Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Biochemistry ; 60(37): 2773-2780, 2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34469142

RESUMO

The prevailing opinion is that prefibrillar ß-amyloid (Aß) species, rather than end-stage amyloid fibrils, cause neuronal dysfunction in Alzheimer's disease, although the mechanisms behind Aß neurotoxicity remain to be elucidated. Luminescent conjugated oligothiophenes (LCOs) exhibit spectral properties upon binding to amyloid proteins and have previously been reported to change the toxicity of Aß1-42 and prion protein. In a previous study, we showed that an LCO, pentamer formyl thiophene acetic acid (p-FTAA), changed the toxicity of Aß1-42. Here we investigated whether an LCO, heptamer formyl thiophene acetic acid (h-FTAA), could change the toxicity of Aß1-42 by comparing its behavior with that of p-FTAA. Moreover, we investigated the effects on toxicity when Aß with the Arctic mutation (AßArc) was aggregated with both LCOs. Cell viability assays on SH-SY5Y neuroblastoma cells demonstrated that h-FTAA has a stronger impact on Aß1-42 toxicity than does p-FTAA. Interestingly, h-FTAA, but not p-FTAA, rescued the AßArc-mediated toxicity. Aggregation kinetics and binding assay experiments with Aß1-42 and AßArc when aggregated with both LCOs showed that h-FTAA and p-FTAA either interact with different species or affect the aggregation in different ways. In conclusion, h-FTAA protects against Aß1-42 and AßArc toxicity, thus showing h-FTAA to be a useful tool for improving our understanding of the process of Aß aggregation linked to cytotoxicity.


Assuntos
Acetatos/química , Precursor de Proteína beta-Amiloide/metabolismo , Tiofenos/química , Acetatos/metabolismo , Amiloide/química , Peptídeos beta-Amiloides/química , Precursor de Proteína beta-Amiloide/fisiologia , Precursor de Proteína beta-Amiloide/toxicidade , Proteínas Amiloidogênicas/química , Corantes Fluorescentes/química , Humanos , Cinética , Luminescência , Fragmentos de Peptídeos/metabolismo , Agregados Proteicos/efeitos dos fármacos , Agregados Proteicos/fisiologia , Coloração e Rotulagem/métodos , Tiofenos/metabolismo
2.
Biochim Biophys Acta Mol Basis Dis ; 1864(9 Pt B): 3060-3068, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29960040

RESUMO

Parkinson's disease (PD) and other synucleinopathies are characterized by accumulation of misfolded aggregates of α-synuclein (α-syn). The normal function of α-syn is still under investigation, but it has been generally linked to synaptic plasticity, neurotransmitter release and the maintenance of the synaptic pool. α-Syn localizes at synaptic terminals where it can bind to synaptic vesicles as well as to other cellular membranes. It has become clear that these interactions have an impact on both α-syn functional role and its propensity to aggregate. In this study, we investigated the aggregation process of α-syn covalently modified with 4-hydroxy-2-nonenal (HNE). HNE is a product of lipid peroxidation and has been implicated in the pathogenesis of different neurodegenerative diseases by modifying the kinetics of soluble toxic oligomers. Although HNE-modified α-syn has been reported to assemble into stable oligomers, we found that slightly acidic conditions promoted further protein aggregation. Lipid vesicles delayed the aggregation process in a concentration-dependent manner, an effect that was observed only when they were added at the beginning of the aggregation process. Co-aggregation of lipid vesicles with HNE-modified α-syn also induced cytotoxic effects on differentiated SHSY-5Y cells. Under conditions in which the aggregation process was delayed cell viability was reduced. By exploring the behavior and potential cytotoxic effects of HNE-α-syn under acidic conditions in relation to protein-lipid interactions our study gives a framework to examine a possible pathway leading from a physiological setting to the pathological outcome of PD.


Assuntos
Aldeídos/metabolismo , Doença de Parkinson/patologia , Agregação Patológica de Proteínas/patologia , Multimerização Proteica/fisiologia , alfa-Sinucleína/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Metabolismo dos Lipídeos/fisiologia , Peroxidação de Lipídeos , Lipossomos/farmacologia , Microscopia Eletrônica de Transmissão , Estresse Oxidativo , Agregação Patológica de Proteínas/tratamento farmacológico , Multimerização Proteica/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Vesículas Sinápticas/patologia , alfa-Sinucleína/ultraestrutura
3.
Acta Neuropathol ; 136(1): 41-56, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29934873

RESUMO

The gradual deterioration of cognitive functions in Alzheimer's disease is paralleled by a hierarchical progression of amyloid-beta and tau brain pathology. Recent findings indicate that toxic oligomers of amyloid-beta may cause propagation of pathology in a prion-like manner, although the underlying mechanisms are incompletely understood. Here we show that small extracellular vesicles, exosomes, from Alzheimer patients' brains contain increased levels of amyloid-beta oligomers and can act as vehicles for the neuron-to-neuron transfer of such toxic species in recipient neurons in culture. Moreover, blocking the formation, secretion or uptake of exosomes was found to reduce both the spread of oligomers and the related toxicity. Taken together, our results imply that exosomes are centrally involved in Alzheimer's disease and that they could serve as targets for development of new diagnostic and therapeutic principles.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Exossomos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Fragmentos de Peptídeos/toxicidade , Idoso , Idoso de 80 Anos ou mais , Peptídeos beta-Amiloides/toxicidade , Linhagem Celular Transformada , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Exossomos/metabolismo , Exossomos/ultraestrutura , Feminino , Regulação da Expressão Gênica/genética , Humanos , L-Lactato Desidrogenase/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Compostos Orgânicos/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
J Biol Chem ; 291(17): 9233-43, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26907684

RESUMO

Aggregation of the amyloid-ß peptide (Aß) in the brain leads to the formation of extracellular amyloid plaques, which is one of the pathological hallmarks of Alzheimer disease (AD). It is a general hypothesis that soluble prefibrillar assemblies of the Aß peptide, rather than mature amyloid fibrils, cause neuronal dysfunction and memory impairment in AD. Thus, reducing the level of these prefibrillar species by using molecules that can interfere with the Aß fibrillation pathway may be a valid approach to reduce Aß cytotoxicity. Luminescent-conjugated oligothiophenes (LCOs) have amyloid binding properties and spectral properties that differ when they bind to protein aggregates with different morphologies and can therefore be used to visualize protein aggregates. In this study, cell toxicity experiments and biophysical studies demonstrated that the LCO p-FTAA was able to reduce the pool of soluble toxic Aß species in favor of the formation of larger insoluble nontoxic amyloid fibrils, there by counteracting Aß-mediated cytotoxicity. Moreover, p-FTAA bound to early formed Aß species and induced a rapid formation of ß-sheet structures. These p-FTAA generated amyloid fibrils were less hydrophobic and more resistant to proteolysis by proteinase K. In summary, our data show that p-FTAA promoted the formation of insoluble and stable Aß species that were nontoxic which indicates that p-FTAA might have therapeutic potential.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/metabolismo , Tiofenos/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Linhagem Celular Tumoral , Humanos , Agregação Patológica de Proteínas/tratamento farmacológico , Agregação Patológica de Proteínas/patologia , Estabilidade Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína
5.
Cell Microbiol ; 17(1): 131-45, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25154738

RESUMO

An overproduction of reactive oxygen species (ROS) mediated by NADPH oxidase 2 (NOX2) has been related to airway inflammation typical of influenza infection. Virus-induced oxidative stress may also control viral replication, but the mechanisms underlying ROS production, as well as their role in activating intracellular pathways and specific steps of viral life cycle under redox control have to be fully elucidated. In this study, we demonstrate that influenza A virus infection of lung epithelial cells causes a significant ROS increase that depends mainly on NOX4, which is upregulated at both mRNA and protein levels, while the expression of NOX2, the primary source of ROS in inflammatory cells, is downregulated. Inhibition of NOX4 activity through chemical inhibitors or RNA silencing blocks the ROS increase, prevents MAPK phosphorylation, and inhibits viral ribonucleoprotein (vRNP) nuclear export and viral release. Overall these data, obtained in cell lines and primary culture, describe a so far unrecognized role for NOX4-derived ROS in activating redox-regulated intracellular pathways during influenza virus infection and highlight their relevance in controlling specific steps of viral replication in epithelial cells. Pharmacological modulation of NOX4-mediated ROS production may open the way for new therapeutic approaches to fighting influenza by targeting cell and not the virus.


Assuntos
Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Vírus da Influenza A/fisiologia , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Replicação Viral , Animais , Células Cultivadas , Células Epiteliais/enzimologia , Expressão Gênica , Humanos , Camundongos , NADPH Oxidase 4 , Oxirredução , Regulação para Cima
6.
Neurobiol Dis ; 83: 122-33, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26334479

RESUMO

The hallmarks of Alzheimer disease are amyloid-ß plaques and neurofibrillary tangles accompanied by signs of neuroinflammation. Lysozyme is a major player in the innate immune system and has recently been shown to prevent the aggregation of amyloid-ß1-40 in vitro. In this study we found that patients with Alzheimer disease have increased lysozyme levels in the cerebrospinal fluid and lysozyme co-localized with amyloid-ß in plaques. In Drosophila neuronal co-expression of lysozyme and amyloid-ß1-42 reduced the formation of soluble and insoluble amyloid-ß species, prolonged survival and improved the activity of amyloid-ß1-42 transgenic flies. This suggests that lysozyme levels rise in Alzheimer disease as a compensatory response to amyloid-ß increases and aggregation. In support of this, in vitro aggregation assays revealed that lysozyme associates with amyloid-ß1-42 and alters its aggregation pathway to counteract the formation of toxic amyloid-ß species. Overall, these studies establish a protective role for lysozyme against amyloid-ß associated toxicities and identify increased lysozyme in patients with Alzheimer disease. Therefore, lysozyme has potential as a new biomarker as well as a therapeutic target for Alzheimer disease.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Muramidase/metabolismo , Fragmentos de Peptídeos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Animais , Encéfalo/patologia , Morte Celular , Drosophila melanogaster , Feminino , Humanos , Proteínas de Insetos/metabolismo , Locomoção , Masculino , Pessoa de Meia-Idade , Muramidase/sangue , Muramidase/líquido cefalorraquidiano , Muramidase/farmacologia , Fragmentos de Peptídeos/ultraestrutura , Placa Amiloide/metabolismo , Placa Amiloide/ultraestrutura , Células Tumorais Cultivadas , Proteínas tau/metabolismo
7.
J Neurovirol ; 21(5): 480-90, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25925093

RESUMO

Several data indicate that neuronal infection with herpes simplex virus type 1 (HSV-1) causes biochemical alterations reminiscent of Alzheimer's disease (AD) phenotype. They include accumulation of amyloid-ß (Aß), which originates from the cleavage of amyloid precursor protein (APP), and hyperphosphorylation of tau protein, which leads to neurofibrillary tangle deposition. HSV-1 infection triggers APP processing and drives the production of several fragments including APP intracellular domain (AICD) that exerts transactivating properties. Herein, we analyzed the production and intracellular localization of AICD following HSV-1 infection in neurons. We also checked whether AICD induced the transcription of two target genes, neprilysin (nep) and glycogen synthase kinase 3ß (gsk3ß), whose products play a role in Aß clearance and tau phosphorylation, respectively. Our data indicate that HSV-1 led to the accumulation and nuclear translocation of AICD in neurons. Moreover, results from chromatin immunoprecipitation assay showed that AICD binds the promoter region of both nep and gsk3ß. Time course analysis of NEP and GSK3ß expression at both mRNA and protein levels demonstrated that they are differently modulated during infection. NEP expression and enzymatic activity were initially stimulated but, with the progression of infection, they were down-regulated. In contrast, GSK3ß expression remained nearly unchanged, but the analysis of its phosphorylation suggests that it was inactivated only at later stages of HSV-1 infection. Thus, our data demonstrate that HSV-1 infection induces early upstream events in the cell that may eventually lead to Aß deposition and tau hyperphosphorylation and further suggest HSV-1 as a possible risk factor for AD.


Assuntos
Doença de Alzheimer/virologia , Precursor de Proteína beta-Amiloide/metabolismo , Herpes Simples/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Doença de Alzheimer/metabolismo , Animais , Western Blotting , Células Cultivadas , Córtex Cerebral , Imunoprecipitação da Cromatina , Quinase 3 da Glicogênio Sintase/biossíntese , Glicogênio Sintase Quinase 3 beta , Herpesvirus Humano 1 , Imuno-Histoquímica , Imunoprecipitação , Neprilisina/biossíntese , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
8.
Adv Healthc Mater ; 11(11): e2102097, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35114074

RESUMO

Laminins (LNs) are key components in the extracellular matrix of neuronal tissues in the developing brain and neural stem cell niches. LN-presenting hydrogels can provide a biologically relevant matrix for the 3D culture of neurons toward development of advanced tissue models and cell-based therapies for the treatment of neurological disorders. Biologically derived hydrogels are rich in fragmented LN and are poorly defined concerning composition, which hampers clinical translation. Engineered hydrogels require elaborate and often cytotoxic chemistries for cross-linking and LN conjugation and provide limited possibilities to tailor the properties of the materials. Here a modular hydrogel system for neural 3D cell cultures, based on hyaluronan and poly(ethylene glycol), that is cross-linked and functionalized with human recombinant LN-521 using bioorthogonal copper-free click chemistry, is shown. Encapsulated human neuroblastoma cells demonstrate high viability and grow into spheroids. Long-term neuroepithelial stem cells (lt-NES) cultured in the hydrogels can undergo spontaneous differentiation to neural fate and demonstrate significantly higher viability than cells cultured without LN. The hydrogels further support the structural integrity of 3D bioprinted structures and maintain high viability of bioprinted and syringe extruded lt-NES, which can facilitate biofabrication and development of cell-based therapies.


Assuntos
Bioimpressão , Hidrogéis , Técnicas de Cultura de Células , Humanos , Ácido Hialurônico , Hidrogéis/química , Hidrogéis/farmacologia , Laminina/farmacologia , Neurônios , Engenharia Tecidual
9.
Front Mol Neurosci ; 12: 200, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31555088

RESUMO

Recently, extracellular vesicles (EVs), such as exosomes, have been proposed to play an influential role in the cell-to-cell spread of neurodegenerative diseases, including the intercellular transmission of α-synuclein (α-syn). However, the regulation of EV biogenesis and its relation to Parkinson's disease (PD) is only partially understood. The generation of EVs through the ESCRT-independent pathway depends on the hydrolysis of sphingomyelin by neutral sphingomyelinase 2 (nSMase2) to produce ceramide, which causes the membrane of endosomal multivesicular bodies to bud inward. nSMase2 is sensitive to oxidative stress, a common process in PD brains; however, little is known about the role of sphingomyelin metabolism in the pathogenesis of PD. This is the first study to show that inhibiting nSMase2 decreases the transfer of oligomeric aggregates of α-syn between neuron-like cells. Furthermore, it reduced the accumulation and aggregation of high-molecular-weight α-syn. Hypoxia, as a model of oxidative stress, reduced the levels of nSMase2, but not its enzymatic activity, and significantly altered the lipid composition of cells without affecting EV abundance or the transfer of α-syn. These data show that altering sphingolipids can mitigate the spread of α-syn, even under hypoxic conditions, potentially suppressing PD progression.

10.
Phytomedicine ; 21(6): 857-65, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24629600

RESUMO

Several essential oils exert in vitro activity against bacteria and viruses and, among these latter, herpes simplex virus type 1 (HSV-1) is known to develop resistance to commonly used antiviral agents. Thus, the effects of the essential oil derived from Mentha suaveolens (EOMS) and its active principle piperitenone oxide (PEO) were tested in in vitro experimental model of infection with HSV-1. The 50% inhibitory concentration (IC50) was determined at 5.1µg/ml and 1.4µg/ml for EOMS and PEO, respectively. Australian tea tree oil (TTO) was used as control, revealing an IC50 of 13.2µg/ml. Moreover, a synergistic action against HSV-1 was observed when each oil was added in combination with acyclovir. In order to find out the mechanism of action, EOMS, PEO and TTO were added to the cells at different times during the virus life-cycle. Results obtained by yield reduction assay indicated that the antiviral activity of both compounds was principally due to an effect after viral adsorption. Indeed, no reduction of virus yield was observed when cells were treated during viral adsorption or pre-treated before viral infection. In particular, PEO exerted a strong inhibitory effect by interfering with a late step of HSV-1 life-cycle. HSV-1 infection is known to induce a pro-oxidative state with depletion of the main intracellular antioxidant glutathione and this redox change in the cell is important for viral replication. Interestingly, the treatment with PEO corrected this deficit, thus suggesting that the compound could interfere with some redox-sensitive cellular pathways exploited for viral replication. Overall our data suggest that both EOMS and PEO could be considered good candidates for novel anti-HSV-1 strategies, and need further exploration to better characterize the targets underlying their inhibition.


Assuntos
Antivirais/farmacologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Mentha/química , Monoterpenos/farmacologia , Óleos Voláteis/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Chlorocebus aethiops , Infecções por Herpesviridae/tratamento farmacológico , Herpesvirus Humano 1/fisiologia , Técnicas In Vitro , Concentração Inibidora 50 , Masculino , Óleos Voláteis/química , Fitoterapia , Extratos Vegetais/farmacologia , Óleo de Melaleuca/farmacologia , Células Vero
11.
Cell Cycle ; 13(15): 2469-74, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483196

RESUMO

The stability of mammalian telomeres depends upon TRF2, which prevents inappropriate repair and checkpoint activation. By using a plasmid integration assay in yeasts carrying humanized telomeres, we demonstrated that TRF2 possesses the intrinsic property to both stimulate initial homologous recombination events and to prevent their resolution via its basic N-terminal domain. In human cells, we further showed that this TRF2 domain prevents telomere shortening mediated by the resolvase-associated protein SLX4 as well as GEN1 and MUS81, 2 different types of endonucleases with resolvase activities. We propose that various types of resolvase activities are kept in check by the basic N-terminal domain of TRF2 in order to favor an accurate repair of the stalled forks that occur during telomere replication.


Assuntos
Recombinação Genética , Telômero/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endonucleases/metabolismo , Células HEK293 , Resolvases de Junção Holliday/metabolismo , Humanos , Plasmídeos , Recombinases/metabolismo , Homeostase do Telômero , Proteína 2 de Ligação a Repetições Teloméricas/genética , Transfecção , Proteína Supressora de Tumor p53/metabolismo
12.
Mol Neurobiol ; 46(3): 614-38, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22899188

RESUMO

A growing body of epidemiologic and experimental data point to chronic bacterial and viral infections as possible risk factors for neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and amyotrophic lateral sclerosis. Infections of the central nervous system, especially those characterized by a chronic progressive course, may produce multiple damage in infected and neighbouring cells. The activation of inflammatory processes and host immune responses cause chronic damage resulting in alterations of neuronal function and viability, but different pathogens can also directly trigger neurotoxic pathways. Indeed, viral and microbial agents have been reported to produce molecular hallmarks of neurodegeneration, such as the production and deposit of misfolded protein aggregates, oxidative stress, deficient autophagic processes, synaptopathies and neuronal death. These effects may act in synergy with other recognized risk factors, such as aging, concomitant metabolic diseases and the host's specific genetic signature. This review will focus on the contribution given to neurodegeneration by herpes simplex type-1, human immunodeficiency and influenza viruses, and by Chlamydia pneumoniae.


Assuntos
Degeneração Neural/microbiologia , Degeneração Neural/virologia , Animais , Infecções Bacterianas/epidemiologia , Sistema Nervoso Central/microbiologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Humanos , Modelos Biológicos , Degeneração Neural/epidemiologia , Degeneração Neural/patologia , Estresse Oxidativo , Viroses/epidemiologia
13.
PLoS One ; 7(6): e39853, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22768144

RESUMO

BACKGROUND: Susceptibility to viral infections as well as their severity are higher in men than in women. Heightened antiviral responses typical of women are effective for rapid virus clearance, but if excessively high or prolonged, can result in chronic/inflammatory pathologies. We investigated whether this variability could be in part attributable to differences in the response to the Toll-Like Receptors (TLR) more involved in the virus recognition. METHODS: Cytokine production by peripheral blood mononuclear cells (PBMCs) from male and female healthy donors after stimulation with Toll-like receptors (TLR) 3, 7, 8, 9 ligands or with viruses (influenza and Herpes-simplex-1) was evaluated. RESULTS: Compared to females, PBMCs from males produced not only lower amounts of IFN-α in response to TLR7 ligands but also higher amounts of the immunosuppressive cytokine IL10 after stimulation with TLR8 and TLR9 ligands or viruses. IL10 production after TLR9 ligands or HSV-1 stimulation was significantly related with plasma levels of sex hormones in both groups, whereas no correlation was found in cytokines produced following TLR7 and TLR8 stimulation. CONCLUSIONS: Given the role of an early production of IL10 by cells of innate immunity in modulating innate and adaptive immune response to viruses, we suggest that sex-related difference in its production following viral nucleic acid stimulation of TLRs may be involved in the sex-related variability in response to viral infections.


Assuntos
Interleucina-10/biossíntese , Caracteres Sexuais , Receptor 8 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Viroses/imunologia , Adulto , Feminino , Hormônios Esteroides Gonadais/metabolismo , Células HEK293 , Herpesvirus Humano 1/imunologia , Humanos , Vírus da Influenza A/imunologia , Interferon-alfa/biossíntese , Interleucina-12/biossíntese , Interleucina-13/biossíntese , Leucócitos Mononucleares/metabolismo , Ligantes , Masculino , Pessoa de Meia-Idade , Ácidos Nucleicos/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
14.
Neurobiol Aging ; 32(12): 2323.e13-26, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20674092

RESUMO

Epidemiological and experimental findings suggest that chronic infection with Herpes simplex virus type 1 (HSV-1) may be a risk factor for Alzheimer's disease (AD), but the molecular mechanisms underlying this association have not been fully identified. We investigated the effects of HSV-1 on excitability and intracellular calcium signaling in rat cortical neurons and the impact of these effects on amyloid precursor protein (APP) processing and the production of amyloid-ß peptide (Aß). Membrane depolarization triggering firing rate increases was observed shortly after neurons were challenged with HSV-1 and was still evident 12 hours postinfection. These effects depended on persistent sodium current activation and potassium current inhibition. The virally induced hyperexcitability triggered intracellular Ca(2+) signals that significantly increased intraneuronal Ca(2+) levels. It also enhanced activity- and Ca(2+)-dependent APP phosphorylation and intracellular accumulation of Aß42. These findings indicate that HSV-1 causes functional changes in cortical neurons that promote APP processing and Aß production, and they are compatible with the co-factorial role for HSV-1 in the pathogenesis of AD suggested by previous findings.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Cálcio/fisiologia , Córtex Cerebral/virologia , Herpesvirus Humano 1/fisiologia , Neurônios/virologia , Fragmentos de Peptídeos/metabolismo , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Potenciais da Membrana/genética , Neurônios/metabolismo , Fosforilação/genética , Ratos , Regulação para Cima/genética , Replicação Viral/genética
15.
PLoS One ; 5(11): e13989, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21085580

RESUMO

Lifelong latent infections of the trigeminal ganglion by the neurotropic herpes simplex virus type 1 (HSV-1) are characterized by periodic reactivation. During these episodes, newly produced virions may also reach the central nervous system (CNS), causing productive but generally asymptomatic infections. Epidemiological and experimental findings suggest that HSV-1 might contribute to the pathogenesis of Alzheimer's disease (AD). This multifactorial neurodegenerative disorder is related to an overproduction of amyloid beta (Aß) and other neurotoxic peptides, which occurs during amyloidogenic endoproteolytic processing of the transmembrane amyloid precursor protein (APP). The aim of our study was to identify the effects of productive HSV-1 infection on APP processing in neuronal cells. We found that infection of SH-SY5Y human neuroblastoma cells and rat cortical neurons is followed by multiple cleavages of APP, which result in the intra- and/or extra-cellular accumulation of various neurotoxic species. These include: i) APP fragments (APP-Fs) of 35 and 45 kDa (APP-F35 and APP-F45) that comprise portions of Aß; ii) N-terminal APP-Fs that are secreted; iii) intracellular C-terminal APP-Fs; and iv) Aß(1-40) and Aß(1-42). Western blot analysis of infected-cell lysates treated with formic acid suggests that APP-F35 may be an Aß oligomer. The multiple cleavages of APP that occur in infected cells are produced in part by known components of the amyloidogenic APP processing pathway, i.e., host-cell ß-secretase, γ-secretase, and caspase-3-like enzymes. These findings demonstrate that HSV-1 infection of neuronal cells can generate multiple APP fragments with well-documented neurotoxic potentials. It is tempting to speculate that intra- and extracellular accumulation of these species in the CNS resulting from repeated HSV-1 reactivation could, in the presence of other risk factors, play a co-factorial role in the development of AD.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Herpesvirus Humano 1/fisiologia , Neurônios/metabolismo , Neurônios/virologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Células HeLa , Herpesvirus Humano 1/genética , Interações Hospedeiro-Patógeno , Humanos , Microscopia Confocal , Mutação , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neuroblastoma/virologia , Neurônios/citologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Multimerização Proteica , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
EMBO J ; 22(7): 1697-706, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12660175

RESUMO

The number of telomeric DNA repeats at chromosome ends is maintained around a mean value by a dynamic balance between elongation and shortening. In particular, proteins binding along the duplex part of telomeric DNA set the number of repeats by progressively limiting telomere growth. The paradigm of this counting mechanism is the Rap1 protein in Saccharomyces cerevisiae. We demonstrate here that a Rap1-independent mechanism regulates the number of yeast telomeric repeats (TG(1-3)) and of vertebrate repeats (T(2)AG(3)) when TEL1, a yeast ortholog of the human gene encoding the ATM kinase, is inactivated. In addition, we show that a T(2)AG(3)-only telomere can be formed and maintained in humanized yeast cells carrying a template mutation of the gene encoding the telomerase RNA, which leads to the synthesis of vertebrate instead of yeast repeats. Genetic and biochemical evidences indicate that this telomere is regulated in a Rap1-independent manner, both in TEL1 and in tel1Delta humanized yeast cells. Altogether, these findings shed light on multiple repeat-counting mechanisms, which may share critical features between lower and higher eukaryotes.


Assuntos
Saccharomyces cerevisiae/ultraestrutura , Telômero , Proteínas rap1 de Ligação ao GTP/fisiologia , Sequência de Bases , Primers do DNA , Humanos , Plasmídeos , Saccharomyces cerevisiae/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA