Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Cell Neurosci ; 88: 201-211, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29427776

RESUMO

Hyperphosphorylation of tau is one of the main hallmarks for Alzheimer's disease (AD) and many other tauopathies. Norepinephrine (NE), a stress-related hormone and 17-ß-estradiol (E2) thought to influence tau phosphorylation (p-tau) and AD pathology. The controversy around the impact of NE and E2 requires further clarification. Moreover, the combination effect of physiological and psychological stress and estrogen alteration during menopause, which affect p-tau, has not been addressed. Exposure to E2 is believed to reduce NE release, however, the link between these two hormones and AD at cellular level was also remained unknown. Here, we examined whether NE and E2 treatment of differentiated SH-SY5Y cells affected tau phosphorylation. The involvement of adenosine monophosphate kinase protein kinase (AMPK) and target of Rapamycin (mTOR) as the possible mechanisms, underlying this effect was also investigated. Subsequent to SH-SY5Y differentiation to mature neurons, we treated the cells with NE, E2 and NE plus E2 in presence and absence of Compound C and Rapamycin. Cell viability was not affected by our treatment while our Western blot and immunofluorescent findings showed that exposure to NE and E2 separately, and in combination enhanced p-tau (Ser396) and (Ser262)/tau but not (Ser202/Thr205)/tau. Blocking AMPK by Compound C reduced p-tau (Ser396) and (Ser262), while GSK-3ß and PP2A activities were remained unchanged. We also found that blocking mTOR by Rapamycin did not change increased p-tau (Ser396) and (Ser262) due to NE + E2 treatment. Collectively, our results suggested that tau hyperphosphorylation due to exposure to NE/E2 was mediated by AMPK, the main energy regulator of cells during stress with no significant involvement of mTOR, GSK-3ß and PP2A.


Assuntos
Adenilato Quinase/metabolismo , Diferenciação Celular/efeitos dos fármacos , Norepinefrina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/efeitos dos fármacos , Monofosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Estradiol/farmacologia , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Fosforilação/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Proteínas tau/metabolismo
2.
BMC Cell Biol ; 19(1): 7, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29921218

RESUMO

BACKGROUND: Cellular energy failure in high metabolic rate organs is one of the underlying causes for many disorders such as neurodegenerative diseases, cardiomyopathies, liver and renal failures. In the past decade, numerous studies have discovered the cellular axis of LKB1/AMPK/mTOR as an essential modulator of cell homeostasis in response to energy stress. Through regulating adaptive mechanisms, this axis adjusts the energy availability to its demand by a systematized control on metabolism. Energy stress, however, could be sensed at different levels in various tissues, leading to applying different strategies in response to hypoxic insults. METHODS: Here the immediate strategies of high metabolic rate organs to time-dependent short episodes of ischaemia were studied by using a rat model (n = 6/group) of cardiac arrest (CA) (15 and 30 s, 1, 2, 4 and 8 min CA). Using western blot analysis, we examined the responses of LKB1/AMPK/mTOR pathway in brain, heart, liver and kidney from 15 s up to 8 min of global ischaemia. The ratio of ADP/ATP was assessed in all ischemic and control groups, using ApoSENSOR bioluminescent assay kit. RESULTS: Brain, followed by kidney showed the early dephosphorylation response in AMPK (Thr172) and LKB1 (Ser431); in the absence of ATP decline (ADP/ATP elevation). Dephosphorylation of AMPK was followed by rephosphorylation and hyperphosphorylation, which was associated with a significant ATP decline. While heart's activity of AMPK and LKB1 remained at the same level during short episodes of ischaemia, liver's LKB1 was dephosphorylated after 2 min. AMPK response to ischaemia in liver was mainly based on an early alternative and a late constant hyperphosphorylation. No significant changes was observed in mTOR activity in all groups. CONCLUSION: Together our results suggest that early AMPK dephosphorylation followed by late hyperphosphorylation is the strategy of brain and kidney in response to ischaemia. While the liver seemed to get benefit of its AMPK system in early ischameia, possibly to stabilize ATP, the level of LKB1/AMPK activity in heart remained unchanged in short ischaemic episodes up to 8 min. Further researches must be conducted to elucidate the molecular mechanism underlying LKB1/AMPK response to oxygen supply.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Parada Cardíaca/metabolismo , Parada Cardíaca/patologia , Isquemia/metabolismo , Especificidade de Órgãos , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Modelos Animais de Doenças , Eletrocardiografia , Isquemia/patologia , Rim/irrigação sanguínea , Rim/patologia , Fígado/irrigação sanguínea , Fígado/patologia , Miocárdio/patologia , Fosforilação , Ratos Sprague-Dawley
3.
Eur J Neurosci ; 44(3): 1987-97, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27177932

RESUMO

Abnormal tau phosphorylation (p-tau) has been shown after hypoxic damage to the brain associated with traumatic brain injury and stroke. As the level of p-tau is controlled by Glycogen Synthase Kinase (GSK)-3ß, Protein Phosphatase 2A (PP2A) and Adenosine Monophosphate Kinase (AMPK), different activity levels of these enzymes could be involved in tau phosphorylation following ischaemia. This study assessed the effects of global brain ischaemia/reperfusion on the immediate status of p-tau in a rat model of cardiac arrest (CA) followed by cardiopulmonary resuscitation (CPR). We reported an early dephosphorylation of tau at its AMPK sensitive residues, Ser(396) and Ser(262) after 2 min of ischaemia, which did not recover during the first two hours of reperfusion, while the tau phosphorylation at GSK-3ß sensitive but AMPK insensitive residues, Ser(202) /Thr(205) (AT8), as well as the total amount of tau remained unchanged. Our data showed no alteration in the activities of GSK-3ß and PP2A during similar episodes of ischaemia of up to 8 min and reperfusion of up to 2 h, and 4 weeks recovery. Dephosphorylation of AMPK followed the same pattern as tau dephosphorylation during ischaemia/reperfusion. Catalase, another AMPK downstream substrate also showed a similar pattern of decline to p-AMPK, in ischaemic/reperfusion groups. This suggests the involvement of AMPK in changing the p-tau levels, indicating that tau dephosphorylation following ischaemia is not dependent on GSK-3ß or PP2A activity, but is associated with AMPK dephosphorylation. We propose that a reduction in AMPK activity is a possible early mechanism responsible for tau dephosphorylation.


Assuntos
Isquemia Encefálica/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Parada Cardíaca/metabolismo , Proteínas Quinases/metabolismo , Proteína Fosfatase 2/metabolismo , Traumatismo por Reperfusão/metabolismo , Proteínas tau/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Isquemia Encefálica/etiologia , Catalase/metabolismo , Feminino , Parada Cardíaca/complicações , Fosforilação , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/etiologia
4.
BMC Neurosci ; 16: 69, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26499115

RESUMO

Accumulation of protein aggregates is the leading cause of cellular dysfunction in neurodegenerative disorders. Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, Prion disease and motor disorders such as amyotrophic lateral sclerosis, present with a similar pattern of progressive neuronal death, nervous system deterioration and cognitive impairment. The common characteristic is an unusual misfolding of proteins which is believed to cause protein deposition and trigger degenerative signals in the neurons. A similar clinical presentation seen in many neurodegenerative disorders suggests the possibility of shared neuronal responses in different disorders. Despite the difference in core elements of deposits in each neurodegenerative disorder, the cascade of neuronal reactions such as activation of glycogen synthase kinase-3 beta, mitogen-activated protein kinases, cell cycle re-entry and oxidative stress leading to a progressive neurodegeneration are surprisingly similar. This review focuses on protein toxicity in two neurodegenerative diseases, AD and PD. We reviewed the activated mechanisms of neurotoxicity in response to misfolded beta-amyloid and α-synuclein, two major toxic proteins in AD and PD, leading to neuronal apoptosis. The interaction between the proteins in producing an overlapping pathological pattern will be also discussed.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doença de Parkinson/metabolismo , Dobramento de Proteína , Transdução de Sinais/fisiologia , alfa-Sinucleína/metabolismo , Humanos
5.
Neurochem Res ; 36(6): 967-77, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21380845

RESUMO

The loss of nigral dopaminergic (DA) neurons is the disease-defining pathological change responsible for progressive motor dysfunction in Parkinson's disease. In this study, we sought to establish a culture method for adult rat tyrosine hydroxylase (TH)-immunoreactive DA neurons. In this context, we investigated the role of fibroblast growth factor 2 (FGF2), brain-derived neurotrophic factor (BDNF), transforming growth factor-ß3 (TGF-ß3), glial-derived neurotrophic factor (GDNF) and dibutyryl-cyclic AMP (dbcAMP) in these cultures. Culturing in the presence of FGF2, BDNF and GDNF enhanced the survival of DA neurons by 15-fold and promoted neurite growth. In contrast, dbcAMP promoted neurite growth in all neurons but did not enhance DA cell survival. This study demonstrates that long-term cultures of DA neurons can be established from the mature rat brain and that survival and regeneration of DA neurons can be manipulated by epigenetic factors such as growth factors and intracellular cAMP pathways.


Assuntos
Encéfalo/enzimologia , Dopamina/metabolismo , Neurônios/enzimologia , Regeneração , Tirosina 3-Mono-Oxigenase/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Feminino , Técnicas In Vitro , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
6.
Front Neurosci ; 13: 155, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30881282

RESUMO

Alzheimer's disease (AD) and cancer are among the leading causes of human death around the world. While neurodegeneration is the main feature of AD, the most important characteristic of malignant tumors is cell proliferation, placing these two diseases in opposite sides of cell division spectrum. Interestingly, AD and cancer's pathologies consist of a remarkable common feature and that is the presence of active cell cycle in both conditions. In an in vitro model of primary adult neuronal culture, we previously showed that treating cell with beta amyloid forced neurons to start a cell cycle. Instead of cell division, however, neuronal cell cycle was aborted and a massive neurodegeneration was left behind as the consequence. A high level of cell cycle entry, which is a requirement for cancer pathogenesis, was reported in clinically diagnosed cases of AD, leading to neurodegeneration. The diverse clinical manifestation of a similar etiology, have puzzled researchers for many years. In fact, the evidence showed an inverse association between AD and cancer prevalence, suggesting that switching pathogenesis toward AD protects patients against cancer and vice versa. In this mini review, we discussed the possibility of involvement of cell proliferation and survival dysregulation as the underlying mechanism of neurodegeneration in AD, and the leading event to develop both disorders' pathology. As examples, the role of phosphoinositide 3 kinase/Akt/ mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway in cell cycle re-entry and blocking autophagy are discussed as potential common intracellular components between AD and cancer pathogenesis, with diverse clinical diagnosis.

7.
Brain Res ; 1218: 224-9, 2008 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-18533137

RESUMO

In Alzheimer's disease (AD) cell cycle reentry precedes neuronal death, which could be induced by many cytotoxic factors. It is believed that beta amyloid (Abeta), the major component of extracellular plaques in AD, is potent in inducing neurons to reenter cell cycle. In AD brains, neurons expressing cell cycle markers are reported in many brain regions without any plaque formation, although very low levels of Abeta may still be detected. In the other side, because cell cycle reentry is not an immediate cause of apoptosis, neurons may remain in cell cycle phases for some time prior to their final death. In this study we examined if very low concentrations of Abeta 1-42 (picomolar) can trigger the adult neurons to reenter the cell cycle, and the effect of different Abeta concentrations on neuronal progression through different cell cycle phases. Primary adult neurons were treated with Abeta 1-42 at 2 x 10(-6), 2 x 10(-5), 2 x 10(-4), 0.5 and 2.5 microM concentrations. Cyclin D1 and cyclin B1 (the markers for G1 and G2 phases of the cell cycle, respectively) and apoptosis were assessed. Treatment with Abeta at 2.5 microM induced apoptosis. At lower levels however, Abeta promoted neurons entering G1 and G2 phases without apoptosis, with 0.5 microM of Abeta inducing neurons into G2, and 2 x 10(-5,) 2 x 10(-4) into G1 phases. Our results suggested that lower concentrations of Abeta induced neurons to reenter the cell cycle, and different concentrations had differential abilities to promote neurons into various cell cycle phases or trigger their death.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Ciclo Celular/efeitos dos fármacos , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Ciclina B/metabolismo , Ciclina B1 , Ciclina D1/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Ratos
8.
Iran Biomed J ; 12(2): 101-7, 2008 04.
Artigo em Inglês | MEDLINE | ID: mdl-18506216

RESUMO

BACKGROUND: Primary cultures of embryonic neurons have been used to introduce a model of neurons in physiological and pathological conditions. However, age-related cellular events limit this method as an optimal model in adult neurodegenerative diseases studies. Besides, short-interval changing media in previous cultures decreases the effectiveness of this model. As an example of this matter, we can refer to the study on some special neuronal secreted factors or the influence of some experimental materials on neurons. Meanwhile, short-interval changing media could remove the effects of some released factors from the environment. In this study, the method for isolation and culturing adult rat hippocampal neurons with long-intervals medium changing has been described. METHODS: The hippocampal neurons of adult male rats were cultured. We used Neurobasal A/B27 culture medium, papain (2 mg/ml), trypsin 0.25% and collagenase (1 mg/ml) for neuronal isolation, OptiPrep density gradient for separation of neurons from other cell types and also debris and FGF2 (10 ng/ml) for increasing neuronal survival and regeneration. RESULTS: The neuronal sprouting and viability were increased by using papain and mild triturating (P<0.05). Adult neuronal culturing and their regeneration were impossible without FGF2. It was shown that adding new fresh medium every 4 days and exchanging half of it every 8 days had no detrimental effect on neuronal viability. CONCLUSION: This investigation shows the possibility of culturing adult neuronal cells and their maintaining in long-interval media. It could be happened because of adult neurons rely significantly on the neighboring cells secreted factors for living and making synaptic connections. This model is very useful in physiological and pathological studies which need stable conditions of neuronal culture in a long period of time.


Assuntos
Meios de Cultura , Hipocampo/citologia , Neurônios/citologia , Animais , Técnicas de Cultura de Células/métodos , Divisão Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Hipocampo/metabolismo , Hipocampo/fisiologia , Masculino , Neurônios/metabolismo , Neurônios/fisiologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
9.
Curr Alzheimer Res ; 15(8): 764-776, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29473507

RESUMO

BACKGROUND: Emerging evidence supports the hypothesis that metabolism dysfunction is involved in pathogenesis of Alzheimer's disease (AD). One aspect of metabolic dysfunction includes dysregulation of adenosine monophosphate kinase protein kinase (AMPK) and mammalian target of rapamycin (mTOR) metabolic axis, which is extensively present in some of the leading causes of AD such as cerebrovascular diseases, type 2 diabetes and brain ischaemic events. While the molecular basis underlying this metabolic dysregulation remains a significant challenge, mitochondrial dysfunction due to aging appears to be an essential factor to activate AMPK/mTOR signaling pathway, leading to abnormal neuronal energy metabolism and AD pathology. METHODS: Using immunofluorescent imaging by Lecia confocal microscopy, we analyzed the activation of AMPK/mTOR. Concurrently, the level of mitochondrial antioxidant enzymes of superoxide dismutase 2 (SOD2) and peroxiredoxin 1 and 4 (p1 and p4) along with protein and DANA oxidation were examined to in postmortem brains of AD (n= 8) and normal (n= 7) subjects to evaluate the metabolism dysfunction role in AD pathology. RESULTS: In spite of AMPK inhibitory control on mTOR, concurrent phosphorylation of AMPK and mTOR (p-AMPK and p-mTOR) was observed in AD brains with high colocalization with hyperphosphorylated tau. Mitochondrial antioxidant enzymes of SOD2 and p1 and p4 were substantially decreased in p-AMPK, p-mTOR and p-tau positive cells along with higher levels of DNA and protein oxidation. CONCLUSION: Collectively, we conclude that AMPK and mTOR metabolic axis is highly activated in AD brains. While the inhibitory link between AMPK and mTOR seems to be disrupted, we suggest oxidative stress as the underlying mechanism for concurrent activation of AMPK and mTOR in AD.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Doença de Alzheimer/metabolismo , Peroxirredoxinas/metabolismo , Superóxido Dismutase/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Estresse Oxidativo/fisiologia
10.
Neurosci Lett ; 670: 53-61, 2018 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-29398519

RESUMO

Aggregation of hyperphosphorylated tau (p-tau) in the form of neurofibrillary tangles (NFT) is a main hallmark for Alzheimer's disease (AD). Activation of cellular metabolic axis, made of adenosine monophosphate kinase protein kinase (AMPK) and mammalian target of rapamycin (mTOR) have been implicated in generating tau pathology of AD. Thus, blocking either of these two proteins or both, are suggested as the future therapeutic approaches for AD. How and to what level these approaches could be applied, however are not entirely clear. By using Compound C (CC) in this study, we showed a substantial decrease in mTOR activity in a rapamycin-independent way without blocking AMPK. This decline in mTOR activity was accompanied by an increase in phosphoinositide 3 kinase (PI3K)/Akt activity and a parallel increase in p-tau (Ser396) but not p-tau (Ser262) in differentiated SH-SY5Y neuroblastoma cells. This elevation was blocked when the cells were treated with 15 µM of LY294002, a specific PI3K inhibitor, suggesting PI3K involvement in CC-mediated tau hyperphosphorylation at Ser396. For all groups the activity levels of glycogen synthase kinase-3ß (GSK-3ß), cyclin-dependent kinase-5 (cdk5) and protein phosphatase 2A (PP2A), the other main kinases and phosphatase responsible for tau phosphorylation/dephosphorylation remained unchanged. Collectively, our results demonstrate that rapamycin-independent blocking of mTOR enhances p-tau (Ser396) in a PI3K-dependent way, suggesting the careful consideration of future therapeutic approaches for AD, which will be based on mTOR inhibition.


Assuntos
Adenilato Quinase/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas tau/metabolismo , Adenilato Quinase/metabolismo , Linhagem Celular Tumoral , Quinase 5 Dependente de Ciclina/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos
11.
Curr Alzheimer Res ; 15(10): 928-937, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29895248

RESUMO

BACKGROUND: Alzheimer's disease (AD) pathology consists of intraneuronal neurofibrillary tangles, made of hyperphosphorylated tau and extracellular accumulation of beta amyloid (Aß) in Aß plaques. There is an extensive debate as to which pathology initiates and is responsible for cellular loss in AD. METHODS: Using confocal and light microscopy, post mortem brains from control and AD cases, an antibody to SOD2 as a marker for mitochondria and an antibody to all forms of tau, we analyzed mitochondrial density in tau positive neurons along with nuclear degradation by calculating the raw integrative density. RESULTS: Our findings showed an extensive staining of aggregated tau in cell bodies, dystrophic neurites and neurofilaments in AD with minimal staining in control tissue, along with a marked decrease in mitochondria in tau positive (tau+) neurons. The control or tau negative (tau-) neurons in AD contained an even distribution of mitochondria, which was greatly diminished in tau+ neurons by 40%. There were no significant differences between control and tau- neurons in AD. Tau+ neurons showed marked nuclear degradation which appeared to progress with the extent of tau aggregation. The aggregated tau infiltrated and appeared to break the nuclear envelope with progressively more DNA exiting the nucleus and associating with the aggregated intracellular tau. CONCLUSION: We report that the mitochondrial decrease is likely due to a decrease in the protein synthesis rather than a redistribution of mitochondria because of the decreased axonal transport. We suggest that the decrease in mitochondria and nuclear degradation are key mechanisms for the neuronal loss seen in AD.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Nucléolo Celular/patologia , Mitocôndrias/patologia , Neurônios , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/metabolismo , Autopsia , Feminino , Humanos , Indóis/metabolismo , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Emaranhados Neurofibrilares/patologia , Neurônios/metabolismo , Neurônios/patologia , Neurônios/ultraestrutura , Superóxido Dismutase/metabolismo , Proteínas tau
12.
Front Neurosci ; 12: 533, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30150923

RESUMO

The overwhelming majority of dominant mutations causing early onset familial Alzheimer's disease (EOfAD) occur in only three genes, PSEN1, PSEN2, and APP. An effect-in-common of these mutations is alteration of production of the APP-derived peptide, amyloid ß (Aß). It is this key fact that underlies the authority of the Amyloid Hypothesis that has informed Alzheimer's disease research for over two decades. Any challenge to this authority must offer an alternative explanation for the relationship between the PSEN genes and APP. In this paper, we explore one possible alternative relationship - the dysregulation of cellular iron homeostasis as a common effect of EOfAD mutations in these genes. This idea is attractive since it provides clear connections between EOfAD mutations and major characteristics of Alzheimer's disease such as dysfunctional mitochondria, vascular risk factors/hypoxia, energy metabolism, and inflammation. We combine our ideas with observations by others to describe a "Stress Threshold Change of State" model of Alzheimer's disease that may begin to explain the existence of both EOfAD and late onset sporadic (LOsAD) forms of the disease. Directing research to investigate the role of dysregulation of iron homeostasis in EOfAD may be a profitable way forward in our struggle to understand this form of dementia.

13.
Brain Res ; 1185: 321-7, 2007 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-17961521

RESUMO

Alzheimer's disease (AD) is a chronic disorder with progressive neurodegeneration associated with aging and is characterized by fibrillar beta-amyloid (Abeta) deposits in the brain. Although the increased production of Abeta seems to play a noticeable role in AD pathogenesis and its progression, all the mechanisms which are involved in this extracellular Abeta elevation are not known completely. In the present study, we used adult hippocampal neuronal culture as an in vitro model which is favorable for adult neurodegenerative diseases' studies. We introduced a toxic concentration for fibrillar Abeta1-42 in adult neurons which was much lower from the toxic concentration in embryonic neurons. To determine the effect of fibrillar Abeta1-42 which is the most toxic part of amyloid plaques, on extracellular Abeta1-40, as the main part of betaAPP proteolysis products, we treated the neurons with fibrillar Abeta1-42 at nontoxic concentrations of 2 x 10(-6), 2 x 10(-5) and 2 x 10(-4) microM and measured extracellular Abeta1-40. Our findings show that even very low levels of fibrillar Abeta1-42 can contribute to subsequent extracellular Abeta elevation in a dose dependent manner. These results suggest that even low levels of fibrillar Abeta may have deleterious actions if it remains in extracellular space for a period of time.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Líquido Extracelular/efeitos dos fármacos , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeos beta-Amiloides/farmacologia , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Relação Dose-Resposta a Droga , Marcação In Situ das Extremidades Cortadas , Técnicas de Cultura de Órgãos , Ratos
14.
IBRO Rep ; 2: 1-13, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30135928

RESUMO

An increase in phosphorylated tau (p-tau) is associated with Alzheimer's disease (AD), and brain hypoxia. Investigation of the association of residue-specific tau hyperphosphorylation and changes in cognition, leads to greater understanding of its potential role in the pathology of memory impairment. The aims of this study are to investigate the involvement of the main metabolic kinases, Liver Kinase B1 (LKB1) and Adenosine Monophosphate Kinase Protein Kinase (AMPK), in tau phosphorylation-derived memory impairment, and to study the potential contribution of the other tau kinases and phosphatases including Glycogen Synthase Kinase (GSK-3ß), Protein kinase A (PKA) and Protein Phosphatase 2A (PP2A). Spatial memory and learning were tested in a rat global brain ischemic model of reversible cardiac arrest (CA). The phosphorylation levels of LKB1, AMPK, GSK-3ß, PP2A, PKA and tau-specific phosphorylation were assessed in rats, subjected to ischaemia/reperfusion and in clinically diagnosed AD and normal human brains. LKB1 and AMPK phosphorylation increased 4 weeks after CA as did AMPK related p-tau (Ser262). The animals showed unchanged levels of GSK-3ß specific p-tau (Ser202/Thr205), phospho-PP2A (Tyr307), total GSK-3ß, PP2A, phospho-cAMP response element-binding protein (CREB) which is an indicator of PKA activity, and no memory deficits. AD brains had hyperphosphorylated tau in all the residues of Ser262, Ser202 and Thr205, with increased phosphorylation of both AMPK (Thr172) and GSK-3ß (Ser9), and reduced PP2A levels. Our data suggests a crucial role for a combined activation of tau kinases and phosphatases in adversely affecting memory and that hyperphosphorylation of tau in more than one specific site may be required to create memory deficits.

15.
Neurotox Res ; 23(1): 69-78, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22610785

RESUMO

In spite of definite roles for ß-amyloid (Aß) in familial Alzheimer's disease (AD), the cause of sporadic AD remains unknown. Amyloid senile plaques and Lewy body pathology frequently coexist in neocortical and hippocampal regions of AD and Parkinson's diseases. However, the relationship between Aß and α-synuclein (α-Syn), the principle components in the pathological structures, in neuronal toxicity and the mechanisms of their interaction are not well studied. As Aß and α-Syn accumulate in aging patients, the biological functions and toxicity of these polypeptides in the aging brain may be different from those in young brain. We examined the neurotoxicity influences of Aß1-42 or α-Syn on mature neurons and the effects of Aß1-42 or α-Syn on the production of endogenous α-Syn or Aß1-40 reciprocally using a model of culture enriched with primary neurons from the hippocampus of adult rats. Treatment of neurons with high concentrations of Aß1-42 or α-Syn caused significant apoptosis of neurons. Following Aß1-42 treatment at sub apoptotic concentrations, both intra- and extra-cellular α-Syn levels were significantly increased. Reciprocally, the non-toxic levels of α-Syn treatment also increased intra- and extra-cellular Aß1-40 levels. The phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002, suppressed α-Syn-induced Aß1-40 elevation, as well as Aß1-42-induced α-Syn elevation. Thus, high concentrations of Aß1-42 and α-Syn exert toxic effects on mature neurons; however, non-toxic concentration treatment of these polypeptides induced the production of each other reciprocally with possible involvement of PI3K pathway.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/toxicidade , alfa-Sinucleína/biossíntese , alfa-Sinucleína/toxicidade , Animais , Células Cultivadas , Sinergismo Farmacológico , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Neurotoxinas/biossíntese , Neurotoxinas/toxicidade , Ratos , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
16.
Neurotox Res ; 17(3): 257-67, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19680737

RESUMO

Alzheimer's disease (AD) is characterized pathologically by the deposition of amyloid-beta peptides (Abeta), neurofibrillary tangles, distinctive neuronal loss and neurite dystrophy. Nerve growth factor (NGF) has been suggested to be involved in the pathogenesis of AD, however, the role of its precursor (proNGF) in AD remains unknown. In this study, we investigated the effect of proNGF on neuron death, neurite growth and Abeta production, in vitro and in vivo. We found that proNGF promotes the death of different cell lines and primary neurons in culture, likely dependent on the expression of p75(NTR). We for the first time found that proNGF has an opposite role in neurite growth to that of mature NGF, retarding neurite growth in both cell lines and primary neurons. proNGF is localized to the Abeta plaques in AD mice brain, however, it had no significant effect on Abeta production in vitro and in vivo. Our findings suggest that proNGF is an important factor involving AD pathogenesis.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fator de Crescimento Neural/toxicidade , Neuritos/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Precursores de Proteínas/toxicidade , Precursor de Proteína beta-Amiloide/genética , Análise de Variância , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/citologia , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Camundongos , Camundongos Transgênicos , Fator de Crescimento Neural/farmacologia , Neuroblastoma , Presenilina-1/genética , Receptor de Fator de Crescimento Neural/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA