Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Neurochem ; 157(3): 684-694, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33251603

RESUMO

Various lines of evidence implicate oxidative stress in the pathogenic mechanism(s) underpinning tauopathies. Consequently, antioxidant therapies have been considered in clinical practice for the treatment of tauopathies such as Alzheimer's disease (AD), but with mixed results. We and others have previously reported increased protein oxidation upon expression of both human 0N3R (hTau0N3R ) and 0N4R (hTau0N4R ) tau in vivo. Building on these studies, we demonstrate here the suppression of hTau0N3R associated phenotypes in Drosophila melanogaster after treatment with vitamin C or vitamin E. Curiously the rescue of phenotype was seen without alteration in total tau level or alteration in phosphorylation at a number of disease-associated sites. Moreover, treatment with paraquat, a pro-oxidant drug, did not exacerbate the hTau0N3R phenotypes. This result following paraquat treatment is reminiscent of our previous findings with hTau0N4R which also causes greater oxidative stress when compared to hTau0N3R but has a milder phenotype. Collectively our data imply that the role of oxidative stress in tau-mediated toxicity is not straight forward and there may be isoform-specific effects as well as contribution of other factors. This may explain the ambiguous effects of anti-oxidant treatments on clinical outcome in dementia patients.


Assuntos
Antioxidantes/farmacologia , Vitamina E/farmacologia , Proteínas tau/antagonistas & inibidores , Proteínas tau/toxicidade , Animais , Ácido Ascórbico/farmacologia , Drosophila melanogaster , Larva , Locomoção/efeitos dos fármacos , Masculino , Oxidantes/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Paraquat/toxicidade , Fosforilação/efeitos dos fármacos
2.
Neurobiol Dis ; 105: 74-83, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28502805

RESUMO

Tau exists as six closely related protein isoforms in the adult human brain. These are generated from alternative splicing of a single mRNA transcript and they differ in the absence or presence of two N-terminal and three or four microtubule binding domains. Typically all six isoforms have been considered functionally similar. However, their differential involvement in particular tauopathies raises the possibility that there may be isoform-specific differences in physiological function and pathological role. To explore this, we have compared the phenotypes induced by the 0N3R and 0N4R isoforms in Drosophila. Expression of the 3R isoform causes more profound axonal transport defects and locomotor impairments, culminating in a shorter lifespan than the 4R isoform. In contrast, the 4R isoform leads to greater neurodegeneration and impairments in learning and memory. Furthermore, the phosphorylation patterns of the two isoforms are distinct, as is their ability to induce oxidative stress. These differences are not consequent to different expression levels and are suggestive of bona fide physiological differences in isoform biology and pathological potential. They may therefore explain isoform-specific mechanisms of tau-toxicity and the differential susceptibility of brain regions to different tauopathies.


Assuntos
Modelos Animais de Doenças , Sequências de Repetição em Tandem/genética , Tauopatias/genética , Tauopatias/fisiopatologia , Proteínas tau/genética , Fatores Etários , Animais , Animais Geneticamente Modificados , Transporte Axonal , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Feminino , Humanos , Larva/genética , Aprendizagem/fisiologia , Locomoção/genética , Masculino , Memória/fisiologia , Fenótipo , Fosforilação , Isoformas de Proteínas/metabolismo , Tauopatias/mortalidade , Tauopatias/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vias Visuais/metabolismo , Proteínas tau/metabolismo
3.
Neural Regen Res ; 19(9): 1899-1907, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38227514

RESUMO

Aging is the leading risk factor for Alzheimer's disease and other neurodegenerative diseases. We now understand that a breakdown in the neuronal cytoskeleton, mainly underpinned by protein modifications leading to the destabilization of microtubules, is central to the pathogenesis of Alzheimer's disease. This is accompanied by morphological defects across the somatodendritic compartment, axon, and synapse. However, knowledge of what occurs to the microtubule cytoskeleton and morphology of the neuron during physiological aging is comparatively poor. Several recent studies have suggested that there is an age-related increase in the phosphorylation of the key microtubule stabilizing protein tau, a modification, which is known to destabilize the cytoskeleton in Alzheimer's disease. This indicates that the cytoskeleton and potentially other neuronal structures reliant on the cytoskeleton become functionally compromised during normal physiological aging. The current literature shows age-related reductions in synaptic spine density and shifts in synaptic spine conformation which might explain age-related synaptic functional deficits. However, knowledge of what occurs to the microtubular and actin cytoskeleton, with increasing age is extremely limited. When considering the somatodendritic compartment, a regression in dendrites and loss of dendritic length and volume is reported whilst a reduction in soma volume/size is often seen. However, research into cytoskeletal change is limited to a handful of studies demonstrating reductions in and mislocalizations of microtubule-associated proteins with just one study directly exploring the integrity of the microtubules. In the axon, an increase in axonal diameter and age-related appearance of swellings is reported but like the dendrites, just one study investigates the microtubules directly with others reporting loss or mislocalization of microtubule-associated proteins. Though these are the general trends reported, there are clear disparities between model organisms and brain regions that are worthy of further investigation. Additionally, longitudinal studies of neuronal/cytoskeletal aging should also investigate whether these age-related changes contribute not just to vulnerability to disease but also to the decline in nervous system function and behavioral output that all organisms experience. This will highlight the utility, if any, of cytoskeletal fortification for the promotion of healthy neuronal aging and potential protection against age-related neurodegenerative disease. This review seeks to summarize what is currently known about the physiological aging of the neuron and microtubular cytoskeleton in the hope of uncovering mechanisms underpinning age-related risk to disease.

4.
Acta Neuropathol Commun ; 12(1): 52, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38576010

RESUMO

The transcellular propagation of the aberrantly modified protein tau along the functional brain network is a key hallmark of Alzheimer's disease and related tauopathies. Inoculation-based tau propagation models can recapitulate the stereotypical spread of tau and reproduce various types of tau inclusions linked to specific tauopathy, albeit with varying degrees of fidelity. With this systematic review, we underscore the significance of judicious selection and meticulous functional, biochemical, and biophysical characterization of various tau inocula. Furthermore, we highlight the necessity of choosing suitable animal models and inoculation sites, along with the critical need for validation of fibrillary pathology using confirmatory staining, to accurately recapitulate disease-specific inclusions. As a practical guide, we put forth a framework for establishing a benchmark of inoculation-based tau propagation models that holds promise for use in preclinical testing of disease-modifying drugs.


Assuntos
Doença de Alzheimer , Tauopatias , Animais , Doença de Alzheimer/patologia , Emaranhados Neurofibrilares/patologia , Modelos Animais de Doenças , Tauopatias/patologia , Proteínas tau/metabolismo , Encéfalo/patologia
5.
Brain Commun ; 5(2): fcad052, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37013175

RESUMO

Tauopathy is characterized by neuronal dysfunction and degeneration occurring as a result of changes to the microtubule-associated protein tau. The neuronal changes evident in tauopathy bear striking morphological resemblance to those reported in models of Wallerian degeneration. The mechanisms underpinning Wallerian degeneration are not fully understood although it can be delayed by the expression of the slow Wallerian degeneration (WldS) protein, which has also been demonstrated to delay axonal degeneration in some models of neurodegenerative disease. Given the morphological similarities between tauopathy and Wallerian degeneration, this study investigated whether tau-mediated phenotypes can be modulated by co-expression of WldS. In a Drosophila model of tauopathy in which expression of human 0N3R tau protein leads to progressive age-dependent phenotypes, WldS was expressed with and without activation of the downstream pathway. The olfactory receptor neuron circuit OR47b was used for these studies in adults, and the larval motor neuron system was employed in larvae. Tau phenotypes studied included neurodegeneration, axonal transport, synaptic deficits and locomotor behaviour. Impact on total tau was ascertained by assessing total, phosphorylated and misfolded tau levels by immunohistochemistry. Activation of the pathway downstream of WldS completely suppressed tau-mediated degeneration. This protective effect was evident even if the pathway downstream of WldS was activated several weeks after tau-mediated degeneration had become established. Though total tau levels were not altered, the protected neurons displayed significantly reduced MC1 immunoreactivity suggestive of clearance of misfolded tau, as well as a trend for a decline in tau species phosphorylated at the AT8 and PHF1 epitopes. In contrast, WldS expression without activation of the downstream protective pathway did not rescue tau-mediated degeneration in adults or improve tau-mediated neuronal dysfunction including deficits in axonal transport, synaptic alterations and locomotor behaviour in tau-expressing larvae. This collectively implies that the pathway mediating the protective effect of WldS intersects with the mechanism(s) of degeneration initiated by tau and can effectively halt tau-mediated degeneration at both early and late stages. Understanding the mechanisms underpinning this protection could identify much-needed disease-modifying targets for tauopathies.

6.
PLoS One ; 18(1): e0262792, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36701399

RESUMO

Tau becomes abnormally hyper-phosphorylated and aggregated in tauopathies like Alzheimers disease (AD). As age is the greatest risk factor for developing AD, it is important to understand how tau protein itself, and the pathways implicated in its turnover, change during aging. We investigated age-related changes in total and phosphorylated tau in brain samples from two cohorts of cognitively normal individuals spanning 19-74 years, without overt neurodegeneration. One cohort utilised resected tissue and the other used post-mortem tissue. Total soluble tau levels declined with age in both cohorts. Phosphorylated tau was undetectable in the post-mortem tissue but was clearly evident in the resected tissue and did not undergo significant age-related change. To ascertain if the decline in soluble tau was correlated with age-related changes in autophagy, three markers of autophagy were tested but only two appeared to increase with age and the third was unchanged. This implies that in individuals who do not develop neurodegeneration, there is an age-related reduction in soluble tau which could potentially be due to age-related changes in autophagy. Thus, to explore how an age-related increase in autophagy might influence tau-mediated dysfunctions in vivo, autophagy was enhanced in a Drosophila model and all age-related tau phenotypes were significantly ameliorated. These data shed light on age-related physiological changes in proteins implicated in AD and highlights the need to study pathways that may be responsible for these changes. It also demonstrates the therapeutic potential of interventions that upregulate turnover of aggregate-prone proteins during aging.


Assuntos
Doença de Alzheimer , Tauopatias , Animais , Humanos , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Idoso , Proteínas tau/metabolismo , Tauopatias/metabolismo , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Drosophila/metabolismo , Autofagia/genética , Fosforilação
7.
Biochem Soc Trans ; 40(4): 641-3, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22817708

RESUMO

It is an exciting time for tau researchers as it is now generally accepted that abnormal tau species are required to mediate the toxic effects of amyloid ß-peptide oligomers in Alzheimer's disease. Tau may play multiple roles in neurophysiology and there may be further pathologically relevant tau alterations, besides hyperphosphorylation and aggregation. The recent Biology and Pathology of Tau and its Role in Tauopathies II meeting explored these various aspects of tau, and presentations at the meeting, described in the following articles in this issue of Biochemical Society Transactions, are outlined in the present paper.


Assuntos
Tauopatias/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Fosforilação
8.
Biochem Soc Trans ; 40(4): 693-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22817718

RESUMO

Insoluble aggregates of the microtubule-associated protein tau characterize a number of neurodegenerative diseases collectively termed tauopathies. These aggregates comprise abnormally hyperphosphorylated and misfolded tau proteins. Research in this field has traditionally focused on understanding how hyperphosphorylated and aggregated tau mediates dysfunction and toxicity in tauopathies. Recent findings from both Drosophila and rodent models of tauopathy suggest that large insoluble aggregates such as tau filaments and tangles may not be the key toxic species in these diseases. Thus some investigators have shifted their focus to study pre-filament tau species such as tau oligomers and hyperphosphorylated tau monomers. Interestingly, tau oligomers can exist in a variety of states including hyperphosphorylated and unphosphorylated forms, which can be both soluble and insoluble. It remains to be determined which of these oligomeric states of tau are causally involved in neurodegeneration and which signal the beginning of the formation of inert/protective filaments. It will be important to better understand this so that tau-based therapeutic interventions can target the most toxic tau species.


Assuntos
Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Animais , Humanos , Fosforilação , Tauopatias/metabolismo
9.
Front Pharmacol ; 13: 903119, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35662729

RESUMO

Global forecasts for prevalence of Alzheimer's Disease (AD) estimate that 152.8 million people will have dementia in 2050, a sharp rise from 57.4 million in 2019 (GBD 2019). This rise can be attributable to increases in population growth and aging, but in the absence of disease-modifying therapies it poses a huge societal challenge that must be addressed urgently. One way to combat this challenge is to explore the utility of holistic treatments that may protect against AD, including traditional herbs, spices and other nutraceuticals that are pharmacologically safe, inexpensive and readily available. In this light, the spice turmeric, and its active ingredient curcumin, has been investigated as a potential holistic treatment for AD over the past 2 decades; however, promising results with animal studies have not translated to success in clinical trials. One issue is that most animal models examining the effects of curcumin and curcumin derivatives in AD have been done with a focus at ameliorating amyloid pathology. Due to the limited success of Amyloid-ß-based drugs in recent clinical trials, tau-focused therapeutics provide a promising alternative. In this article, we aim to provide a clearer picture of what is currently known about the effectiveness of curcumin and curcumin derivatives to ameliorate tau pathology. Tau focused studies may help inform more successful clinical studies by placing greater emphasis on the development and optimised delivery of curcumin derivatives that more effectively target tau pathology.

10.
Biochem Soc Trans ; 38(4): 953-4, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20658983

RESUMO

Tauopathies are a clinically diverse group of neurodegenerative dementias involving perturbations of the level or phosphorylation state of the microtubule-binding axonal protein tau. Despite intense effort in recent years, the precise role of tau in the pathology of the various behaviourally and neuropathologically distinct tauopathies, the mechanisms of tau toxicity and the potential functional interaction of tau and amyloid in Alzheimer's disease remain elusive. Nevertheless, novel observations regarding the various aspects of taumisregulation-dependent pathogenesis are emerging from various cellular, vertebrate and invertebrate animal models and are supported by new clinical data. This Focused Meeting brought together scientists working on tau and tauopathies from different disciplines and various experimental models. The aim was to enhance our understanding of the protein itself and disorders associated with its misregulation through synergy.


Assuntos
Tauopatias/etiologia , Proteínas tau/fisiologia , Animais , Humanos , Modelos Animais , Sistema Nervoso/metabolismo , Sistema Nervoso/patologia , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/química , Proteínas tau/metabolismo
11.
Biochem Soc Trans ; 38(4): 988-92, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20658990

RESUMO

AD (Alzheimer's disease) is a neurodegenerative disorder characterized by the abnormal hyperphosphorylation and aggregation of the microtubule-associated protein tau and the misfolding and deposition of Abeta peptide. The mechanisms by which tau and Abeta become abnormal is not clearly understood, neither is it known what role either protein plays in the neurodegenerative process underlying AD. We have modelled aspects of AD in Drosophila melanogaster to shed light on these processes and to further our understanding of the relationship between tau and amyloid in this disease.


Assuntos
Doença de Alzheimer/patologia , Modelos Animais de Doenças , Drosophila melanogaster , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/fisiologia , Animais , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Humanos , Modelos Biológicos , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Peptídeo Hidrolases/metabolismo , Fosforilação , Transmissão Sináptica/fisiologia , Proteínas tau/metabolismo , Proteínas tau/fisiologia
12.
Biochem Soc Trans ; 38(2): 564-70, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20298222

RESUMO

Axonal microtubules are essential for transport of materials to the synapse. Compromised microtubules and synaptic loss have been demonstrated in AD (Alzheimer's disease), which is believed to contribute to cognitive dysfunction before neuronal death in the early stages of the disease. The mechanism by which hyperphosphorylated tau, the building block of neurofibrillary tangles, one of the pathological hallmarks of AD, disrupts neuronal and synaptic function is unclear. There is a theory that hyperphosphorylated tau does not bind effectively to microtubules and is no longer able to function in stabilizing them, thus axonal transport can no longer proceed efficiently. This leads to synaptic dysfunction. We have tested this theory in a Drosophila model of tauopathies in which we expressed human tau (h-tau). Using this model, we have tested all aspects of this hypothesis and have demonstrated that axonal transport does become compromised in the presence of hyperphosphorylated h-tau and this leads to synaptic and behavioural defects. We are currently investigating the mechanism by which hyperphosphorylated h-tau mediates this effect and are preliminary data indicate that this entails phospho-tau-mediated effects that are predicted by the tau-microtubule hypothesis, as well as novel effects. These deleterious effects of h-tau occur in the absence of tau filaments and before neuronal death. This sequence of pathogenic events may constitute the mechanism by which abnormal tau disrupts neuronal and synaptic function and contributes to cognitive impairment before neuronal death in the early stages of tauopathies such as AD.


Assuntos
Modelos Animais de Doenças , Drosophila , Neurônios/efeitos dos fármacos , Tauopatias/patologia , Proteínas tau/farmacologia , Animais , Drosophila/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Modelos Biológicos , Neurônios/fisiologia , Fosforilação/fisiologia , Solubilidade , Tauopatias/metabolismo , Tauopatias/fisiopatologia , Regulação para Cima/fisiologia , Proteínas tau/metabolismo
13.
Acta Neuropathol ; 120(5): 593-604, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20617325

RESUMO

It has been hypothesised that tau protein, when hyper-phosphorylated as in Alzheimer's disease (AD), does not bind effectively to microtubules and is no longer able to stabilise them; thus microtubules break down, and axonal transport can no longer proceed efficiently in affected brain regions in AD and related tauopathies (tau-microtubule hypothesis). We have used Drosophila models of tauopathy to test all components of this hypothesis in vivo. We have previously shown that upon expression of human 0N3R tau in Drosophila motor neurons it becomes highly phosphorylated, resulting in disruptions to both axonal transport and synaptic function which culminate in behavioural phenotypes. We now show that the mechanism by which the human tau mediates these effects is twofold: first, as predicted by the tau-microtubule hypothesis, the highly phosphorylated tau exhibits significantly reduced binding to microtubules; and second, it participates in a pathogenic interaction with the endogenous normal Drosophila tau and sequesters it away from microtubules. This causes disruption of the microtubular cytoskeleton as evidenced by a reduction in the numbers of intact correctly-aligned microtubules and the appearance of microtubules that are not correctly oriented within the axon. These deleterious effects of human tau are phosphorylation dependent because treatment with LiCl to suppress tau phosphorylation increases microtubule binding of both human and Drosophila tau and restores cytoskeletal integrity. Notably, all these phospho-tau-mediated phenotypes occur in the absence of tau filament/neurofibrillary tangle formation or neuronal death, and may thus constitute the mechanism by which hyper-phosphorylated tau disrupts neuronal function and contributes to cognitive impairment prior to neuronal death in the early stages of tauopathies.


Assuntos
Microtúbulos/metabolismo , Microtúbulos/patologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Proteínas tau/metabolismo , Animais , Western Blotting , Drosophila , Humanos , Imuno-Histoquímica , Imunoprecipitação , Microscopia Eletrônica de Transmissão , Fosforilação , Tauopatias/metabolismo , Tauopatias/patologia
14.
J Neurosci Methods ; 319: 77-88, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30633936

RESUMO

Work spanning almost two decades using the fruit fly, Drosophila melanogaster, to study tau-mediated neurodegeneration has provided valuable and novel insights into the causes and mechanisms of tau-mediated toxicity and dysfunction in tauopathies such as Alzheimer's disease (AD). The fly has proven to be an excellent model for human diseases because of its cost efficiency, and the availability of powerful genetic tools for use in a comparatively less-complicated, but evolutionarily conserved, in vivo system. In this review, we provide a critical evaluation of the insights provided by fly models, highlighting both the advantages and limitations of the system. The fly has contributed to a greater understanding of the causes of tau abnormalities, the role of these abnormalities in mediating toxicity and/or dysfunction, and the nature of causative species mediating tau-toxicity. However, it is not possible to perfectly model all aspects of human degenerative diseases. What sets the fly apart from other animal models is its genetic tractability, which makes it highly amenable to overcoming experimental limitations. The explosion of genetic technology since the first fly disease models were established has translated into fly lines that allow for greater temporal control in restricting tau expression to single neuron types, and lines that can label and monitor the function of subcellular structures and components; thus, fly models offer an unprecedented view of the neurodegenerative process. Emerging genetic technology means that the fly provides an ever-evolving experimental platform for studying disease.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Drosophila melanogaster/metabolismo , Tauopatias/metabolismo , Doença de Alzheimer/genética , Animais , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Humanos , Neurônios/metabolismo , Especificidade da Espécie , Tauopatias/genética , Proteínas tau/metabolismo
15.
Front Neurosci ; 13: 801, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31427921

RESUMO

Almost 50 million people in the world are affected by dementia; the most prevalent form of which is Alzheimer's disease (AD). Although aging is considered to be the main risk factor for AD, growing evidence from epidemiological studies suggests that type 2 diabetes mellitus (T2DM) increases the risk of dementia including AD. Defective brain insulin signaling has been suggested as an early event in AD and other tauopathies but the mechanisms that link these diseases are largely unknown. Tau hyperphosphorylation is a hallmark of neurofibrillary pathology and insulin resistance increases the number of neuritic plaques particularly in AD. Utilizing a combination of our Drosophila models of tauopathy (expressing the 2N4R-Tau) and neuroblastoma cells, we have attempted to decipher the pathways downstream of the insulin signaling cascade that lead to tau hyperphosphorylation, aggregation and autophagic defects. Using cell-based, genetic, and biochemical approaches we have demonstrated that tau phosphorylation at AT8 and PHF1 residues is enhanced in an insulin-resistant environment. We also show that insulin-induced changes in total and phospho-tau are mediated by the crosstalk of AKT, glycogen synthase kinase-3ß, and extracellular regulating kinase located downstream of the insulin receptor pathway. Finally, we demonstrate a significant change in the levels of the key proteins in the mammalian target of rapamycin/autophagy pathway, implying an increased impairment of aggregated protein clearance in our transgenic Drosophila models and cultured neuroblastoma cells.

16.
Front Neurosci ; 12: 383, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29950970

RESUMO

Alzheimer's disease (AD) and Type 2 Diabetes Mellitus (T2DM) are two of the most prevalent diseases in the elderly population worldwide. A growing body of epidemiological studies suggest that people with T2DM are at a higher risk of developing AD. Likewise, AD brains are less capable of glucose uptake from the surroundings resembling a condition of brain insulin resistance. Pathologically AD is characterized by extracellular plaques of Aß and intracellular neurofibrillary tangles of hyperphosphorylated tau. T2DM, on the other hand is a metabolic disorder characterized by hyperglycemia and insulin resistance. In this review we have discussed how Insulin resistance in T2DM directly exacerbates Aß and tau pathologies and elucidated the pathophysiological traits of synaptic dysfunction, inflammation, and autophagic impairments that are common to both diseases and indirectly impact Aß and tau functions in the neurons. Elucidation of the underlying pathways that connect these two diseases will be immensely valuable for designing novel drug targets for Alzheimer's disease.

17.
Acta Neuropathol Commun ; 6(1): 3, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29298722

RESUMO

Alzheimer's disease (AD) is the most common cause of dementia among older adults. Accumulation of amyloid-ß (Aß) in the brain is considered central in AD pathogenesis and its understanding crucial for developing new diagnostic and therapeutic approaches. Recent literature suggests that ageing may induce post translational modifications in Aß, in the form of spontaneous amino acid modifications, which enhance its pathogenic properties, contributing to its aggregation.In this study, we have investigated whether the isoaspartate (IsoD-Aß) and pyroglutamate (pE3-Aß) modified forms of Aß are significantly associated with AD pathology or represent markers of ageing. Cerebral neocortex of 27 AD cases, 32 old controls (OC) and 11 young controls (YC) was immunostained for pE3-Aß and IsoD-Aß, quantified as protein load and correlated with other Aß forms and p-TAU. IsoD-Aß and pE3-Aß were detected at low levels in non-demented controls, and significantly increased in AD (p ≤ 0.001), with a characteristic deposition of IsoD-Aß in blood vessel walls and pE3-Aß within neurons. Both AD and OC showed positive associations between IsoD-Aß and Aß (p = 0.003 in AD and p = 0.001 in OC) and between IsoD-Aß and pE3-Aß (p = 0.001 in AD and OC). This last association was the only significant pE3-Aß correlation identified in AD, whereas in the control cohorts pE3-Aß also correlated with Aß and AßPP (p = 0.001 in OC and p = 0.010 in YC).Our analyses suggest that IsoD-Aß accumulation starts with ageing; whereas pE3-Aß deposition is more closely linked to AD. Our findings support the importance of age-related modifications of Aß in AD pathogenesis.


Assuntos
Envelhecimento/patologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Ácido Isoaspártico/metabolismo , Ácido Pirrolidonocarboxílico/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Peptídeos beta-Amiloides/química , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neocórtex , Processamento de Proteína Pós-Traducional , Estatísticas não Paramétricas , Adulto Jovem
18.
Invert Neurosci ; 7(3): 165-71, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17636367

RESUMO

Hyperphosphorylation and aggregation of tau into tangles is a feature of disorders such as Alzheimer's disease and other Tauopathies. To model these disorders in Drosophila melanogaster, human tau has been over-expressed and a variety of phenotypes have been observed including neurotoxicity, disrupted neuronal and synaptic function and locomotor impairments. Neuronal dysfunction has been seen prior to neuronal death and in the absence of tangle formation. The Drosophila tau protein shares a large degree of homology with human tau but differs in the crucial microtubule binding domains. Although like human tau Drosophila tau can induce neurotoxicity, little is known about its ability to disrupt neuronal function. In this study we demonstrate that like human tau, over-expression of Drosophila tau results in disrupted axonal transport, altered neuromuscular junction morphology and locomotor impairments. This indicates that like human tau, over-expression of Drosophila tau compromises neuronal function despite significant differences in microtubule binding regions.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Neurônios/patologia , Tauopatias/patologia , Proteínas tau/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Axonal/fisiologia , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Larva , Movimento/fisiologia , Junção Neuromuscular/patologia , Neurônios/metabolismo , Tauopatias/metabolismo
19.
Trends Neurosci ; 25(1): 22-6, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11801334

RESUMO

The amyloid cascade hypothesis has been the predominant model of molecular pathogenesis in Alzheimer's disease. The finding of tau mutations in other dementias has added weight to the hypothesis as it suggests that tau-pathology is a downstream but essential part of the dementing process. However, some observations remain difficult to reconcile with the hypothesis. In transgenic mice, for example, amyloid generation does not induce the predicted cascade and in man, plaques and tangles are separated temporally and spatially. One alternative possibility is that some common factor, loss of wnt signalling for example, might induce both plaques and tangles.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra , Proteínas tau/metabolismo , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Humanos , Camundongos , Emaranhados Neurofibrilares/metabolismo , Placa Amiloide/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Wnt
20.
Sci Rep ; 6: 38224, 2016 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-27910888

RESUMO

The microtubule cytoskeleton is a highly dynamic, filamentous network underpinning cellular structure and function. In Alzheimer's disease, the microtubule cytoskeleton is compromised, leading to neuronal dysfunction and eventually cell death. There are currently no disease-modifying therapies to slow down or halt disease progression. However, microtubule stabilisation is a promising therapeutic strategy that is being explored. We previously investigated the disease-modifying potential of a microtubule-stabilising peptide NAP (NAPVSIPQ) in a well-established Drosophila model of tauopathy characterised by microtubule breakdown and axonal transport deficits. NAP prevented as well as reversed these phenotypes even after they had become established. In this study, we investigate the neuroprotective capabilities of an analogous peptide SAL (SALLRSIPA). We found that SAL mimicked NAP's protective effects, by preventing axonal transport disruption and improving behavioural deficits, suggesting both NAP and SAL may act via a common mechanism. Both peptides contain a putative 'SIP' (Ser-Ile-Pro) domain that is important for interactions with microtubule end-binding proteins. Our data suggests this domain may be central to the microtubule stabilising function of both peptides and the mechanism by which they rescue phenotypes in this model of tauopathy. Our observations support microtubule stabilisation as a promising disease-modifying therapeutic strategy for tauopathies like Alzheimer's disease.


Assuntos
Doença de Alzheimer/prevenção & controle , Microtúbulos/metabolismo , Peptídeos/farmacologia , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Axonal/efeitos dos fármacos , Transporte Axonal/genética , Drosophila melanogaster , Humanos , Microtúbulos/genética , Microtúbulos/patologia , Proteínas tau/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA