Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 157(4): 1284-1299, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33180957

RESUMO

Diminished glutamate (Glu) uptake via the excitatory amino acid transporter EAAT2, which normally accounts for ~90% of total forebrain EAAT activity, may contribute to neurodegeneration via Glu-mediated excitotoxicity. C-terminal cleavage by caspase-3 (C3) was reported to mediate EAAT2 inactivation and down-regulation in the context of neurodegeneration. For a detailed analysis of C3-dependent EAAT2 degradation, we employed A172 glioblastoma as well as hippocampal HT22 cells and murine astrocytes over-expressing VSV-G-tagged EAAT2 constructs. C3 activation was induced by staurosporine (STR). In HT22 cells, STR-induced C3 activation-induced rapid EAAT2 protein degradation. The mutation of asparagine 504 to aspartate (D504N), which should inactivate the putative C3 cleavage site, increased EAAT2 activity in A172 cells. In contrast, the D504N mutation did not protect EAAT2 protein against STR-induced degradation in HT22 cells, whereas inhibition of caspases, ubiquitination and the proteasome did. Similar results were obtained in astrocytes. Phylogenetic analysis showed that C-terminal ubiquitin acceptor sites-but not the putative C3 cleavage site-exhibit a high degree of conservation. Moreover, C-terminal truncation mimicking C3 cleavage increased rather than decreased EAAT2 activity and stability as well as protected EAAT2 against STR-induced ubiquitination-dependent degradation. We conclude that cellular stress associated with endogenous C3 activation degrades EAAT2 via a pathway involving ubiquitination and the proteasome but not direct C3-mediated cleavage. In addition, C3 cleavage of EAAT2, described to occur in other models, is unlikely to inactivate EAAT2. However, mutation of the highly conserved D504 within the putative C3 cleavage site increases EAAT2 activity via an unknown mechanism.


Assuntos
Caspase 3/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Degeneração Neural/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Estresse Fisiológico/fisiologia , Animais , Células Cultivadas , Inibidores Enzimáticos/toxicidade , Humanos , Camundongos , Estaurosporina/toxicidade , Ubiquitinação
2.
Int J Med Sci ; 17(14): 2207-2213, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32922183

RESUMO

2,3,5,4'-Tetrahydroxystilbene-2-O-ß-d-glucoside (THSG) is the major active ingredient in Plygonum multiflorum that displays a great deal of health-benefits including anti-oxidation, anti-hyperlipidemia, anti-cancer, anti-inflammation and neuroprotection. However, it is unclear whether THSG exerts neuroprotective functions by regulating neurotrophic factors and their associated signaling pathways. In this study, hippocampal neurons were challenged with staurosporine (STS) to establish a neural damage model. We found that STS-induced cytotoxicity introduced significant morphological collapse and initiating cell apoptosis, along with the down regulation of BDNF and TrkB/Akt signaling axis. In contrast, neurons pretreated with THSG showed resistance to STS-induced toxicity and maintained cell survival. THSG rescued STS induced dysfunctions of BDNF and its associated TrkB/Akt signaling, and restored the expression of Bcl-2 and Caspase-3. However, inhibition of TrkB activity by K252a or Akt signaling by LY294002 abolished the neuroprotective effects of THSG. Therefore, BDNF and TrkB/Akt signaling axis is a promise target for THSG mediated neuroprotective functions.


Assuntos
Glucosídeos/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estilbenos/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Carbazóis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Fallopia multiflora/química , Hipocampo/citologia , Alcaloides Indólicos/farmacologia , Morfolinas/farmacologia , Neurônios/patologia , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Receptor trkB/antagonistas & inibidores , Receptor trkB/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estaurosporina/toxicidade
3.
Biochimie ; 177: 78-86, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32835737

RESUMO

l-Dopa Decarboxylase (DDC) is a pyridoxal requiring enzyme that catalyzes the decarboxylation of L-3,4-dihydroxyphenylalanine (l-Dopa) to Dopamine (DA). The function of DDC in physiological and pathological biochemical pathways remains poorly understood, while the function and regulation of human DDC isoforms is almost completely elusive. We have shown that Annexin V, a fundamental apoptosis marker, is an inhibitor of l-Dopa decarboxylase activity. Here we show the interaction of both the full-length DDC and the truncated isoform alternative DDC (Alt-DDC) with Annexin V in human tissue and cell lines. Interestingly, DDC isoform expression is enhanced or remains unaffected following staurosporine (STS) treatment, despite increased levels of cytotoxicity and apoptosis. The findings presented here provide novel insights concerning the involvement of DDC in programmed cell death.


Assuntos
Anexina A5/metabolismo , Anexina A5/farmacologia , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Descarboxilases de Aminoácido-L-Aromático/genética , Morte Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Cobalto/toxicidade , Cricetinae , Feminino , Humanos , Placenta/metabolismo , Gravidez , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estaurosporina/toxicidade
4.
Int J Mol Sci ; 20(20)2019 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-31640160

RESUMO

Tauopathy is a class of a neurodegenerative disorder linked with tau hyperphosphorylation, proteolysis, and aggregation. Tau can be subjected to proteolysis upon calpain activation in Alzheimer disease (AD), and traumatic brain injury (TBI). We and others have extensively researched calpain-mediated tau breakdown products (Tau-BDP; 45K, 35K, and 17K). Tau proteolysis might also generate low molecular weight (LMW ≤10K) proteolytic peptides after neurodegenerative damage. In this study, we have subjected purified tau protein (phospho and non-phospho) and mouse brain lysate to calpain-1 digestion to characterize the LMW generated by nano-liquid chromatography coupled to electrospray ionization to tandem mass spectrometry (nano-LC-ESI-MS/MS). We have also challenged differentiated primary cerebrocortical neuronal cultures (CTX) with neurotoxic agents (calcium ionophore calcimycin (A23187), staurosporine (STS), N-methyl-D-aspartate (NMDA), and Maitotoxin (MTX)) that mimic neurodegeneration to investigate the peptidome released into the conditioned cell media. We used a simple workflow in which we fractionate LMW calpain-mediated tau peptides by ultrafiltration (molecular weight cut-off value (MWCO) of 10K) and subject filtrate fractions to nano-LC-MS/MS analysis. The high molecular weight (HMW) peptides and intact proteins retained on the filter were analyzed separately by western blotting using total and phospho-specific tau antibodies. We have identified several novel proteolytic tau peptides (phosphorylated and non-phosphorylated) that are only present in samples treated with calpain or cell-based calpain activation model (particularly N- and C-terminal peptides). Our findings can help in developing future research strategies emphasizing on the suppression of tau proteolysis as a target.


Assuntos
Calpaína/metabolismo , Neurônios/citologia , Peptídeos/análise , Cultura Primária de Células/métodos , Proteínas tau/química , Animais , Calcimicina/toxicidade , Células Cultivadas , Cromatografia Líquida , Toxinas Marinhas/toxicidade , Camundongos , Camundongos Transgênicos , Peso Molecular , N-Metilaspartato/toxicidade , Nanotecnologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oxocinas/toxicidade , Fosforilação , Proteólise , Ratos , Espectrometria de Massas por Ionização por Electrospray , Estaurosporina/toxicidade , Espectrometria de Massas em Tandem
5.
Cytometry A ; 95(6): 664-671, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30508273

RESUMO

Most cancer patients die from metastatic disease as a result of a circulating tumor cell (CTC) spreading from a primary tumor through the blood circulation to distant organs. Many studies have demonstrated the tremendous potential of using CTC counts as prognostic markers of metastatic development and therapeutic efficacy. However, it is only the viable CTCs capable of surviving in the blood circulation that can create distant metastasis. To date, little progress has been made in understanding what proportion of CTCs is viable and what proportion is in an apoptotic state. Here, we introduce a novel approach toward in situ characterization of CTC apoptosis status using a multicolor in vivo flow cytometry platform with fluorescent detection for the real-time identification and enumeration of such cells directly in blood flow. The proof of concept was demonstrated with two-color fluorescence flow cytometry (FFC) using breast cancer cells MDA-MB-231 expressing green fluorescein protein (GFP), staurosporine as an activator of apoptosis, Annexin-V apoptotic kit with orange dye color, and a mouse model. The future application of this new platform for real-time monitoring of antitumor drug efficiency is discussed. © 2018 International Society for Advancement of Cytometry.


Assuntos
Apoptose , Vasos Sanguíneos/diagnóstico por imagem , Neoplasias da Mama/diagnóstico por imagem , Citometria de Fluxo/métodos , Células Neoplásicas Circulantes/química , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/sangue , Vasos Sanguíneos/efeitos dos fármacos , Contagem de Células , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Fluorescência , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Nus , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patologia , Células Neoplásicas Circulantes/efeitos da radiação , Estaurosporina/toxicidade
6.
J Neurosci Res ; 96(3): 427-435, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28877366

RESUMO

ACTH, a melanocortin peptide used to treat multiple sclerosis (MS) relapses, acts by stimulating adrenal corticosteroid (CS) production via melanocortin receptor 2 (MC2R), but it may also exert a therapeutic effect independent of CS by stimulating other melanocortin receptors (MCR) distributed in many tissues, including the brain. We reported that oligodendroglia (OL) and oligodendroglial precursor cells (OPC) express MC4R, and that ACTH 1-39 protects OL and OPC in vitro from cell death induced by mechanisms likely involved in white matter damage in MS. This study investigates expression of MC1R, MC2R, MC3R and MC5R in OL and MC4R in OPC using immunocytochemistry with MCR subtype specific antibodies. OL express surface MC1R, MC3R and MC5R, in addition to MC4R. To investigate whether these receptors are functional, we asked if signaling through MCR is involved in ACTH protection of cultured rat OL from apoptosis (staurosporine), or cell death induced by excitotoxicity (glutamate), reactive oxygen species (ROS), or an inflammatory mediator (quinolinic acid). Like ACTH 1-39, MCR subtype specific agonists for MC1R, MC3R, MC4R and MC5R all protected OL from these insults. Conversely, antagonists for MC3R and MC4R blocked ACTH protection of OL. We then investigated the role of MC4R, as a prototype MCR, in protection and proliferation of OPC; MC4R agonists protected OPC and increased their proliferation, while antagonists blocked these effects. Our results demonstrate that MCR on OL and OPC are functional and activate signaling pathways that protect against mechanisms involved in OL damage in MS, suggesting potential beneficial effects in neurologic diseases.


Assuntos
Hormônio Adrenocorticotrópico/farmacologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Receptores de Melanocortina/biossíntese , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Ácido Glutâmico/toxicidade , Peróxido de Hidrogênio/toxicidade , Imuno-Histoquímica , Cultura Primária de Células , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/metabolismo , Ácido Quinolínico/toxicidade , Ratos Sprague-Dawley , Receptores de Melanocortina/agonistas , Receptores de Melanocortina/antagonistas & inibidores , Estaurosporina/toxicidade
7.
PLoS One ; 12(8): e0182150, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28787459

RESUMO

Synaptic activity increases the resistance of neurons to diverse apoptotic insults; however, the underlying mechanisms remain less well understood. Zinc promotes cell survival under varied conditions, but the role of synaptically released zinc in the activity-dependent anti-apoptotic effect is unknown. Using cultured hippocampal slices and primary neurons we show that a typical apoptosis inducer-staurosporine (STP) was able to cause concentration-dependent apoptotic cell death in brain slices; Enhanced synaptic activity by bicuculline (Bic)/4-Aminopyridine (AP) treatment effectively prevented neurons from STP-induced cell apoptosis, as indicated by increased cell survival and suppressed caspase-3 activity. Application of Ca-EDTA, a cell membrane-impermeable zinc chelator which can efficiently capture the synaptically released zinc, completely blocked the neuronal activity-dependent anti-apoptotic effect. Same results were also observed in cultured primary hippocampal neurons. Therefore, our results indicate that synaptic activity improves neuronal resistance to apoptosis via synaptically released zinc.


Assuntos
Apoptose/fisiologia , Neurônios/fisiologia , Neuroproteção/fisiologia , Transmissão Sináptica/fisiologia , Zinco/metabolismo , 4-Aminopiridina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Bicuculina/farmacologia , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Quelantes/farmacologia , Relação Dose-Resposta a Droga , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Hipocampo/fisiologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ratos Sprague-Dawley , Estaurosporina/toxicidade , Transmissão Sináptica/efeitos dos fármacos , Técnicas de Cultura de Tecidos
8.
Neurotox Res ; 31(1): 63-76, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27577743

RESUMO

As a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase, demalonylase, and desuccinylase, sirtuin 5 (SIRT5) in host cells has been reportedly observed in the mitochondria, in the cytosol/cytoplasm or in the nucleus. Various functional roles of SIRT5 have also been described in cellular metabolism, energy production, detoxification, oxidative stress, and apoptosis, but some of the reported results are seemingly inconsistent or even contradictory to one another. Using immunocytochemistry, molecular biology, gene transfection, and flow cytometry, we investigated the expression, subcellular distribution, and possible functional roles of SIRT5 in regulating apoptosis and oxidative stress of cultured SH-EP neuroblastoma cells. Both endogenous and transfected exogenous SIRT5 were observed in mitochondria of host SH-EP cells. Overexpression of SIRT5 markedly protected SH-EP cells from apoptosis induced by staurosporine or by incubation in Hank's balanced salt solution. SIRT5 also lowered the level of oxidative stress and countered the toxicity of hydrogen peroxide to SH-EP cells. It was suggested that the anti-apoptotic role of SIRT5 was mediated, at least in part, by its anti-oxidative effect in SH-EP neuroblastoma cells although the involved molecular mechanisms remain to be elucidated in details.


Assuntos
Antioxidantes/metabolismo , Apoptose/fisiologia , Neuroproteção/fisiologia , Sirtuínas/metabolismo , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Microscopia Confocal , Mitocôndrias/metabolismo , Neuroproteção/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sirtuínas/genética , Estaurosporina/toxicidade , Transfecção
9.
Open Biol ; 6(11)2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27810968

RESUMO

Amyloidogenic protein aggregation impairs cell function and is a hallmark of many chronic degenerative disorders. Protein aggregation is also a major event during acute injury; however, unlike amyloidogenesis, the process of injury-induced protein aggregation remains largely undefined. To provide this insight, we profiled the insoluble proteome of several cell types after acute injury. These experiments show that the disulfide-driven process of nucleocytoplasmic coagulation (NCC) is the main form of injury-induced protein aggregation. NCC is mechanistically distinct from amyloidogenesis, but still broadly impairs cell function by promoting the aggregation of hundreds of abundant and essential intracellular proteins. A small proportion of the intracellular proteome resists NCC and is instead released from necrotic cells. Notably, the physicochemical properties of NCC-resistant proteins are contrary to those of NCC-sensitive proteins. These observations challenge the dogma that liberation of constituents during necrosis is anarchic. Rather, inherent physicochemical features including cysteine content, hydrophobicity and intrinsic disorder determine whether a protein is released from necrotic cells. Furthermore, as half of the identified NCC-resistant proteins are known autoantigens, we propose that physicochemical properties that control NCC also affect immune tolerance and other host responses important for the restoration of homeostasis after necrotic injury.


Assuntos
Etoposídeo/toxicidade , Agregados Proteicos , Proteoma/efeitos dos fármacos , Estaurosporina/toxicidade , Apoptose , Linhagem Celular , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/metabolismo , Proteína Ligante Fas/toxicidade , Humanos , Células Jurkat , Proteômica/métodos
10.
Neuropharmacology ; 111: 195-211, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27600687

RESUMO

Activation of metabotropic glutamate receptors (mGluRs) modulates neuronal excitability. Here, we evaluated the neuroprotective potential of four structurally diverse activators of group II and III mGluRs: an orthosteric agonist of group II (LY354740), an orthosteric agonist of group III (ACPT-I), an allosteric agonist of mGluR7 (AMN082) and a positive allosteric modulator (PAM) of mGluR4 (VU0361737). Neurotoxicity was induced by the pro-apoptotic agents: staurosporine (St) and doxorubicin (Dox) or the excitotoxic factor glutamate (Glu). The effects were analyzed in primary hippocampal (HIP) and cerebellar granule cell (CGC) cultures at two developmental stages, at 7 and 12 days in vitro (DIV). The data reveal a general neuroprotective effect of group II and III mGluR activators against the St- and Glu- but not Dox-induced cell damage. We found that neuroprotective effects of group II and III mGluR orthosteric agonists (LY354740 and ACPT-I) were higher at 12 DIV when compared to 7 DIV cells. In contrast, the efficiency of allosteric mGluR agents (AMN082 and VU0361737) did not differ between 7 and 12 DIV in both, St and Glu models of neuronal cell damage. Interestingly, the protective effects of activators of group II and III mGluRs were blocked by relevant antagonists only against Glu-induced neurotoxicity. Moreover, the observed neuroprotective action of group II and III mGluR activators in the St model was associated with a decreased number of PI-positive cells and no alterations in the caspase-3 activity. Finally, we showed that MAPK/ERK pathway activation was potentially involved in the mechanism of ACPT-I- and AMN082-induced neuroprotection against the St-evoked cellular damage. Our comparative study demonstrated the developmental stage-dependent neuroprotective effect of orthosteric group II and III mGluR agonists. In comparison to allosteric modulators, orthosteric compounds may provide more specific tools for suppression of neuronal cell loss associated with various chronic neurodegenerative conditions. Our results also suggest that the inhibition of intracellular pathways mediating necrotic, rather than apoptotic cascades, may be involved in neuroprotective effects of activators of group II and III mGluRs.


Assuntos
Compostos de Anilina/administração & dosagem , Compostos Benzidrílicos/administração & dosagem , Compostos Bicíclicos com Pontes/administração & dosagem , Morte Celular/efeitos dos fármacos , Ciclopentanos/administração & dosagem , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Ácidos Picolínicos/administração & dosagem , Receptores de Glutamato Metabotrópico/agonistas , Ácidos Tricarboxílicos/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Cerebelo/efeitos dos fármacos , Doxorrubicina/toxicidade , Ácido Glutâmico/toxicidade , Hipocampo/efeitos dos fármacos , Camundongos , Cultura Primária de Células , Estaurosporina/toxicidade
11.
BMB Rep ; 49(10): 560-565, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27439606

RESUMO

Granzyme A (GzmA) was first identified as a cytotoxic T lymphocyte protease protein with limited tissue expression. A number of cellular proteins are known to be cleaved by GzmA, and its function is to induce apoptosis. Histones H1, H2B, and H3 were identified as GzmA substrates during apoptotic cell death. Here, we demonstrated that histone H4 was cleaved by GzmA during staurosporine-induced cell death; however, in the presence of caspase inhibitors, staurosporine-treated Raji cells underwent necroptosis instead of apoptosis. Furthermore, histone H4 cleavage was blocked by the GzmA inhibitor nafamostat mesylate and by GzmA knockdown using siRNA. These results suggest that histone H4 is a novel substrate for GzmA in staurosporine-induced cells. [BMB Reports 2016; 49(10): 560-565].


Assuntos
Apoptose/efeitos dos fármacos , Granzimas/metabolismo , Histonas/metabolismo , Estaurosporina/toxicidade , Clorometilcetonas de Aminoácidos/farmacologia , Benzamidinas , Western Blotting , Inibidores de Caspase/farmacologia , Linhagem Celular Tumoral , Citometria de Fluxo , Granzimas/antagonistas & inibidores , Granzimas/genética , Guanidinas/farmacologia , Células HEK293 , Células HeLa , Humanos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Estaurosporina/farmacologia
12.
Microsc Res Tech ; 79(6): 532-40, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27059940

RESUMO

It is known that, besides a wide range of functions, melatonin provides protection against oxidative stress, thanks to its ability to act, directly, as a free radical scavenger and, indirectly, by stimulating antioxidant enzymes production and mitochondrial electron transport chain efficiency. Oxidative stress is one of the major players in initiating apoptotic cell death in skeletal muscle, as well as in other tissues. Apoptosis is essential for skeletal muscle development and homeostasis; nevertheless, its misregulation has been frequently observed in several myopathies, in sarcopenia, as well as in denervation and disuse. Melatonin activity was investigated in undifferentiated C2C12 skeletal muscle cells, after exposure to various apoptotic chemical triggers, chosen for their different mechanisms of action. Cells were pretreated with melatonin and then exposed to hydrogen peroxide, etoposide and staurosporine. Morphofunctional and molecular analyses show that in myoblasts melatonin prevents oxidative stress and apoptosis induced by chemicals following, at least in part, the mitochondria pathway. These results confirm melatonin ability to act as an antioxidant and antiapoptotic molecule in skeletal muscle cells, thus suggesting a possible therapeutic strategy for myopathies involving apoptosis misregulation. Microsc. Res. Tech. 79:532-540, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Apoptose/efeitos dos fármacos , Melatonina/farmacologia , Mioblastos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular , Etoposídeo/toxicidade , Peróxido de Hidrogênio/toxicidade , Camundongos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Mitocôndrias , Estaurosporina/toxicidade
13.
J Neurol Sci ; 362: 21-6, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26944112

RESUMO

Damage to myelin and oligodendroglia (OL) in multiple sclerosis (MS) results from a wide array of mechanisms including excitotoxicity, neuroinflammation and oxidative stress. We previously showed that ACTH 1-39, a melanocortin, protects OL in mixed glial cultures and enriched OL cultures, inhibiting OL death induced by staurosporine, ionotropic glutamate receptors, quinolinic acid or reactive oxygen species (ROS), but not nitric oxide (NO) or kynurenic acid. OL express melanocortin receptor 4 (MC4R), suggesting a direct protective effect of ACTH 1-39 on OL. However, these results do not rule out the possibility that astroglia (AS) or microglia (MG) also play roles in protection. To investigate this possibility, we prepared conditioned medium (CM) from AS and MG treated with ACTH, then assessed the protective effects of the CM on OL. CM from AS treated with ACTH protected OL from glutamate, NMDA, AMPA, quinolinic acid and ROS but not from kainate, staurosporine, NO or kynurenic acid. CM from MG treated with ACTH did not protect from any of these molecules, nor did CM from AS or MG not treated with ACTH. While protection of OL by ACTH from several toxic molecules involves direct effects on OL, ACTH can also stimulate AS to produce mediators that protect against some molecules but not others. Thus the cellular mechanisms underlying the protective effects of ACTH for OL are complex, varying with the toxic molecules.


Assuntos
Hormônio Adrenocorticotrópico/farmacologia , Astrócitos/fisiologia , Hormônios/farmacologia , Oligodendroglia/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Astrócitos/química , Bassia scoparia/citologia , Morte Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Inibidores Enzimáticos/toxicidade , Agonistas de Aminoácidos Excitatórios/toxicidade , Peróxido de Hidrogênio/toxicidade , Oxidantes/toxicidade , Ratos , Estaurosporina/toxicidade
14.
Nanotechnology ; 27(16): 164001, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26963176

RESUMO

The response of cells to toxins is commonly investigated by detecting intracellular markers for cell death, such as caspase proteins. This requires the introduction of labels by the permeabilization or complete lysis of cells. Here we introduce a non-invasive tool for monitoring a caspase protein in the extracellular medium. The tool is based on highly sensitive optical micro-devices, referred to as whispering-gallery mode biosensors (WGMBs). WGMBs are functionalized with antibodies for the specific and label-free detection of procaspase-3 released from human embryonic kidney HEK293 and neuroglioma H4 cells after introducing staurosporine and rotenone toxins, respectively. Additional tests show that the extracellular accumulation of procaspase-3 is concomitant with a decrease in cell viability. The hitherto unknown release of procaspase-3 from cells in response to toxins and its accumulation in the medium is further investigated by Western blot, showing that the extracellular detection of procaspase-3 is interrelated with cytotoxicity of alpha-synuclein protein (aSyn) overexpressed in H4 cells. These studies provide evidence for procaspase-3 as a novel extracellular biomarker for cell death, with applications in cytotoxicity tests. Such WGMBs could be applied to further identify as-yet unknown extracellular biomarkers using established antibodies against intracellular antigens.


Assuntos
Técnicas Biossensoriais/instrumentação , Caspase 3/metabolismo , Rotenona/toxicidade , Estaurosporina/toxicidade , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , alfa-Sinucleína/metabolismo
15.
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
16.
Graefes Arch Clin Exp Ophthalmol ; 254(2): 285-95, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26686232

RESUMO

PURPOSE: To examine the effects of media and deswelling agents on human corneal endothelial and epithelial cell viability using a previously developed screening system. METHODS: The human corneal endothelial cell line HCEC-12 and the human corneal epithelial cell line HCE-T were cultured in four different corneal organ culture media (serum-supplemented: MEM +2 % FCS, CorneaMax®/CorneaJet®, serum-free: Human Endothelial-SFM, Stemalpha-2 and -3) with and without 6 % dextran T500 or 7 % HES 130/0.4. Standard growth media F99HCEC and DMEM/F12HCE-T served as controls. In additional controls, the stress inducers staurosporine or hydrogen peroxide were added. After 5 days in the test media, cell viability was assessed by flow cytometrically quantifying apoptotic and necrotic cells (sub-G1 DNA content, vital staining with YO-PRO-1® and propidium iodide) and intracellular reactive oxygen species (ROS). RESULTS: The MEM-based media were unable to support HCEC-12 and HCE-T survival under stress conditions, resulting in significantly increased numbers of apoptotic and necrotic cells. HCEC-12 survival was markedly improved in SFM-based media even under staurosporine or hydrogen peroxide. Likewise, HCE-T survival was improved in SFM with or without dextran. The media CorneaMax®, CorneaJet®, and CorneaMax® with HES supported HCEC-12 survival better than MEM-based media, but less well than SFM-based media. HCE-T viability was also supported by CorneaJet®, but not by CorneaMax® with or without HES. Stemalpha-based media were not suitable for maintaining viability of HCEC-12 or HCE-T in the applied cell culture system. CONCLUSIONS: The use of serum-supplemented MEM-based media for corneal organ culture should be discontinued in favour of serum-free media like SFM.


Assuntos
Meios de Cultura/farmacologia , Dextranos/farmacologia , Endotélio Corneano/patologia , Epitélio Corneano/patologia , Derivados de Hidroxietil Amido/farmacologia , Substitutos do Plasma/farmacologia , Apoptose , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultura Livres de Soro/farmacologia , Inibidores Enzimáticos/toxicidade , Citometria de Fluxo , Humanos , Peróxido de Hidrogênio/toxicidade , Microscopia de Contraste de Fase , Necrose , Técnicas de Cultura de Órgãos , Oxidantes/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Estaurosporina/toxicidade
17.
BMC Ophthalmol ; 15: 174, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26653778

RESUMO

BACKGROUND: The effect of excess glucose on retinal cellular health remains controversial, and cellular reducing equivalents, as indicators of cellular energy production, are widely used as substitute indicators of retinal cellular health. These investigations hypothesised that excess energy substrate availability, as occurs in the diabetic retina, increases the susceptibility of retinal neurons to injury in the presence of increased cellular reducing equivalents. METHODS: The response of 661W cells to phototoxicity, oxidative stress induced by H2O2 and apoptosis induction by staurosporine was characterised in the presence of 5mM glucose and B27 defined media without insulin. Cellular insult was produced by phototoxicity, H2O2 and the apoptosis induction agent staurosporine. The effect of physiologically relevant alterations in environmental glucose on cellular reducing equivalents was assessed by MTT dye reduction and NAD(P)H assays, and cell survival was assessed via caspase 3/7 activation and Annexin V/PI flow cytometry. RESULTS: 661W photoreceptor-like cells underwent dose dependent cell death primarily by apoptosis in response to phototoxic insult, H2O2, and staurosporine by all measures of cellular viability. Exposure of cells to 25mM glucose (diabetic-type conditions) increased cell death in response to all insults as measured by caspase 3/7 activation and Annexin V/PI flow cytometry. Cellular reducing equivalents were nonetheless increased in all models of injury in the presence of excess glucose. The mechanism of this increase was partly due to increased NADPH but not NADH levels in the presence of 25mM glucose. CONCLUSIONS: Acute exposure to 25mM glucose decreased the resilience of 661W photoreceptor-like cells to a range of cellular stressors whilst maintaining or increasing cellular reducing equivalents, partly be increasing NADPH levels. This shows that in 661W cells, diabetic levels of glucose decrease cellular resilience to injury. The decoupling of cellular reducing equivalents levels from cell survival has important implications when investigating the mechanisms of neuronal damage in diabetic retinal neuropathy.


Assuntos
Glucose/farmacologia , Células Fotorreceptoras/efeitos dos fármacos , Células Fotorreceptoras/patologia , Animais , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Inibidores Enzimáticos/toxicidade , Citometria de Fluxo , Humanos , Peróxido de Hidrogênio/toxicidade , Modelos Biológicos , NADP/metabolismo , Oxidantes/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Células Fotorreceptoras/efeitos da radiação , Lesões Experimentais por Radiação/etiologia , Estaurosporina/toxicidade
18.
Colloids Surf B Biointerfaces ; 135: 8-17, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26231861

RESUMO

The main objectives of the present study were to investigate the biocompatibility of polyelectrolyte-coated nanocapsules and to evaluate the neuroprotective action of the nanoencapsulated water-insoluble neuroprotective drug-undecylenic acid (UDA), in vitro. Core-shell nanocapsules were synthesized using nanoemulsification and the layer-by-layer (LbL) technique (by saturation method). The average size of synthesized nanocapsules was around 80 nm and the concentration was 2.5 × 10(10) particles/ml. Their zeta potential values ranged from less than -30 mV for the ones with external polyanion layers through -4 mV for the PEG-ylated layers to more than 30 mV for the polycation layers. Biocompatibility of synthesized nanocarriers was evaluated in the SH-SY5Y human neuroblastoma cell line using cell viability/toxicity assays (MTT reduction, LDH release). The results obtained showed that synthesized nanocapsules coated with PLL and PGA (also PEG-ylated) were non-toxic to SH-SY5Y cells, therefore, they were used as nanocarriers for UDA. Moreover, studies with ROD/FITC-labeled polyelectrolytes demonstrated approximately 20% cellular uptake of synthetized nanocapsules. Further studies showed that nanoencapsulated form of UDA was biocompatible and protected SH-SY5Y cells against the staurosporine-induced damage in lower concentrations than those of the same drug added directly to the culture medium. These data suggest that designed nanocapsules might serve as novel, promising delivery systems for neuroprotective agents.


Assuntos
Eletrólitos/química , Nanocápsulas/química , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacologia , Ácidos Undecilênicos/química , Ácidos Undecilênicos/farmacologia , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos , Estabilidade de Medicamentos , Humanos , Teste de Materiais , Fármacos Neuroprotetores/síntese química , Tamanho da Partícula , Estaurosporina/antagonistas & inibidores , Estaurosporina/toxicidade , Ácidos Undecilênicos/síntese química
19.
Clin Lab ; 61(7): 717-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26299070

RESUMO

BACKGROUND: Staurosporine (STS), a microbial alkaloid and potent PKC inhibitor, has become one of the most promising anti-cancer drugs. STS effectively induces apoptosis in many nucleated cells; however, it is still unclear whether STS induces apoptosis in enucleated platelets. METHODS: Apoptotic events in platelets treated with STS were assessed by flow cytometry or western blotting. RESULTS: STS induced depolarization of mitochondrial inner transmembrane potential (ΔΨm), up-regulation of Bax and Bak, phosphatidylserine (PS) exposure, release of mitochondrial cytochrome c, and activation of caspase-8 and caspase-9 in human platelets. Furthermore, STS stimulation induced phosphorylation of p38 mitogen-activated protein kinase (MAPK). Inhibition of p38 MAPK activation significantly reduced ΔΨm depolarization and PS exposure in platelets stimulated with STS. CONCLUSIONS: These data indicate that STS induces platelet apoptosis via the p38 MAPK signaling pathway. These findings suggest that platelet apoptosis-related hemorrhage should be noticed in STS and its derivatives in clinical tests.


Assuntos
Antineoplásicos/toxicidade , Apoptose/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Estaurosporina/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Plaquetas/enzimologia , Plaquetas/patologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
20.
PLoS One ; 10(6): e0128601, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26047500

RESUMO

Traumatic Brain Injury (TBI) is the result of a mechanical impact on the brain provoking mild, moderate or severe symptoms. It is acknowledged that TBI leads to apoptotic and necrotic cell death; however, the exact mechanism by which brain trauma leads to neural injury is not fully elucidated. Some studies have highlighted the pivotal role of the Kallikrein-Kinin System (KKS) in brain trauma but the results are still controversial and inconclusive. In this study, we investigated both the expression and the role of Bradykinin 1 and 2 receptors (B1R and B2R), in mediating neuronal injury under chemical neurotoxicity paradigm in PC12 cell lines. The neuronal cell line PC12 was treated with the apoptotic drug Staurosporine (STS) to induce cell death. Intracellular calcium release was evaluated by Fluo 4-AM staining and showed that inhibition of the B2R prevented calcium release following STS treatment. Differential analyses utilizing immunofluorescence, Western blot and Real-time Polymerase Chain Reaction revealed an upregulation of both bradykinin receptors occurring at 3h and 12h post-STS treatment, but with a higher induction of B2R compared to B1R. This implies that STS-mediated apoptosis in PC12 cells is mainly conducted through B2R and partly via B1R. Finally, a neuroproteomics approach was conducted to find relevant proteins associated to STS and KKS in PC12 cells. Neuroproteomics results confirmed the presence of an inflammatory response leading to cell death during apoptosis-mediated STS treatment; however, a "survival" capacity was shown following inhibition of B2R coupled with STS treatment. Our data suggest that B2R is a key player in the inflammatory pathway following STS-mediated apoptosis in PC12 cells and its inhibition may represent a potential therapeutic tool in TBI.


Assuntos
Inibidores Enzimáticos/toxicidade , Sistema Calicreína-Cinina/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Estaurosporina/toxicidade , Animais , Morte Celular/efeitos dos fármacos , Neurônios/imunologia , Neurônios/metabolismo , Células PC12 , Proteômica , Ratos , Receptor B1 da Bradicinina/análise , Receptor B1 da Bradicinina/genética , Receptor B1 da Bradicinina/imunologia , Receptor B2 da Bradicinina/análise , Receptor B2 da Bradicinina/genética , Receptor B2 da Bradicinina/imunologia , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA