampus of senescent mice. TauC3 is also elevated in AD patients. Through quantitative live-cell imaging, we show that TauC3 has a drastically reduced dynamics of its microtubule interaction. Single-molecule tracking of tau confirmed that TauC3 has a longer residence time on axonal microtubules. The reduced dynamics of the TauC3-microtubule interaction correlated with a decreased transport of mitochondria, a reduced processivity of APP-vesicle transport and an induction of region-specific dendritic atrophy in CA1 neurons of the hippocampus. The microtubule-targeting drug Epothilone D normalized the interaction of TauC3 with microtubules and modulated the transport of APP-vesicles dependent on the presence of overexpressed human tau. The results indicate a novel toxic gain of function, in which a post-translational modification of tau changes the dynamics of the tau-microtubule interaction and thus leads to axonal transport defects and neuronal degeneration. The data also introduce microtubule-targeting drugs as pharmacological modifiers of the tau-microtubule interaction with the potential to restore the physiological interaction of pathologically altered tau with microtubules.


Assuntos
Doença de Alzheimer , Tauopatias , Doença de Alzheimer/metabolismo , Animais , Transporte Axonal , Caspases/metabolismo , Mutação com Ganho de Função , Humanos , Camundongos , Microtúbulos/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo
2.
J Biol Chem ; 293(21): 8065-8076, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29636414

RESUMO

During neuronal development, the microtubule-associated protein tau becomes enriched in the axon, where it remains concentrated in the healthy brain. In tauopathies such as Alzheimer's disease, tau redistributes from the axon to the somatodendritic compartment. However, the cellular mechanism that regulates tau's localization remains unclear. We report here that tau interacts with the Ca2+-regulated plasma membrane-binding protein annexin A2 (AnxA2) via tau's extreme N terminus encoded by the first exon (E1). Bioinformatics analysis identified two conserved eight-amino-acids-long motifs within E1 in mammals. Using a heterologous yeast system, we found that disease-related mutations and pseudophosphorylation of Tyr-18, located within E1 but outside of the two conserved regions, do not influence tau's interaction with AnxA2. We further observed that tau interacts with the core domain of AnxA2 in a Ca2+-induced open conformation and interacts also with AnxA6. Moreover, lack of E1 moderately increased tau's association rate to microtubules, consistent with the supposition that the presence of the tau-annexin interaction reduces the availability of tau to interact with microtubules. Of note, intracellular competition through overexpression of E1-containing constructs reduced tau's axonal enrichment in primary neurons. Our results suggest that the E1-mediated tau-annexin interaction contributes to the enrichment of tau in the axon and is involved in its redistribution in pathological conditions.


Assuntos
Anexina A2/metabolismo , Anexina A6/metabolismo , Axônios/metabolismo , Microtúbulos/metabolismo , Proteínas tau/metabolismo , Animais , Anexina A2/genética , Anexina A6/genética , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células PC12 , Fosforilação , Ligação Proteica , Ratos , Proteínas tau/genética
3.
Mol Cell Neurosci ; 84: 132-141, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28318914

RESUMO

Tau is a microtubule-associated neuronal protein, whose primary role was long thought to regulate axonal microtubule assembly. Tau is subject to many posttranslational modifications and can aggregate into neurofibrillary tangles, which are considered to be a hallmark of several neurodegenerative diseases collectively called "tauopathies". The most common tauopathy is Alzheimer's disease, where tau pathology correlates with sites of neurodegeneration. Tau belongs to the class of intrinsically disordered proteins, which are known to interact with many partners and are considered to be involved in various signaling, regulation and recognition processes. Thus more recent evidence indicates that tau functionally interacts with many proteins and different cellular structures, which may have an important physiological role and may be involved in neurodegenerative processes. Furthermore, tau can be released from neurons and exert functional effects on other cells. This review article weighs the evidence that tau has subtle but important systemic effects on neuronal network function by maintaining physiological neuronal transmission and synaptic plasticity, which are possibly independent from tau's microtubule modulating activities. Implications for tau-based therapeutic approaches are discussed.


Assuntos
Doença de Alzheimer/terapia , Encéfalo/metabolismo , Emaranhados Neurofibrilares/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Animais , Humanos , Neurônios/metabolismo
4.
J Neurochem ; 143(4): 409-417, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28267200

RESUMO

Alzheimer's disease (AD) is the most common neurodegenerative disorder and is, on a histopathological level, characterized by the presence of extracellular amyloid plaques composed of the protein fragment Aß, and intracellular neurofibrillary tangles, which contain the microtubule-associated protein tau in a hyperphosphorylated state. In AD defects in microtubule (MT) assembly and organization have also been reported; however, it is unclear whether MT abnormalities have a causal and early role in the disease process or represent a common end point downstream of the neurodegenerative cascade. Recent evidence indicates that microtubule-stabilizing drugs prevent axonopathy in animal models of tauopathies and reverse Aß-induced loss of synaptic connectivity in an ex vivo model of amyloidosis. This could suggest that MT dysfunction connects some of the degenerative events and provides a useful target to simultaneously prevent several neurodegenerative processes in AD. Here, we describe how changes in the structure and dynamics of MTs are involved in the different aspects of the neurodegenerative triad of AD. We discuss evidence that MTs are affected both by tau-dependent and tau-independent mechanisms but appear to be regulated in a distinct way in different neuronal compartments. We argue that modulation of MT dynamics could be of potential benefit but needs to be precisely controlled in a cell and compartment-specific manner to avoid harmful side effects. This article is part of the series "Beyond Amyloid".


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Microtúbulos/metabolismo , Microtúbulos/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Dendritos/metabolismo , Dendritos/patologia , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Proteínas tau/metabolismo
5.
J Biol Chem ; 289(24): 16814-25, 2014 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-24755223

RESUMO

The neuronal microtubule-associated protein Tau is expressed in different variants, and changes in Tau isoform composition occur during development and disease. Here, we investigate a potential role of the multivalent tau mRNA-binding proteins G3BP1 and IMP1 in regulating neuronal tau expression. We demonstrate that G3BP1 and IMP1 expression induces the formation of structures, which qualify as neuronal ribonucleoprotein (RNP) granules and concentrate multivalent proteins and mRNA. We show that RNP granule formation leads to a >30-fold increase in the ratio of high molecular weight to low molecular weight tau mRNA and an ∼12-fold increase in high molecular weight to low molecular weight Tau protein. We report that RNP granule formation is associated with increased neurite formation and enhanced process growth. G3BP1 deletion constructs that do not induce granule formation are also deficient in inducing neuronal sprouting or changing the expression pattern of tau. The data indicate that granule formation driven by multivalent proteins modulates tau isoform expression and suggest a morphoregulatory function of RNP granules during health and disease.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas tau/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Processos de Crescimento Celular , DNA Helicases , Deleção de Genes , Humanos , Neurônios/fisiologia , Células PC12 , Proteínas de Ligação a Poli-ADP-Ribose , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Helicases , Proteínas com Motivo de Reconhecimento de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ratos , Ribonucleoproteínas/genética , Proteínas tau/genética
6.
Cytoskeleton (Hoboken) ; 81(1): 47-52, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37694806

RESUMO

Tau is a microtubule-associated protein that is enriched in the axonal process of neurons. Post-translational modifications of tau have been implicated in the development of tauopathies characterized by defects in axonal transport, neuronal atrophy, and microtubule disassembly. Although tau is almost quantitatively bound to microtubules under physiological conditions, it does not significantly affect axonal transport. Furthermore, acute or chronic tau deficiency does not result in significant destabilization of neuronal microtubules, challenging the classical view that disease-related tau modifications directly cause axonal microtubule collapse. Here, we discuss how the rapid interaction kinetics of the tau-microtubule interaction, which we previously termed the kiss-and-hop interaction, explains why tau does not affect microtubule-dependent axonal transport but still allows tau to modulate microtubule polymerization. In contrast, tau modifications that slow down the kinetics of the tau-microtubule interaction and increase the residence time of tau at a microtubule interaction site can disrupt axonal transport and cause dendritic atrophy. We discuss the consequences of such a gain-of-toxicity mechanism in terms of the development of disease-modulating drugs that target the tau protein.


Assuntos
Tauopatias , Proteínas tau , Humanos , Transporte Axonal , Microtúbulos/metabolismo , Tauopatias/metabolismo , Atrofia/metabolismo
7.
Nat Commun ; 15(1): 1679, 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38396035

RESUMO

Tauopathies such as Alzheimer's disease are characterized by aggregation and increased phosphorylation of the microtubule-associated protein tau. Tau's pathological changes are closely linked to neurodegeneration, making tau a prime candidate for intervention. We developed an approach to monitor pathological changes of aggregation-prone human tau in living neurons. We identified 2-phenyloxazole (PHOX) derivatives as putative polypharmacological small molecules that interact with tau and modulate tau kinases. We found that PHOX15 inhibits tau aggregation, restores tau's physiological microtubule interaction, and reduces tau phosphorylation at disease-relevant sites. Molecular dynamics simulations highlight cryptic channel-like pockets crossing tau protofilaments and suggest that PHOX15 binding reduces the protofilament's ability to adopt a PHF-like conformation by modifying a key glycine triad. Our data demonstrate that live-cell imaging of a tauopathy model enables screening of compounds that modulate tau-microtubule interaction and allows identification of a promising polypharmacological drug candidate that simultaneously inhibits tau aggregation and reduces tau phosphorylation.


Assuntos
Doença de Alzheimer , Tauopatias , Humanos , Tauopatias/tratamento farmacológico , Tauopatias/metabolismo , Proteínas tau/metabolismo , Microtúbulos/metabolismo , Doença de Alzheimer/metabolismo , Citoesqueleto/metabolismo , Fosforilação
8.
Brain Res Bull ; 194: 13-22, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36626968

RESUMO

The unique morphology of neurons consists of a long axon and a highly variable arbour of dendritic processes, which assort neuronal cells into the main classes. The dendritic tree serves as the main domain for receiving synaptic input. Therefore, to maintain the structure and to be able to plastically change according to the incoming stimuli, molecules and organelles need to be readily available. This is achieved mainly via bi-directional transport of cargo along the microtubule lattices. Analysis of dendritic transport is lagging behind the investigation of axonal transport. Moreover, addressing transport mechanisms in tissue environment is very challenging and, therefore, rare. We employed high-speed volumetric lattice light-sheet microscopy and single particle tracking of truncated KIF1A motor protein lacking the cargo-binding domain. We focused our analysis on dendritic processes of CA1 pyramidal neurons in cultured hippocampal tissue. Analysis of individual trajectories revealed detailed information about stalling and high variability in movement and speed, and biased directionality of KIF1A. Furthermore, we could also observe KIF1A shortly entering into dendritic spines. We provide a workflow to analyse variations in the speed and direction of motor protein movement in dendrites that are either intrinsic properties of the motor domain or depend on the structure and modification of the microtubule trails.


Assuntos
Espinhas Dendríticas , Cinesinas , Microscopia , Camundongos , Axônios/metabolismo , Dendritos , Espinhas Dendríticas/metabolismo , Hipocampo/metabolismo , Cinesinas/metabolismo , Cinesinas/fisiologia , Microscopia/métodos , Neurônios/metabolismo , Animais
9.
Mol Genet Genomics ; 287(4): 313-24, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22327349

RESUMO

Today, a large variety of viral vectors is available for ectopic gene expression in mammalian cell cultures or in vivo. Among them, infection with Sindbis virus- or Lentivirus-derived constructs is often used to address biological questions or for applications in neuronal therapies. However, cloning of genes of interest is time consuming, since it relies on restriction and ligation, frequently of PCR-generated DNA fragments with suitable restriction sites introduced by the primers employed. We here take advantage of the unusually high capacity for homologous recombination in Saccharomyces cerevisiae to circumvent this problem, and introduce a new set of triple shuttle vectors, which can be shuffled between E. coli, yeast, and mammalian cells. The system allows the introduction of genes of interest largely independent of the target site in the vectors. It also allows the removal of the yeast selection marker by Cre-recombinase directed recombination in E. coli, if vector size limits transfection efficiency in the mammalian cells. We demonstrate the expression of genes encoding fluorescent proteins (EGFP and mCherry) both separately and in combination, using two different viral systems in mammalian cell lines, primary neurons and organotypic slices.


Assuntos
Vetores Genéticos , Lentivirus/genética , Neurônios/metabolismo , Sindbis virus/genética , Animais , Escherichia coli/genética , Proteínas de Fluorescência Verde/genética , Camundongos , Saccharomyces cerevisiae/genética
10.
Curr Neuropharmacol ; 20(4): 782-798, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34852744

RESUMO

The microtubule skeleton plays an essential role in nerve cells as the most important structural determinant of morphology and as a highway for axonal transport processes. Many neurodegenerative diseases are characterized by changes in the structure and organization of microtubules and microtubule-regulating proteins such as the microtubule-associated protein tau, which exhibits characteristic changes in a whole class of diseases collectively referred to as tauopathies. Changes in the dynamics of microtubules appear to occur early under neurodegenerative conditions and are also likely to contribute to age-related dysfunction of neurons. Thus, modulating microtubule dynamics and correcting impaired microtubule stability can be a useful neuroprotective strategy to counteract the disruption of the microtubule system in disease and aging. In this article, we review current microtubule- directed approaches for the treatment of neurodegenerative diseases with microtubules as a drug target, tau as a drug target, and post-translational modifications as potential modifiers of the microtubule system. We discuss limitations of the approaches that can be traced back to the rather unspecific mechanism of action, which causes undesirable side effects in non-neuronal cell types or which are due to the disruption of non-microtubule-related interactions. We also develop some thoughts on how the specificity of the approaches can be improved and what further targets could be used for modulating substances.


Assuntos
Doenças Neurodegenerativas , Tauopatias , Humanos , Microtúbulos/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Processamento de Proteína Pós-Traducional , Tauopatias/tratamento farmacológico , Tauopatias/metabolismo
11.
Brain Res Bull ; 190: 234-243, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36244582

RESUMO

Microtubules are essential for the development of neurons and the regulation of their structural plasticity. Microtubules also provide the structural basis for the long-distance transport of cargo. Various factors influence the organization and dynamics of neuronal microtubules, and disturbance of microtubule regulation is thought to play a central role in neurodegenerative diseases. However, imaging and quantitative assessment of the microtubule organization in the densely packed neuronal processes is challenging. The development of super-resolution techniques combined with the use of nanobodies offers new possibilities to visualize microtubules in neurites in high resolution. In combination with recently developed computational analysis tools, this allows automated quantification of neuronal microtubule organization with high precision. Here we have implemented three-dimensional DNA-PAINT (Point Accumulation in Nanoscale Topography), a single-molecule localization microscopy (SMLM) technique, which allows us to acquire 3D arrays of the microtubule lattice in axons of model neurons (neuronally differentiated PC12 cells) and dendrites of primary neurons. For the quantitative analysis of the microtubule organization, we used the open-source software package SMLM image filament extractor (SIFNE). We found that treatment with nanomolar concentrations of the microtubule-targeting drug epothilone D (EpoD) increased microtubule density in axon-like processes of model neurons and shifted the microtubule length distribution to shorter ones, with a mean microtubule length of 2.39 µm (without EpoD) and 1.98 µm (with EpoD). We also observed a significant decrease in microtubule straightness after EpoD treatment. The changes in microtubule density were consistent with live-cell imaging measurements of ensemble microtubule dynamics using a previously established Fluorescence Decay After Photoactivation (FDAP) assay. For comparison, we determined the organization of the microtubule array in dendrites of primary hippocampal neurons. We observed that dendritic microtubules have a very similar length distribution and straightness compared to microtubules in axon-like processes of a neuronal cell line. Our data show that super-resolution imaging of microtubules followed by algorithm-based image analysis represents a powerful tool to quantitatively assess changes in microtubule organization in neuronal processes, useful to determine the effect of microtubule-modulating conditions. We also provide evidence that the approach is robust and can be applied to neuronal cell lines or primary neurons, both after incorporation of labeled tubulin and by anti-tubulin antibody staining.


Assuntos
Axônios , Microtúbulos , Ratos , Animais , Microtúbulos/metabolismo , Axônios/metabolismo , Neurônios/metabolismo , Células PC12
12.
Front Neurol ; 11: 590059, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193056

RESUMO

Tau protein (MAPT) is classified as a microtubule-associated protein (MAP) and is believed to regulate the axonal microtubule arrangement. It belongs to the tau/MAP2/MAP4 family of MAPs that have a similar microtubule binding region at their carboxy-terminal half. In tauopathies, such as Alzheimer's disease, tau is distributed more in the somatodendritic compartment, where it aggregates into filamentous structures, the formation of which correlates with cognitive impairments in patients. While microtubules are the dominant interaction partners of tau under physiological conditions, tau has many additional interaction partners that can contribute to its physiological and pathological role. In particular, the amino-terminal non-microtubule binding domain (N-terminal projection region, NTR) of tau interacts with many partners that are involved in membrane organization. The NTR contains intrinsically disordered regions (IDRs) that show a strong evolutionary increase in the disorder and may have been the basis for the development of new, tau-specific interactions. In this review we discuss the functional organization of the tau protein and the special features of the tau non-microtubule binding region also in the connection with the results of Tau KO models. We consider possible physiological and pathological functions of tau's non-microtubule interactions, which could indicate that interactions mediated by tau's NTR and regulated by far-reaching functional interactions of the PRR and the extreme C-terminus of tau contribute to the pathological processes.

13.
Artigo em Inglês | MEDLINE | ID: mdl-32390822

RESUMO

Alzheimer's disease is regarded as a synaptopathy with a long presymptomatic phase. Soluble, oligomeric amyloid-ß (Aß) is thought to play a causative role in this disease, which eventually leads to cognitive decline. However, most animal studies have employed mice expressing high levels of the Aß precursor protein (APP) transgene to drive pathology. Here, to understand how the principal neurons in different brain regions cope with moderate, chronically present levels of Aß, we employed transgenic mice expressing equal levels of mouse and human APP carrying a combination of three familial AD (FAD)-linked mutations (Swedish, Dutch, and London), that develop plaques only in old age. We analyzed dendritic spine parameters in hippocampal and cortical brain regions after targeted expression of EGFP to allow high-resolution imaging, followed by algorithm-based evaluation of mice of both sexes from adolescence to old age. We report that Aß species gradually accumulated throughout the life of APPSDL mice, but not the oligomeric forms, and that the amount of membrane-associated oligomers decreased at the onset of plaque formation. We observed an age-dependent loss of thin spines under most conditions as an indicator of a loss of synaptic plasticity in older mice. We further found that hippocampal pyramidal neurons respond to increased Aß levels by lowering spine density and shifting spine morphology, which reached significance in the CA1 subfield. In contrast, the spine density in cortical pyramidal neurons of APPSDL mice was unchanged. We also observed an increase in the protein levels of PSD-95 and Arc in the hippocampus and cortex, respectively. Our data demonstrated that increased concentrations of Aß have diverse effects on dendritic spines in the brain and suggest that hippocampal and cortical neurons have different adaptive and compensatory capacity during their lifetime. Our data also indicated that spine morphology differs between sexes in a region-specific manner.

14.
Artigo em Inglês | MEDLINE | ID: mdl-32116638

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder that leads to impaired memory and cognitive deficits. Spine loss as well as changes in spine morphology correlates with cognitive impairment in this neurological disorder. Many studies in animal models and ex vivo cultures indicate that amyloid ß-peptide (Aß) oligomers induce synaptic damage early during the progression of the disease. Here, in order to determine the events that initiate synaptic alterations, we acutely applied oligomeric Aß to primary hippocampal neurons and an ex vivo model of organotypic hippocampal cultures from a mouse after targeted expression of EGFP to allow high-resolution imaging and algorithm-based evaluation of spine changes. Dendritic spines were classified as thin, stubby or mushroom, based on morphology. In vivo, time-lapse imaging showed that the three spine types were relatively stable, although their stability significantly decreased after treatment with Aß oligomers. Unexpectedly, we observed that the density of total dendritic spines increased in organotypic hippocampal slices treated with Aß compared to control cultures. Specifically, the fraction of stubby spines significantly increased, while mushroom and thin spines remained unaltered. Pharmacological tools revealed that acute Aß oligomers induced spine changes through mechanisms involving CaMKII and integrin ß1 activities. Additionally, analysis of dendritic complexity based on a 3D reconstruction of the whole neuron morphology showed an increase in the apical dendrite length and branching points in CA1 organotypic hippocampal slices treated with Aß. In contrast to spines, the morphological changes were affected by integrin ß1 but not by CaMKII inhibition. Altogether, these data indicate that the Aß oligomers exhibit early dual effects by acutely enhancing dendritic complexity and spine density.

15.
Neurochem Int ; 54(1): 28-36, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18984021

RESUMO

The binding of two radiolabelled analogues (N-(5-[125I]Iodo-2-phenoxyphenyl)-N-(2,5-dimethoxybenzyl)acetamide ([125I]desfluoro-DAA1106) and N-(5-[125I]Fluoro-2-phenoxyphenyl)-N-(2-[125I]Iodo-5-methoxybenzyl)acetamide ([125I]desmethoxy-DAA1106) of the peripheral benzodiazepine receptor (PBR) (or TSPO, 18kDa translocator protein) ligand DAA1106 was examined by in vitro autoradiography on human post mortem whole hemisphere brain slices obtained from Alzheimer's disease (AD) patients and age-matched controls. Both [(125)I]desfluoro-IDAA1106 and [(125)I]desmethoxy-IDAA1106 were effectively binding to various brain structures. The binding could be blocked by the unlabelled ligand as well as by other PBR specific ligands. With both radiolabelled compounds, the binding showed regional inhomogeneity and the specific binding values proved to be the highest in the hippocampus, temporal and parietal cortex, the basal ganglia and thalamus in the AD brains. Compared with age-matched control brains, specific binding in several brain structures (temporal and parietal lobes, thalamus and white matter) in Alzheimer brains was significantly higher, indicating that the radioligands can effectively label-activated microglia and the up-regulated PBR/TSPO system in AD. Complementary immunohistochemical studies demonstrated reactive microglia activation in the AD brain tissue and indicated that increased ligand binding coincides with increased regional microglia activation due to neuroinflammation. These investigations yield further support to the PBR/TSPO binding capacity of DAA1106 in human brain tissue, demonstrate the effective usefulness of its radio-iodinated analogues as imaging biomarkers in post mortem human studies, and indicate that its radiolabelled analogues, labelled with short half-time bioisotopes, can serve as prospective in vivo imaging biomarkers of activated microglia and the up-regulated PBR/TSPO system in the human brain.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Receptores de GABA-A/metabolismo , Acetamidas/metabolismo , Idoso de 80 Anos ou mais , Autorradiografia , Feminino , Humanos , Imuno-Histoquímica , Radioisótopos do Iodo , Cinética , Masculino , Pessoa de Meia-Idade , Éteres Fenílicos/metabolismo , Mudanças Depois da Morte , Valores de Referência
16.
Front Aging Neurosci ; 11: 256, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31619983

RESUMO

Tau is a neuronal microtubule-associated protein (MAP) that is involved in the regulation of axonal microtubule assembly. However, as a protein with intrinsically disordered regions (IDRs), tau also interacts with many other partners in addition to microtubules. Phosphorylation at selected sites modulates tau's various intracellular interactions and regulates the properties of IDRs. In Alzheimer's disease (AD) and other tauopathies, tau exhibits pathologically increased phosphorylation (hyperphosphorylation) at selected sites and aggregates into neurofibrillary tangles (NFTs). By bioinformatics means, we tested the hypothesis that the sequence of tau has changed during the vertebrate evolution in a way that novel interactions developed and also the phosphorylation pattern was affected, which made tau prone to the development of tauopathies. We report that distinct regions of tau show functional specialization in their molecular interactions. We found that tau's amino-terminal region, which is involved in biological processes related to "membrane organization" and "regulation of apoptosis," exhibited a strong evolutionary increase in protein disorder providing the basis for the development of novel interactions. We observed that the predicted phosphorylation sites have changed during evolution in a region-specific manner, and in some cases the overall number of phosphorylation sites increased owing to the formation of clusters of phosphorylatable residues. In contrast, disease-specific hyperphosphorylated sites remained highly conserved. The data indicate that novel, non-microtubule related tau interactions developed during evolution and suggest that the biological processes, which are mediated by these interactions, are of pathological relevance. Furthermore, the data indicate that predicted phosphorylation sites in some regions of tau, including a cluster of phosphorylatable residues in the alternatively spliced exon 2, have changed during evolution. In view of the "antagonistic pleiotropy hypothesis" it may be worth to take disease-associated phosphosites with low evolutionary conservation as relevant biomarkers into consideration.

17.
Transl Psychiatry ; 9(1): 7, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30664629

RESUMO

In humans, genetic variants of DLGAP1-4 have been linked with neuropsychiatric conditions, including autism spectrum disorder (ASD). While these findings implicate the encoded postsynaptic proteins, SAPAP1-4, in the etiology of neuropsychiatric conditions, underlying neurobiological mechanisms are unknown. To assess the contribution of SAPAP4 to these disorders, we characterized SAPAP4-deficient mice. Our study reveals that the loss of SAPAP4 triggers profound behavioural abnormalities, including cognitive deficits combined with impaired vocal communication and social interaction, phenotypes reminiscent of ASD in humans. These behavioural alterations of SAPAP4-deficient mice are associated with dramatic changes in synapse morphology, function and plasticity, indicating that SAPAP4 is critical for the development of functional neuronal networks and that mutations in the corresponding human gene, DLGAP4, may cause deficits in social and cognitive functioning relevant to ASD-like neurodevelopmental disorders.


Assuntos
Transtorno do Espectro Autista/genética , Disfunção Cognitiva/genética , Proteínas do Tecido Nervoso/genética , Proteínas Associadas SAP90-PSD95/genética , Animais , Comportamento Animal , Modelos Animais de Doenças , Feminino , Relações Interpessoais , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Comportamento Social , Sinapses/metabolismo
18.
Neurochem Int ; 52(1-2): 265-71, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17664023

RESUMO

The cutaneous and mucosal surfaces in the infraorbital region around the whisker pad are innervated by the maxillary division of the afferent fibers of the trigeminal nerve, while certain ganglion cells project to the principal sensory trigeminal nucleus (Pr5). In turn, some of the neurons in the Pr5 project to the motor trigeminal nucleus (Mo5), whose neurons do not innervate the infraorbital skin. We analyzed the calmodulin (CaM) gene expression in these nuclei after dithranol-induced inflammation and subsequent treatment with corticosteroid in the infraorbital skin. CaM gene-specific mRNA populations were detected through quantitative image analysis of the distribution of CaM gene-specific riboprobes in brain stem cryostat sections of control rats and rats chronically treated with dithranol, corticosteroid or both. These nuclei displayed a differentially altered CaM gene expression in response to the treatments. While the CaM I and II mRNA contents were increased, the CaM III transcripts remained unaltered after chronic dithranol treatment in the Mo5. In the Pr5, however, the CaM mRNA contents were either unchanged (CaM I and III) or increased (CaM II). Subsequent corticosteroid treatment reversed the stimulatory effects of dithranol on the expression of all the CaM genes in the Mo5, but was without significant effects on the CaM I and II genes, or even increased the CaM III mRNA contents in the Pr5. Corticosteroid treatment alone was either ineffective or decreased the levels of CaM mRNAs in these nuclei. These data suggest that peripheral noxae of dermal origin may result in a trans-synaptically acting differential regulation of the multiple CaM genes in the brain.


Assuntos
Corticosteroides/uso terapêutico , Calmodulina/genética , Regulação da Expressão Gênica , Inflamação/genética , Sinapses/metabolismo , Nervo Trigêmeo/patologia , Animais , Encéfalo/metabolismo , Face , Hibridização In Situ , Inflamação/tratamento farmacológico , Masculino , Boca , Ratos , Ratos Sprague-Dawley
19.
Neuropeptides ; 42(1): 57-67, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18068762

RESUMO

[(3)H]Tyr-Tic-(2S,3R)-beta-MePhe-Phe-OH (where Tic: 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid) with a specific radioactivity of 53.7 Ci/mmol was synthesized and characterized in receptor binding assays at 25 degrees C in rat brain membranes. The specific binding was saturable and displayed high affinity, with a K(D) of 0.16+/-0.005 nM and B(max) of 85.9+/-6.3 fmol/mg protein. NaCl increased its affinity by about 4-fold in membranes of rat brain and Chinese Hamster Ovary Cells stably transfected with the human delta-opioid receptors (hDOR-CHO) showing that the new ligand is an antagonist. The prototypic delta-opioid ligands were much more potent than mu- or kappa-specific ligands in competition assays. The autoradiographic distribution of the binding sites of the new ligand agreed with the known locations of the delta-opioid receptors in rat brain. The unlabeled new ligand was about 7-fold more potent than the parent peptide in competing for the binding sites of [(3)H]Tyr-Tic-(2S,3R)-beta-MePhe-Phe-OH in rat brain membranes. Likewise, the threo-beta-methyl analog was 3.8-fold more potent than the parent compound in antagonizing the effect of DPDPE in the [(35)S]GTPgammaS functional assay in hDOR-CHO membranes. The new, highly potent, conformationally constrained antagonist may be a valuable pharmacological tool in understanding the structural and topographical requirements of peptide ligand binding to the delta-opioid receptors.


Assuntos
Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacologia , Receptores Opioides delta/antagonistas & inibidores , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Autorradiografia , Sítios de Ligação , Química Encefálica/efeitos dos fármacos , Células CHO , Cricetinae , Cricetulus , D-Penicilina (2,5)-Encefalina/antagonistas & inibidores , D-Penicilina (2,5)-Encefalina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Humanos , Masculino , Ensaio Radioligante , Ratos , Ratos Wistar
20.
Neuropharmacology ; 113(Pt A): 434-444, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27773645

RESUMO

DMSO has a widespread use as a vehicle for water-insoluble therapeutic drug candidates but may also exert disease-relevant pharmacological effects by itself. However, its influence on the CNS has hardly been addressed. Here we examined the brain structure and function following chronic exposure to low DMSO dose at a paradigm with flawed synaptic connectivity in a preclinical transgenic mouse model for Alzheimer's disease (APPSDL mice). DMSO treatment increased spine density in a region-specific manner in the hippocampus of APPSDL mice ex vivo and in vivo. Moreover, DMSO exhibited clear influence on the behavior of this mouse line by enhancing hippocampal-dependent spatial memory accuracy, modulating hippocampal-independent olfactory habituation and displaying anxiolytic effect. Despite that most of the action of DMSO was observed in animals with elevated Aß levels, the drug did not exert its function via decreasing the oligomeric Aß species. However, challenging organotypic hippocampal slice cultures with NMDA receptor antagonist MK-801 recapitulated the effect of DMSO on spine density, indicating a tuning influence of DMSO on receptor signalization. Our findings demonstrate that DMSO should be considered as a true bioactive compound, which has the potential to be a beneficial adjuvant to counteract Aß-mediated synaptotoxicity and behavioral impairment.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Espinhas Dendríticas/efeitos dos fármacos , Dimetil Sulfóxido/uso terapêutico , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Espinhas Dendríticas/patologia , Dimetil Sulfóxido/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Sequestradores de Radicais Livres/farmacologia , Sequestradores de Radicais Livres/uso terapêutico , Hipocampo/patologia , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA