Preferencje help
Widoczny [Schowaj] Abstrakt
Liczba wyników

Znaleziono wyników: 9

Liczba wyników na stronie
Pierwsza strona wyników Pięć stron wyników wstecz Poprzednia strona wyników Strona / 1 Następna strona wyników Pięć stron wyników wprzód Ostatnia strona wyników

Wyniki wyszukiwania

help Sortuj według:

help Ogranicz wyniki do:
Pierwsza strona wyników Pięć stron wyników wstecz Poprzednia strona wyników Strona / 1 Następna strona wyników Pięć stron wyników wprzód Ostatnia strona wyników
Alpha-Synuclein (ASN), a small cytosolic protein enriched in synaptic terminals, was implicated in the pathomechanism of several neurodegenerative disorders called alpha-synucleinopathies. ASN was shown to be a main component of characteristic intraneuronal protein aggregates called Lewy bodies (LB) and Lewy neurites (LN), observed i.a. in Parkinson’s disease, dementia with LBs and in the LB variant of Alzheimer’s disease. Recent studies demonstrated that ASN may exist also in the extracellular space. Low-molecular ASN aggregates distributed in the brain parenchyma likely may be more toxic than ASN in LB, however, the exact mechanism of cytotoxicity of extracellular ASN is not fully understood. Our previous studies demonstrated the significant impact of extracellular ASN on calcium homeostasis. ASN evoked deregulation of intracellular calcium concentration leading in consequence to enhancement of nitric oxide synthesis. Deregulation of calcium homeostasis affects other calcium-dependent enzymes, including Calpains. The aim of the present study was to investigate the involvement of Calpaindependent activation of Cyclin Dependent Kinase 5 (Cdk5) in molecular mechanism of extracellular ASN cytotoxicity. The activation of Cdk5 is regulated by binding of regulatory subunits p35 and p39. Deregulation of calcium homeostasis may induce the Calpainmediated breakdown of Cdk5/p35 into Cdk5/p25 leading to overactivation of Cdk5. In our studies we used rat Pheochromocytoma PC12 cells incubated with exogenous ASN (10 µM) in the presence of Calpain inhibitor Calpeptin (10 µM) and Cdk5 inhibitors Roscovitine (10 µM) and BML-259 (10 µM). Our results indicated that incubation of PC12 cells in the presence of extracellular ASN (10 µM) for 48 h evoked cell death, and Cdk5 inhibitors efficiently prevented ASN toxicity, indicating an important role of Cdk5 in molecular mechanism of ASN toxicity. The level of Cdk5 protein was unchanged, but phosphorylation of Cdk5 at Tyr15 was significantly increased, suggesting that the enzymatic activity of Cdk5 is increased in ASN-treated cells. The presence of p25 protein was observed, what suggests that Calpain-dependent proteolysis of p35 occurred in ASN-treated cells. Calpeptin, an inhibitor of Calpains, prevented ASN-induced cell death, confirming the important role of Calpain activation in mechanism of ASN toxicity. In summary, our results demonstrated that alteration of calcium homeostasis evoked by extracellular ASN induce Calpain-dependent overactivation of Cdk5. These molecular processes may be involved in ASN-evoked cell death in vitro and probably also in neurodegenerative disorders.
Alterations of phosphorylation-dephosphorylation processes play a crucial role in the pathomechanism of Alzheimer’s disease (AD). They affect signaling cascades and lead to hyperphosphorylation of tau protein. Glycogen synthase kinase 3β (Gsk-3β) is the main tau-kinase; however, little is known about the role of sphingolipid pathway in its regulation. Alteration of sphingolipid biostat may be an early event in etiopathology of AD. The question arises, if the sphingosine kinase (Sphk), a key enzyme in sphingolipid pathway, regulates Gsk-3β? We analyzed acute effects of exogenous amyloid β (Aβ) oligomers in PC12 cells, and prolonged exposition to endogenous Aβ in PC12 cells stably expressing human Swedish mutant APPsw gene. Our data indicated that in cells subjected to exogenous Aβ expression of Sphk1 and phosphorylation of Gsk-3β at Ser9 were enhanced, what could be considered as a component of protective mechanism. However, prolonged liberation of Aβ in APPsw cells evoked inhibition of Sphk1 expression, activation of Gsk-3β and death of significant population of cells. Consequently, an inhibitor of Sphk1 also reduced cell viability. Our data suggest the existence of specific relationship between Sphk1 and Gsk-3β and indicate their role in alteration of cell function and survival. The study was supported by MSHE Grant N401 587040.
Hyperphosphorylation of tau is involved in the pathomechanism of neurodegenerative disorders such as Alzheimer’s and Parkinson’s diseases. Recent studies suggested the significance of alpha-synuclein (ASN) in tau phosphorylation, however, the molecular mechanism responsible for ASN-mediated tau modification remains to be elucidated. In this study, we investigated the role of extracellular ASN in tau phosphorylation in PC12 dopaminergic cells and the involvement of glycogen synthase kinase-3 beta (GSK-3β) and cyclin-dependent kinase 5 (CDK5) in ASN-induced tau modification and cell death. We found that exogenously added ASN (10 µM) stimulates the phosphorylation of tau at Ser396 in PC12 cells. A specific GSK-3β inhibitor (SB-216763) prevented ASN-evoked tau hyperphosphorylation without effect of CDK5 inhibitors. Furthermore, we found that ASN enhanced of GSK-3β protein level and activity. Cell viability determined by MTT assay and Hoechst 33258 staining showed that ASN induced PC12 cell death that presented typical apoptotic morphology. SB-216763 prevented apoptotic cell death evoked by ASN. Concluding, extracellular ASN is involved in GSK-3β-dependent tau modulation and its proapoptotic effect might be mediated at least in part by GSK-3β-catalysed tau phosphorylation and cytoskeleton destabilisation. Supported by a grant from The National Science Centre 2012/05/B/NZ3/02047.
alpha-Synuclein (ASN) play important role in pathogenesis of Parkinson’s disease (PD) and other neurodegenerative disorders. Novel and most interesting data showed elevated tauopathy in PD and suggested relationship between ASN and Tau protein. However, the mechanism of ASN-evoked Tau protein modification is not fully elucidated. In this study, we investigated the role of glycogen synthase kinase-3β (Gsk-3β) and cyclin-dependent kinase 5 (Cdk5) in ASN-evoked Tau modification in dopaminergic PC12 cells. We used real-time quantitative PCR (qRT-PCR) analysis to assess Gsk3β gene expression and Western blot technique to analyse protein phosphorylation. The presence of apoptotic cells was assessed by Hoechst 33258 fluorescent staining, and cell viability was determined by the 2-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay. Our data showed that exogenously added ASN (10 μM) increases Tau phosphorylation on Ser396 and specific Gsk-3β inhibitor (SB-216763, 10 µM) opposite to Cdk5 inhibitor protects cells against Tau hyperphosphorylation. Western blot analysis showed that ASN affected Gsk-3β via increasing of protein level and activation of this enzyme. From immunochemical studies, was found that ASN treatment leads to significant increase in GSK-3β immunoreactivity by about 20%. GSK-3β activity evaluated by its phosphorylation status assay showed that ASN significantly increased the phosphorylation of this enzyme at Tyr216 with parallel decrease in phosphorylation at Ser9, indicative of stimulation of GSK-3β activity. ASN-induced apoptotic processes leads to decrease of PC12 cells viability, the apoptotic cells determined by phase contrast together with Hoechst 33258 fluorescent staining, indicated significantly increase of apoptosis in the presence of ASN. SB-216763 prevented ASN-induced cytotoxicity and enhanced PC12 cell viability. In conclusion, all these findings suggested that extracellular ASN is involved in Gsk-3β-dependent Tau modulation and its proapoptotic effect might be mediated at least in part by the Gsk-3β catalysed Tau hyperphosphorylation and impairment of cytoskeleton stability. GSK-3β inhibitors may offer promising tool against ASN-induced Tau modification and cytotoxicity in neurodegenerative disorders. Supported by statutory theme 9.
BACKGROUND AND AIMS: The prominent features of Alzheimer’s disease (AD) are accumulation of amyloid beta (Aβ) oligomers and neuroinflammatory processes. Previous data demonstrated that activation of cyclin-dependent kinase 5 (Cdk5) may be essential for pathology of AD and other neurodegenerative disorders. In this study we focused on the role of Cdk5 in controlling gene expression in the brain in experimental model of AD as well as during systemic inflammatory reaction (SIR) METHODS: Alzheimer’s Aβ toxicity and SIR were induced in mice by intracerebroventricular injection of Aβ1-42 oligomers and intraperitoneal injection of lipopolysaccharide (LPS), respectively. Roscovitine, the inhibitor of Cdk5, was administered intraperitoneally. RESULTS: Both Aβ and LPS induced an increase in Cdk5 activity in hippocampus, as evidenced by augmented formation of Cdk5 activator, p25, and enhanced phosphorylation of Cdk5. In concordance, Cdk5-related increase in phosphorylation of Gsk-3β (Ser9) and MAP tau (Ser396) was found. Moreover, we found Cdk5-dependent phosphorylation of ERK1/2 (Thr183/Tyr185) and MEF2A (Ser406), that may negatively modulate activity of transcription factors and in consequence the expression of various genes, i.a. those related to prosurvival pathways. Aβ and LPS injection evoked rapid activation of inflammation-related genes in hippocampus, Nos2, Tnfa, Il1b, Il6, Il10, whereas inhibition of Cdk5 with Roscovitine modified the level of micro RNA and augmented Aβ- and LPS-induced changes in mRNA level for several inflammationrelated genes. CONCLUSIONS: Cdk5 participates in regulation of gene expression in Aβ toxicity and in SIR. Our data suggest that modulation of Cdk5 activity may be prospective strategy for protection in neurodegenerative disorders. This study was supported by The National Science Centre grant 2011/03/B/NZ3/04549.
Ceramide and sphingosine-1-phosphate (S1P) are very active sphingolipid messengers which play a crucial role in regulation of neuronal cells survival and death. Alternation of ceramide/S1P rheostat is related to several pathological disorders including Alzheimer’s disease. Ceramides are involved in cells proliferation, differentiation and apoptotic death, while S1P enhances cell proliferation and antagonizes apoptosis. S1P regulates cellular processes by binding to five specific G protein coupled-receptors (S1PR1-5). The aim of the study was to investigate the molecular processes of neuronal death evoked by ceramide and the role of S1P in neuroprotection. Our study indicated that ceramide enhanced significantly the level of free radicals and decreased neuronal cells (SHSY5Y) viability through inhibition of PI3-K/Akt pathway. Ceramide also decreased anti-apoptotic (Bcl-2) and increased pro-apoptotic (Bax, Hrk) gene expression. Exogenously added S1P increased the viability of cells through S1PR (1-3) receptors-dependent mechanism. S1P also increased Bcl-2 gene expression and decreased the gene expression of Hrk protein. Summarizing, our study indicated that the action of ceramide and S1P on mitochondria may control neuronal fate and may play a crucial role in neurodegeneration and neuroprotection.
BACKGROUND AND AIMS: Hyperphosphorylation of Tau is involved in the pathomechanism of neurodegenerative disorders such as Alzheimer’s and Parkinson’s diseases. Epidemiological data suggest the significance of early life exposure to lead (Pb) in etiology of disorders affecting brain function. However, the precise mechanisms by which Pb exerts neurotoxic effects are not fully elucidated. In this study, we investigated the effect of perinatal exposure to Pb on Tau pathology in selected rat brain structures: forebrain cortex (FC), cerebellum (C) and hippocampus (H). Concomitantly, we examined the ultrastructural alterations in these regions. Furthermore, the involvement of two major Tau-kinases: glycogen synthase kinase-3 beta (GSK-3β) and cyclin-dependent kinase 5 (CDK5) in Pb-induced Tau modification was analysed. RESULTS: Our data revealed that pre- and neonatal exposure of rats to Pb (concentration in rat offspring’s blood below a ‘safe level’) evoked significant increase in the phosphorylation of Tau at Ser396 with parallel rise in the level of total Tau protein in FC and C. Moreover, in these brain structures, GSK-3β activity was increased by phosphorylation of a tyrosine residue – Tyr-216. However, GSK3β phosphorylation on serine residue, Ser-9 was unchanged. Parallel with GSK-3β activation we observed increase of total GSK-3β level in FC from rats subjected to Pb. In Pb-treated cerebellum we showed calpain-dependent cleavage of CDK5-activating protein p35 leading to formation of p25 and CDK5 overactivation. Molecular alterations were accompanied by pathological changes in ultrastructure of all examined brain structures from rats subjected to Pb. CONCLUSION: Perinatal exposure to lead induces Tau modification in the rat cortex and cerebellum. We suggest that its neurotoxic effect might be mediated, at least in part, by GSK-3β and CDK5- catalysed Tau hyperphosphorylation, leading to impairment of cytoskeleton stability. Supported from MMRC statutory theme 8.
Pierwsza strona wyników Pięć stron wyników wstecz Poprzednia strona wyników Strona / 1 Następna strona wyników Pięć stron wyników wprzód Ostatnia strona wyników
JavaScript jest wyłączony w Twojej przeglądarce internetowej. Włącz go, a następnie odśwież stronę, aby móc w pełni z niej korzystać.