Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuro Oncol ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38860311

RESUMO

BACKGROUND: Medulloblastoma (MB) is one of the most common malignant brain tumors in children. Current treatments have increased overall survival but can lead to devastating side effects and late complications in survivors, emphasizing the need for new, improved targeted therapies that specifically eliminate tumor cells while sparing the normally developing brain. METHODS: Here, we used a SHH-MB model based on a patient-derived neuroepithelial stem (NES) cell system for an unbiased high-throughput screen with a library of 172 compounds with known targets. Compounds were evaluated in both healthy neural stem cells and tumor cells derived from the same patient. Based on the difference of cell viability and drug sensitivity score between normal cells and tumor cells, hit compounds were selected and further validated in vitro and in vivo. RESULTS: We identified PF4708671 (S6K1 inhibitor) as a potential agent that selectively targets Sonic Hedgehog (SHH) driven MB tumor cells while sparing neural stem cells and differentiated neurons. Subsequent validation studies confirmed that PF4708671 inhibited the growth of SHH-MB tumor cells both in vitro and in vivo, and that knockdown of S6K1 resulted in reduced tumor formation. CONCLUSIONS: Overall, our results suggest that inhibition of S6K1 specifically affects tumor growth, whereas it has less effect on non-tumor cells. Our data also show that the NES cell platform can be used to identify potentially effective new therapies and targets for SHH-MB.

2.
Neuro Oncol ; 25(6): 1058-1072, 2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-36583853

RESUMO

BACKGROUND: High-grade gliomas are malignant brain tumors characterized by aggressiveness and resistance to chemotherapy. Prognosis remains dismal, highlighting the need to identify novel molecular dependencies and targets. Ribosome biogenesis (RiBi), taking place in the nucleolus, represents a promising target as several cancer types rely on high RiBi rates to sustain proliferation. Publicly available transcriptomics data of glioma patients revealed a positive correlation between RiBi rates and histological grades. We, therefore, hypothesized that glioma cells could be susceptible to RiBi inhibition. METHODS: Transcriptomics data from glioma patients were analyzed for RiBi-related processes. BMH-21, a small molecule inhibitor of RNA pol I transcription, was tested in adult and pediatric high-grade glioma cell lines and a zebrafish transplant model. Cellular phenotypes were evaluated by transcriptomics, cell cycle analysis, and viability assays. A chemical synergy screen was performed to identify drugs potentiating BMH-21-mediated effects. RESULTS: BMH-21 reduced glioma cell viability, induced apoptosis, and impaired the growth of transplanted glioma cells in zebrafish. Combining BMH-21 with TMZ potentiated cytotoxic effects. Moreover, BMH-21 synergized with Fibroblast Growth Factor Receptor (FGFR) inhibitor (FGFRi) Erdafitinib, a top hit in the chemical synergy screen. RiBi inhibition using BMH-21, POLR1A siRNA, or Actinomycin D revealed engagement of the FGFR-FGF2 pathway. BMH-21 downregulated FGFR1 and SOX2 levels, whereas FGF2 was induced and released from the nucleolus. CONCLUSIONS: This study conceptualizes the implementation of RiBi inhibition as a viable future therapeutic strategy for glioma and reveals an FGFR connection to the cellular response upon RiBi inhibition with potential translational value.


Assuntos
Glioma , Peixe-Zebra , Animais , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fator 2 de Crescimento de Fibroblastos/uso terapêutico , Linhagem Celular Tumoral , Glioma/genética , Proliferação de Células , Ciclo Celular , Inibidores de Proteínas Quinases/farmacologia , Ribossomos/metabolismo , Ribossomos/patologia
3.
Mol Psychiatry ; 27(8): 3533-3543, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35418601

RESUMO

Liver-generated plasma apolipoprotein E (apoE) does not enter the brain but nonetheless correlates with Alzheimer's disease (AD) risk and AD biomarker levels. Carriers of APOEε4, the strongest genetic AD risk factor, exhibit lower plasma apoE and altered brain integrity already at mid-life versus non-APOEε4 carriers. Whether altered plasma liver-derived apoE or specifically an APOEε4 liver phenotype promotes neurodegeneration is unknown. Here we investigated the brains of Fah-/-, Rag2-/-, Il2rg-/- mice on the Non-Obese Diabetic (NOD) background (FRGN) with humanized-livers of an AD risk-associated APOE ε4/ε4 versus an APOE ε2/ε3 genotype. Reduced endogenous mouse apoE levels in the brains of APOE ε4/ε4 liver mice were accompanied by various changes in markers of synaptic integrity, neuroinflammation and insulin signaling. Plasma apoE4 levels were associated with unfavorable changes in several of the assessed markers. These results propose a previously unexplored role of the liver in the APOEε4-associated risk of neurodegenerative disease.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Animais , Camundongos , Apolipoproteína E4/genética , Camundongos Endogâmicos NOD , Apolipoproteínas E/genética , Encéfalo/metabolismo , Doença de Alzheimer/genética , Genótipo , Biomarcadores , Fígado/metabolismo
4.
Aging Cell ; 17(3): e12728, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29453790

RESUMO

The population of brain pericytes, a cell type important for vessel stability and blood brain barrier function, has recently been shown altered in patients with Alzheimer's disease (AD). The underlying reason for this alteration is not fully understood, but progressive accumulation of the AD characteristic peptide amyloid-beta (Aß) has been suggested as a potential culprit. In the current study, we show reduced number of hippocampal NG2+ pericytes and an association between NG2+ pericyte numbers and Aß1-40 levels in AD patients. We further demonstrate, using in vitro studies, an aggregation-dependent impact of Aß1-40 on human NG2+ pericytes. Fibril-EP Aß1-40 exposure reduced pericyte viability and proliferation and increased caspase 3/7 activity. Monomer Aß1-40 had quite the opposite effect: increased pericyte viability and proliferation and reduced caspase 3/7 activity. Oligomer-EP Aß1-40 had no impact on either of the cellular events. Our findings add to the growing number of studies suggesting a significant impact on pericytes in the brains of AD patients and suggest different aggregation forms of Aß1-40 as potential key regulators of the brain pericyte population size.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Antígenos/metabolismo , Pericitos/metabolismo , Proteoglicanas/metabolismo , Idoso , Idoso de 80 Anos ou mais , Técnicas de Cultura de Células , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
5.
Alzheimers Res Ther ; 10(1): 9, 2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29378650

RESUMO

BACKGROUND: Kallikrein 6 (KLK6) is known to be an age-related protease expressed at high levels in the central nervous system. It was previously shown to be involved in proteolysis of extracellular proteins implicated in neurodegenerative diseases such as Alzheimer's disease (AD), prompting validation of KLK6 as a potential biomarker of disease. However, analyses of both plasma and cerebrospinal fluid (CSF) levels of KLK6 in patients with AD have been inconclusive. We present a detailed analysis of KLK6 in plasma and CSF in two separate cohorts in a cross-sectional and a longitudinal clinical setting. METHODS: The cross-sectional cohort included control subjects without dementia and patients with AD, and the longitudinal cohort included patients with MCI and patients with AD followed over a 2-year period. Plasma and CSF levels of KLK6 were quantified by use of a previously developed and validated enzyme-linked immunosorbent assay. Statistical analyses were performed to compare KLK6 levels between diagnostic groups and to identify potential associations between KLK6 level, age, apolipoprotein E (APOE) genotype, total apoE level and the classical CSF AD biomarkers. RESULTS: In the cross-sectional setting, KLK6 levels in plasma but not in CSF were significantly higher in the AD group than in control subjects. CSF but not plasma KLK6 levels were positively correlated with age in both the cross-sectional and longitudinal settings. In both cohorts, the CSF KLK6 levels were significantly and positively correlated with the CSF levels of core AD biomarkers. Total plasma and CSF apoE levels were positively associated with KLK6 in the cross-sectional study. Finally, during the 2-year monitoring period of the longitudinal cohort, CSF KLK6 levels increased with disease progression over time in the investigated patient groups. CONCLUSIONS: In two separate cohorts we have confirmed the previously reported correlation between age and CSF levels of KLK6. Increased plasma KLK6 levels in patients with AD with a more advanced disease stage suggest KLK6 as a potential biomarker in patients with AD with more severe dementia. Significant correlations between KLK6 levels and core CSF AD biomarkers suggest molecular links between KLK6 and AD-related pathological processes.


Assuntos
Doença de Alzheimer/sangue , Doença de Alzheimer/líquido cefalorraquidiano , Calicreínas/sangue , Calicreínas/líquido cefalorraquidiano , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Apolipoproteína E4/genética , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Estudos Transversais , Feminino , Humanos , Estudos Longitudinais , Masculino , Testes de Estado Mental e Demência , Pessoa de Meia-Idade
6.
Front Neurosci ; 11: 172, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28424577

RESUMO

Misfolding and aggregation of alpha-synuclein (αsyn) resulting in cytotoxicity is a hallmark of Parkinson's disease (PD) and related synucleinopathies. The recent body of evidence indicates that αsyn can be released from neuronal cells by nonconventional exocytosis involving extracellular vesicles (EVs) such as exosomes. The transfer of αsyn between cells has been proposed to be an important mechanism of disease propagation in PD. To date, exosome trafficking mechanisms, including release and cell-cell transmission, have not been fully described. To gain insight into the mechanisms involved, exosomes were purified from conditioned media of stable cells secreting αsyn oligomers. A novel bimolecular protein complementation assay was used to detect exosomes containing αsyn oligomers. Recipient cells were treated with exosomes containing αsyn oligomers or "free" non-exosome-associated αsyn oligomers and internalization was monitored. We demonstrate that cell-derived exosome-associated αsyn oligomers can be efficiently internalized by recipient cells. Interestingly exosome-free αsyn oligomers isolated from conditioned medium were not internalized but remained bound to the extracellular surface. To investigate the endocytic pathway(s) required for the exosome uptake different pharmacological inhibitors of caveolin-dependent, clathrin-dependent, and macropinocytosis pathways were utilized. Surprisingly, none of these pathways appear to play a significant role in the internalization of exosome-associated αsyn oligomers. Finally, the role of heparin sulfate proteoglycans (HSPGs) in exosome-associated αsyn internalization was investigated using genetic approach. Despite previous studies showing HSPGs can modulate internalization of fibrillar αsyn, genetic manipulations did not attenuate internalization of exosome-associated αsyn oligomers in our hands, suggesting that exosome-associated αsyn is internalized via an alternative endocytic pathway(s) that has yet to be elucidated.

7.
Acta Neuropathol ; 132(1): 77-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26839082

RESUMO

Cell-to-cell transmission of α-synuclein (αS) aggregates has been proposed to be responsible for progressive αS pathology in Parkinson disease (PD) and related disorders, including dementia with Lewy bodies. In support of this concept, a growing body of in vitro and in vivo experimental evidence shows that exogenously introduced αS aggregates can spread into surrounding cells and trigger PD-like pathology. It remains to be determined what factor(s) lead to initiation of αS aggregation that is capable of seeding subsequent propagation. In this study we demonstrate that filamentous αS aggregates form in neurons in response to apoptosis induced by staurosporine or other toxins-6-hydroxy-dopamine and 1-methyl-4-phenylpyridinium (MPP+). Interaction between αS and proaggregant nuclear factor(s) is associated with disruption of nuclear envelope integrity. Knocking down a key nuclear envelop constituent protein, lamin B1, enhances αS aggregation. Moreover, in vitro and in vivo experimental models demonstrate that aggregates released upon cell breakdown can be taken up by surrounding cells. Accordingly, we suggest that at least some αS aggregation might be related to neuronal apoptosis or loss of nuclear membrane integrity, exposing cytosolic α-synuclein to proaggregant nuclear factors. These findings provide new clues to the pathogenesis of PD and related disorders that can lead to novel treatments of these disorders. Specifically, finding ways to limit the effects of apoptosis on αS aggregation, deposition, local uptake and subsequent propagation might significantly impact progression of disease.


Assuntos
Apoptose/fisiologia , Lamina Tipo B/metabolismo , Neurônios/metabolismo , Membrana Nuclear/metabolismo , Agregação Patológica de Proteínas/metabolismo , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Tumoral , Citosol/efeitos dos fármacos , Citosol/metabolismo , Citosol/patologia , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/patologia , Membrana Nuclear/efeitos dos fármacos , Membrana Nuclear/patologia , Oxidopamina/toxicidade , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Agregação Patológica de Proteínas/patologia , Estaurosporina/toxicidade
9.
Expert Opin Ther Targets ; 19(5): 589-603, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25785645

RESUMO

OBJECTIVE: Reducing the burden of α-synuclein oligomeric species represents a promising approach for disease-modifying therapies against synucleinopathies such as Parkinson's disease and dementia with Lewy bodies. However, the lack of efficient drug discovery strategies that specifically target α-synuclein oligomers has been a limitation to drug discovery programs. RESEARCH DESIGN AND METHODS: Here we describe an innovative strategy that harnesses the power of bimolecular protein-fragment complementation to monitor synuclein-synuclein interactions. We have developed two robust models to monitor α-synuclein oligomerization by generating novel stable cell lines expressing α-synuclein fusion proteins for either fluorescent or bioluminescent protein-fragment complementation under the tetracycline-controlled transcriptional activation system. MAIN OUTCOME MEASURES: A pilot screen was performed resulting in the identification of two potential hits, a p38 MAPK inhibitor and a casein kinase 2 inhibitor, thereby demonstrating the suitability of our protein-fragment complementation assay for the measurement of α-synuclein oligomerization in living cells at high throughput. CONCLUSIONS: The application of the strategy described herein to monitor α-synuclein oligomer formation in living cells with high throughput will facilitate drug discovery efforts for disease-modifying therapies against synucleinopathies and other proteinopathies.


Assuntos
Descoberta de Drogas/métodos , Doença por Corpos de Lewy/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico , alfa-Sinucleína/metabolismo , Caseína Quinase II/antagonistas & inibidores , Linhagem Celular , Desenho de Fármacos , Ensaios de Triagem em Larga Escala , Humanos , Doença por Corpos de Lewy/fisiopatologia , Modelos Biológicos , Terapia de Alvo Molecular , Doença de Parkinson/fisiopatologia , Projetos Piloto , Multimerização Proteica , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
10.
Front Neurosci ; 9: 511, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26834539

RESUMO

Alpha synuclein (αsyn) aggregates are associated with the pathogenesis of Parkinson's disease and others related disorders. Although modulation of αsyn aggregation is an attractive therapeutic target, new powerful methodologies are desperately needed to facilitate in vivo screening of novel therapeutics. Here, we describe an in vivo rodent model with the unique ability to rapidly track αsyn-αsyn interactions and thus oligomerization using a bioluminescent protein complementation strategy that monitors spatial and temporal αsyn oligomerization ex vivo. We find that αsyn forms oligomers in vivo as early as 1 week after stereotactic AAV injection into rat substantia nigra. Strikingly, although abundant αsyn expression is also detected in striatum at 1 week, no αsyn oligomers are detected at this time point. By 4 weeks, oligomerization of αsyn is detected in both striatum and substantia nigra homogenates. Moreover, in a proof-of-principle experiment, the effect of a previously described Hsp90 inhibitor known to prevent αsyn oligomer formation, demonstrates the utility of this rapid and sensitive animal model to monitor αsyn oligomerization status in the rat brain.

11.
Neurobiol Aging ; 36(2): 1209-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25480524

RESUMO

The pathologic hallmark of Parkinson's disease (PD) is the accumulation of alpha-synuclein (αsyn) in susceptible neurons in the form of Lewy bodies and Lewy neurites. The etiology of PD remains unclear. Because brain injury has been suggested to facilitate αsyn aggregation, we investigated whether cellular breakdown products from damaged cells can act on neighboring healthy cells and cause intracellular αsyn accumulation and/or aggregation. Using 2 neuronal cell models, we found that extracellular adenosine triphosphate (ATP) induced a significant increase in intracellular αsyn levels between 24 and 48 hours after treatment. Further investigation revealed that the observed αsyn accumulation is a result of lysosome dysfunction caused by extracellular ATP-induced elevation of lysosomal pH. Interestingly, P2X1 receptor appears to mediate the cells' response to extracellular ATP. Although Ca(2+) influx via P2X1 receptor is necessary for αsyn accumulation, Ca(2+) influx per se is not sufficient for increased αsyn accumulation. These findings provide new insight into our knowledge of the role of P2X receptors in PD pathogenesis and may be helpful in identifying new therapeutic targets for PD.


Assuntos
Trifosfato de Adenosina/farmacologia , Lisossomos/metabolismo , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Receptores Purinérgicos P2X1/fisiologia , alfa-Sinucleína/metabolismo , Trifosfato de Adenosina/fisiologia , Cálcio/metabolismo , Células Cultivadas , Humanos , Concentração de Íons de Hidrogênio , Neurônios/metabolismo , Doença de Parkinson/genética , Fatores de Tempo
12.
Mol Neurodegener ; 9: 43, 2014 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-25352339

RESUMO

The accumulation of α-synuclein aggregates is the hallmark of Parkinson's disease, and more generally of synucleinopathies. The accumulation of tau aggregates however is classically found in the brains of patients with dementia, and this type of neuropathological feature specifically defines the tauopathies. Nevertheless, in numerous cases α-synuclein positive inclusions are also described in tauopathies and vice versa, suggesting a co-existence or crosstalk of these proteinopathies. Interestingly, α-synuclein and tau share striking common characteristics suggesting that they may work in concord. Tau and α-synuclein are both partially unfolded proteins that can form toxic oligomers and abnormal intracellular aggregates under pathological conditions. Furthermore, mutations in either are responsible for severe dominant familial neurodegeneration. Moreover, tau and α-synuclein appear to promote the fibrillization and solubility of each other in vitro and in vivo. This suggests that interactions between tau and α-synuclein form a deleterious feed-forward loop essential for the development and spreading of neurodegeneration. Here, we review the recent literature with respect to elucidating the possible links between α-synuclein and tau.


Assuntos
Degeneração Neural/metabolismo , Doenças Neurodegenerativas/metabolismo , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Animais , Humanos , Degeneração Neural/patologia , Doenças Neurodegenerativas/patologia
13.
Brain ; 137(Pt 7): 1958-70, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24860142

RESUMO

A growing number of PSEN1 mutations have been associated with dementia with Lewy bodies and familial Alzheimer's disease with concomitant α-synuclein pathology. The objective of this study was to determine if PSEN1 plays a direct role in the development of α-synuclein pathology in these diseases. Using mass spectrometry, immunoelectron microscopy and fluorescence lifetime image microscopy based on Forster resonance energy transfer (FLIM-FRET) we identified α-synuclein as a novel interactor of PSEN1 in wild-type mouse brain tissue. The interaction of α-synuclein with PSEN1 was detected in post-mortem brain tissue from cognitively normal cases and was significantly increased in tissue from cases with dementia with Lewy bodies and familial Alzheimer's disease associated with known PSEN1 mutations. We confirmed an increased interaction of PSEN1 and α-synuclein in cell lines expressing well characterized familial Alzheimer's disease PSEN1 mutations, L166P and delta exon 9, and demonstrated that PSEN1 mutations associate with increased membrane association and accumulation of α-synuclein. Our data provides evidence of a molecular interaction of PSEN1 and α-synuclein that may explain the clinical and pathophysiological overlap seen in synucleinopathies, including Parkinson's disease, dementia with Lewy bodies, and some forms of Alzheimer's disease.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Doença por Corpos de Lewy/patologia , Presenilina-1/metabolismo , alfa-Sinucleína/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Encéfalo/patologia , Encéfalo/ultraestrutura , Células CHO , Células Cultivadas , Córtex Cerebral , Cricetulus , Feminino , Glutationa Transferase/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Microscopia Imunoeletrônica , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Presenilina-1/deficiência , Presenilina-1/genética
14.
Ther Adv Neurol Disord ; 7(1): 33-51, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24409201

RESUMO

Parkinson's disease is a slowly progressive neurodegenerative disorder typically characterized by the loss of dopaminergic neurons within the substantia nigra pars compacta, and the intraneuronal deposition of insoluble protein aggregates chiefly comprised of α-synuclein. Patients experience debilitating symptoms including bradykinesia, rigidity and postural instability. No curative treatment currently exists and therapeutic strategies are restricted to symptomatic treatment only. Over the past decade a class of molecular chaperones called the heat shock proteins has emerged as a potentially promising therapeutic target. Heat shock proteins aid in the folding and refolding of proteins, and target denatured proteins to degradation systems. By targeting heat shock proteins through various means including overexpression and pharmacological enhancement, researchers have shown that α-synuclein aggregation and its associated cytotoxicity can be therapeutically modulated in an array of cell and animal models. This review highlights the relevant progress in this field and discusses the relevance of heat shock proteins as therapeutic modulators of α-synuclein toxicity to the rapidly evolving understanding of Parkinson's disease pathogenesis.

15.
J Neurosci Methods ; 187(2): 243-53, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-20097228

RESUMO

As the major immuno-competent cells of the brain, microglia are highly implicated in neuro-protection as well as in neurodegeneration. Therefore, they are of key interest for research on numerous CNS diseases. Currently, to model inflammation in the brain, microglial cell lines or primary microglia prepared from embryonic or neo-natal rodents are widely used. However, these in vitro microglial models are not suitable for research in the field of neuro-degenerative diseases where aging is a crucial parameter. Only a few in vitro studies on aged microglia have been published so far, most of which use ex vivo microglia which cannot be kept in culture for prolonged periods of time. In the present study, we provide a new approach which allows the isolation and culture of an almost pure population of microglia from adult mouse brains. The isolation is based on a procedure which combines density separation and a subsequent culture selection process. After these steps, microglia form a non-adherent floating cell layer that can be easily and repeatedly harvested and replated. This method is simple and allows for a comparatively high yield and purity of adult microglial cells. The collected primary adult microglia proliferate and can be kept in culture for extended periods of time. We compared the primary adult microglia to primary microglia from neo-natal mice as well as to the C8-B4 microglial cell line. We found that adult microglia have similar, but not identical, immuno-phenotypic, functional and electrophysiological characteristics to the other in vitro models.


Assuntos
Separação Celular/métodos , Microglia/fisiologia , Animais , Animais Recém-Nascidos , Linhagem Celular , Membrana Celular/metabolismo , Células Cultivadas , Citocinas/biossíntese , Eletrofisiologia , Feminino , Citometria de Fluxo , Imunofluorescência , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Óxido Nítrico/biossíntese , Fagocitose/fisiologia , Canais de Potássio/fisiologia
16.
Cell Physiol Biochem ; 24(3-4): 141-52, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19710528

RESUMO

Microglia are the intrinsic immune cells of the brain. As such, they are crucially involved in neuro-protection as well as neuro-degeneration. Their activation leads to the induction of cytokine and chemokine release, the production of reactive oxygen species and nitric oxide and an increased outward potassium conductance. In this study, we focus our interest on potassium currents and channels in the C8-B4 murine microglial cell line and compare them with those of primary cultured microglia from neo-natal mice. Using the whole cell patch-clamp technique, we have recorded prominent inward and outward rectifying voltage-dependent potassium currents but no calcium-activated potassium currents. Using pharmacological, biophysical and molecular approaches, we demonstrate that Kv1.3 and Kir2.1 channels underlie outward and inward rectifying potassium currents, respectively. In contrast to primary cultured microglia, we observe that an outward rectifying potassium current is already present in unstimulated C8-B4 cells. However, as seen in primary microglia, this current increases after treatment with LPS, IFN-gamma, TGF-beta and GM-CSF and is suppressed by treatment with protein kinase inhibitors. Our study indicates that the C8-B4 cell line shows similar though not identical potassium channel properties compared to primary cultured microglia. We demonstrate that despite some differences, they are a useful tool to study potassium currents in microglial activation mechanisms by means of electrophysiological methods without the need for preparation of cells as primary culture.


Assuntos
Microglia/fisiologia , Canais de Potássio/fisiologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , 4-Aminopiridina/farmacologia , Animais , Apamina/farmacologia , Bário/farmacologia , Cálcio/farmacologia , Cátions/farmacologia , Linhagem Celular , Charibdotoxina/farmacologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Venenos Elapídicos/farmacologia , Eletrodos , Eletrofisiologia , Técnica Direta de Fluorescência para Anticorpo , Genisteína/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interferon gama/farmacologia , Lipopolissacarídeos/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/metabolismo , Neurotoxinas/farmacologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização , Pirazóis/farmacologia , Venenos de Escorpião/farmacologia , Fator de Crescimento Transformador beta/farmacologia
17.
J Neurosci ; 27(34): 9220-32, 2007 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-17715357

RESUMO

Aggregation of alpha-synuclein (alpha-syn) has been linked to the pathogenesis of Parkinson's disease (PD) and other neurodegenerative diseases. Increasing evidence suggests that prefibrillar oligomers and protofibrils, rather than mature fibrils of alpha-syn, are the pathogenic species in PD. Despite extensive effort on studying oligomerization of alpha-syn, no studies have compared different oligomer species directly on a single-particle level and investigated their biological effects on cells. In this study, we applied a novel highly sensitive single molecule detection system that allowed a direct comparison of different oligomer types. Furthermore, we studied biological effects of different oligomer types on cells. For this purpose, we developed new oligomerization protocols, that enabled the use of these different oligomers in cell culture. We found that all of our three aggregation protocols resulted in heterogeneous populations of oligomers. Some types of oligomers induced cell death via disruption of cellular ion homeostasis by a presumably pore-forming mechanism. Other oligomer types could directly enter the cell resulting in increased alpha-syn aggregation. Based on our results, we propose that under various physiological conditions, heterogeneous populations of oligomeric forms will coexist in an equilibrium. These different oligomer types lead directly or indirectly to cell damage. Our data indicate that inhibition of early alpha-syn aggregation events would consequently prevent all alpha-syn oligomer related toxicities. This has important implications for the development of disease-modifying drugs for the treatment of PD and other synucleinopathies.


Assuntos
Cálcio/metabolismo , Neurônios/efeitos dos fármacos , alfa-Sinucleína/química , alfa-Sinucleína/farmacologia , Animais , Biopolímeros , Encéfalo/citologia , Caspase 3 , Células Cultivadas , Embrião de Mamíferos , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Microscopia de Força Atômica/métodos , Mutação/fisiologia , Neuroblastoma , Fragmentos de Peptídeos/química , Proteínas Recombinantes , Transfecção/métodos , alfa-Sinucleína/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...