Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Chem Res Toxicol ; 33(9): 2310-2323, 2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32786544

RESUMO

We have previously shown that phenyl saligenin phosphate (PSP), an organophosphorus compound which is classed as a weak inhibitor of acetylcholinesterase, triggered cytotoxicity in mitotic and differentiated H9c2 cardiomyoblasts. The aim of this study was to assess whether sublethal concentrations of PSP could disrupt the morphology of differentiating rat H9c2 cardiomyoblasts and human-induced pluripotent stem-cell-derived cardiomyocyte progenitor cells (hiPSC-CMs) and to assess the underlying cytoskeletal changes. PSP-induced changes in protein expression were monitored via Western blotting, immunocytochemistry, and proteomic analysis. PSP-mediated cytotoxicity was determined by measuring MTT reduction, LDH release, and caspase-3 activity. Sublethal exposure to PSP (3 µM) induced morphological changes in differentiating H9c2 cells (7, 9, and 13 days), reflected by reduced numbers of spindle-shaped cells. Moreover, this treatment (7 days) attenuated the expression of the cytoskeletal proteins cardiac troponin I, tropomyosin-1, and α-actin. Further proteomic analysis identified nine proteins (e.g., heat shock protein 90-ß and calumenin) which were down-regulated by PSP exposure in H9c2 cells. To assess the cytotoxic effects of organophosphorus compounds in a human cell model, we determined their effects on human-induced pluripotent stem-cell-derived cardiomyocyte progenitor cells. Chlorpyrifos and diazinon-induced cytotoxicity (48 h) was evident only at concentrations >100 µM. By contrast, PSP exhibited cytotoxicity in hiPSC-CMs at a concentration of 25 µM following 48 h exposure. Finally, sublethal exposure to PSP (3 µM; 7 days) induced morphological changes and decreased the expression of cardiac troponin I, tropomyosin-1, and α-actin in hiPSC-CMs. In summary, our data suggest cardiomyocyte morphology is disrupted in both cell models by sublethal concentrations of PSP via modulation of cytoskeletal protein expression.


Assuntos
Citoesqueleto de Actina/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Compostos Organofosforados/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Ratos
2.
Arch Toxicol ; 94(11): 3861-3875, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32749514

RESUMO

Organophosphate compounds (OPs) induce both acute and delayed neurotoxic effects, the latter of which is believed to involve their interaction with proteins other than acetylcholinesterase. However, few OP-binding proteins have been identified that may have a direct role in OP-induced delayed neurotoxicity. Given their ability to disrupt Ca2+ homeostasis, a key aim of the current work was to investigate the effects of sub-lethal neurite outgrowth inhibitory levels of OPs on the Ca2+-dependent enzyme tissue transglutaminase (TG2). At 1-10 µM, the OPs phenyl saligenin phosphate (PSP) and chlorpyrifos oxon (CPO) had no effect cell viability but induced concentration-dependent decreases in neurite outgrowth in differentiating N2a neuroblastoma cells. The activity of TG2 increased in cell lysates of differentiating cells exposed for 24 h to PSP and chlorpyrifos oxon CPO (10 µM), as determined by biotin-cadaverine incorporation assays. Exposure to both OPs (3 and/or 10 µM) also enhanced in situ incorporation of the membrane permeable substrate biotin-X-cadaverine, as indicated by Western blot analysis of treated cell lysates probed with ExtrAvidin peroxidase and fluorescence microscopy of cell monolayers incubated with FITC-streptavidin. Both OPs (10 µM) stimulated the activity of human and mouse recombinant TG2 and covalent labelling of TG2 with dansylamine-labelled PSP was demonstrated by fluorescence imaging following SDS-PAGE. A number of TG2 substrates were tentatively identified by mass spectrometry, including cytoskeletal proteins, chaperones and proteins involved protein synthesis and gene regulation. We propose that the elevated TG2 activity observed is due to the formation of a novel covalent adduct between TG2 and OPs.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proteínas de Ligação ao GTP/efeitos dos fármacos , Neuroblastoma/metabolismo , Crescimento Neuronal/efeitos dos fármacos , Organofosfatos/toxicidade , Transglutaminases/efeitos dos fármacos , Aminas/metabolismo , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Clorpirifos/análogos & derivados , Clorpirifos/toxicidade , Humanos , Camundongos , Compostos Organofosforados/toxicidade , Proteína 2 Glutamina gama-Glutamiltransferase , Proteômica , Ratos , Espécies Reativas de Oxigênio
3.
Toxicol Appl Pharmacol ; 308: 20-31, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27521977

RESUMO

Chlorpyrifos (CPF) and CPF-oxon (CPO) are known to inhibit neurite outgrowth but little is known about their ability to induce neurite retraction in differentiating neuronal cells. The aims of this study were to determine the ability of these compounds to destabilize neurites and to identify the key molecular events involved. N2a cells were induced to differentiate for 20h before exposure to CPF or CPO for 2-8h. Fixed cell monolayers labeled with carboxyfluorescein succinimidyl ester or immunofluorescently stained with antibodies to tubulin (B512) or phosphorylated neurofilament heavy chain (Ta51) showed time- and concentration-dependent reductions in numbers and length of axon-like processes compared to the control, respectively, retraction of neurites being observed within 2h of exposure by live cell imaging. Neurofilament disruption was also observed in treated cells stained by indirect immunofluorescence with anti-phosphorylated neurofilament heavy chain (NFH) monoclonal antibody SMI34, while the microtubule network was unaffected. Western blotting analysis revealed transiently increased levels of reactivity of Ta51 after 2h exposure and reduced levels of reactivity of the same antibody following 8h treatment with both compounds, whereas reactivity with antibodies to anti-total NFH or anti-tubulin was not affected. The alteration in NFH phosphorylation at 2h exposure was associated with increased activation of extracellular signal-regulated protein kinase ERK 1/2. However, increased levels of phosphatase activity were observed following 8h exposure. These findings suggest for the first time that organophosphorothionate pesticide-induced neurite retraction in N2a cells is associated with transient increases in NFH phosphorylation and ERK1/2 activation.


Assuntos
Clorpirifos/toxicidade , Inseticidas/toxicidade , Sistema de Sinalização das MAP Quinases , Neuritos/efeitos dos fármacos , Proteínas de Neurofilamentos/metabolismo , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Camundongos , Fosforilação
4.
Chem Res Toxicol ; 28(11): 2179-91, 2015 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-26465378

RESUMO

At present, little is known about the effect(s) of organophosphorous compounds (OPs) on cardiomyocytes. In this study, we have investigated the effects of phenyl saligenin phosphate (PSP), two organophosphorothioate insecticides (diazinon and chlorpyrifos), and their acutely toxic metabolites (diazoxon and chlorpyrifos oxon) on mitotic and differentiated H9c2 cardiomyoblasts. OP-induced cytotoxicity was assessed by monitoring MTT reduction, LDH release, and caspase-3 activity. Cytotoxicity was not observed with diazinon, diazoxon, or chlorpyrifos oxon (48 h exposure; 200 µM). Chlorpyrifos-induced cytotoxicity was only evident at concentrations >100 µM. In marked contrast, PSP displayed pronounced cytotoxicity toward mitotic and differentiated H9c2 cells. PSP triggered the activation of JNK1/2 but not ERK1/2, p38 MAPK, or PKB, suggesting a role for this pro-apoptotic protein kinase in PSP-induced cell death. The JNK1/2 inhibitor SP 600125 attenuated PSP-induced caspase-3 and JNK1/2 activation, confirming the role of JNK1/2 in PSP-induced cytotoxicity. Fluorescently labeled PSP (dansylated PSP) was used to identify novel PSP binding proteins. Dansylated PSP displayed cytotoxicity toward differentiated H9c2 cells. 2D-gel electrophoresis profiles of cells treated with dansylated PSP (25 µM) were used to identify proteins fluorescently labeled with dansylated PSP. Proteomic analysis identified tropomyosin, heat shock protein ß-1, and nucleolar protein 58 as novel protein targets for PSP. In summary, PSP triggers cytotoxicity in differentiated H9c2 cardiomyoblasts via JNK1/2-mediated activation of caspase-3. Further studies are required to investigate whether the identified novel protein targets of PSP play a role in the cytotoxicity of this OP, which is usually associated with the development of OP-induced delayed neuropathy.


Assuntos
Caspase 3/metabolismo , Inibidores da Colinesterase/toxicidade , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mioblastos Cardíacos/efeitos dos fármacos , Compostos Organofosforados/toxicidade , Acetilcolinesterase/metabolismo , Animais , Antracenos/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Mioblastos Cardíacos/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos
5.
Neurotoxicology ; 103: 16-26, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38763473

RESUMO

BACKGROUND: The global coronavirus 2019 (COVID-19) pandemic began in early 2020, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In mid-2020 the CIAO (Modelling the Pathogenesis of COVID-19 Using the Adverse Outcome Pathway Framework) project was established, bringing together over 75 interdisciplinary scientists worldwide to collaboratively investigate the underlying biological mechanisms of COVID-19 and consolidate the data using the Adverse Outcome Pathway (AOP) Framework. Neurological symptoms such as anosmia and encephalitis have been frequently reported to be associated with infection with SARS-CoV-2. OBJECTIVE: Within CIAO, a working group was formed to conduct a systematic scoping review of COVID-19 and its related neurological symptoms to determine which key events and modulating factors are most commonly reported and to identify knowledge gaps. DESIGN: LitCOVID was used to retrieve 86,075 papers of which 10,244 contained relevant keywords. After title and abstract screening, 2,328 remained and their full texts were reviewed based on predefined inclusion and exclusion criteria. 991 studies fulfilled the inclusion criteria and were retrieved to conduct knowledge synthesis. RESULTS: The majority of publications reported human observational studies. Early key events were less likely to be reported compared to middle and late key events/adverse outcomes. The majority of modulating factors described related to age or sex. Less recognised COVID-19 associated AO or neurological effects of COVID-19 were also identified including multiple sclerosis/demyelination, neurodegeneration/cognitive effects and peripheral neuronal effects. CONCLUSION: There were many methodological and reporting issues noted in the reviewed studies. In particular, publication abstracts would benefit from clearer reporting of the methods and endpoints used and the key findings, to ensure relevant papers are included when systematic reviews are conducted. The information extracted from the scoping review may be useful in understanding the mechanisms of neurological effects of COVID-19 and to further develop or support existing AOPs linking COVID-19 and its neurological key events and adverse outcomes. Further evaluation of the less recognised COVID-19 effects is needed.


Assuntos
COVID-19 , Doenças do Sistema Nervoso , Humanos , COVID-19/epidemiologia , Doenças do Sistema Nervoso/epidemiologia , Doenças do Sistema Nervoso/virologia , SARS-CoV-2/patogenicidade
6.
Adv Exp Med Biol ; 724: 189-204, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22411244

RESUMO

Organophosphorous compounds (OPs) are widely used in agriculture, industry and the home. Though best known for their acute effects when used as pesticides, which target acetylcholinesterase (AChE) activity in neuromuscular junctions and the central nervous system, not all OPs are potent inhibitors of this enzyme. The widespread use of OPs has heightened concern regarding their toxicity in man, with numerous reports linking OPs to various forms of delayed neuropathy encompassing a range of neurodegenerative, psychological and neurobehavioral effects. There is mounting evidence to suggest that sub-acute levels of OPs have the ability to interact directly with a range of target proteins in addition to AChE (i.e., noncholinergic targets), causing major disruption of membrane and protein turnover, protein phosphorylation, mitochondrial dysfunction, oxidative stress and cytoskeletal re-organisation, although the mechanisms involved are not fully understood. However, major advances have been made in the study of one OP binding protein neuropathy target esterase (NTE) in terms of its true physiological role. Additionally, there is increasing evidence for the ability of OPs to cause disruption in a number of metabolic and cell signalling pathways that affect neuronal cell proliferation, differentiation and survival and to interact direct with non-esterase proteins such as tubulin. The aim of this chapter is to review our current understanding of delayed neurotoxicity, to discuss how these molecular events may relate to each other and to suggest possible future directions in mechanistic studies of OP toxicity.


Assuntos
Doenças Neurodegenerativas/induzido quimicamente , Síndromes Neurotóxicas/complicações , Compostos Organofosforados/toxicidade , Humanos , Síndromes Neurotóxicas/etiologia
7.
Cells ; 11(21)2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36359807

RESUMO

Several reports have shown that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has the potential to also be neurotropic. However, the mechanisms by which SARS-CoV-2 induces neurologic injury, including neurological and/or psychological symptoms, remain unclear. In this review, the available knowledge on the neurobiological mechanisms underlying COVID-19 was organized using the AOP framework. Four AOPs leading to neurological adverse outcomes (AO), anosmia, encephalitis, stroke, and seizure, were developed. Biological key events (KEs) identified to induce these AOs included binding to ACE2, blood-brain barrier (BBB) disruption, hypoxia, neuroinflammation, and oxidative stress. The modularity of AOPs allows the construction of AOP networks to visualize core pathways and recognize neuroinflammation and BBB disruption as shared mechanisms. Furthermore, the impact on the neurological AOPs of COVID-19 by modulating and multiscale factors such as age, psychological stress, nutrition, poverty, and food insecurity was discussed. Organizing the existing knowledge along an AOP framework can represent a valuable tool to understand disease mechanisms and identify data gaps and potentially contribute to treatment, and prevention. This AOP-aligned approach also facilitates synergy between experts from different backgrounds, while the fast-evolving and disruptive nature of COVID-19 emphasizes the need for interdisciplinarity and cross-community research.


Assuntos
Rotas de Resultados Adversos , COVID-19 , Acidente Vascular Cerebral , Humanos , SARS-CoV-2 , Barreira Hematoencefálica
8.
ALTEX ; 39(2): 322­335, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35032963

RESUMO

On April 28-29, 2021, 50 scientists from different fields of expertise met for the 3rd online CIAO workshop. The CIAO project "Modelling the Pathogenesis of COVID-19 using the Adverse Outcome Pathway (AOP) framework" aims at building a holistic assembly of the available scientific knowledge on COVID-19 using the AOP framework. An individual AOP depicts the disease progression from the initial contact with the SARS-CoV-2 virus through biological key events (KE) toward an adverse outcome such as respiratory distress, anosmia or multiorgan failure. Assembling the individual AOPs into a network highlights shared KEs as central biological nodes involved in multiple outcomes observed in COVID-19 patients. During the workshop, the KEs and AOPs established so far by the CIAO members were presented and posi­tioned on a timeline of the disease course. Modulating factors influencing the progression and severity of the disease were also addressed as well as factors beyond purely biological phenomena. CIAO relies on an interdisciplinary crowd­sourcing effort, therefore, approaches to expand the CIAO network by widening the crowd and reaching stakeholders were also discussed. To conclude the workshop, it was decided that the AOPs/KEs will be further consolidated, inte­grating virus variants and long COVID when relevant, while an outreach campaign will be launched to broaden the CIAO scientific crowd.


Assuntos
Rotas de Resultados Adversos , COVID-19 , COVID-19/complicações , Humanos , SARS-CoV-2 , Síndrome de COVID-19 Pós-Aguda
9.
J Proteome Res ; 10(4): 1974-86, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21322648

RESUMO

Increasing evidence points to mitochondrial dysfunction in Parkinson's disease (PD) associated with complex I dysfunction, but the exact pathways which lead to cell death have not been resolved. 2D-gel electrophoresis profiles of isolated mitochondria from neuroblastoma cells treated with subcytotoxic concentrations of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a well-characterized complex I inhibitor, were assessed to identify associated targets. Up to 27 differentially expressed proteins were observed, of which 16 were identified using peptide mass fingerprinting. Changes in protein levels were validated by immunoprobing 1D blots, confirming increases in heat shock cognate 71 kDa (Hsc70), 60 kDa heat shock protein (Hsp60), fumarase, glutamate oxaloacetate transaminase 2, ATP synthase subunit d, and voltage-dependent anion-channel 1 (VDAC1). Immunoprobing of 2D blots revealed isoform changes in Hsc70, Hsp60, and VDAC1. Subcytotoxic concentrations of MPTP modulated a host of mitochondrial proteins including chaperones, metabolic enzymes, oxidative phosphorylation-related proteins, an inner mitochondrial protein (mitofilin), and an outer mitochondrial membrane protein (VDAC1). Early changes in chaperones suggest a regulated link between complex 1 inhibition and protein folding. VDAC1, a multifunctional protein, may have a key role in signaling between mitochondria and the rest of the cell prior to cell death. Our work provides new important information of relevance to PD.


Assuntos
Complexo I de Transporte de Elétrons/antagonistas & inibidores , Mitocôndrias/química , Proteínas Mitocondriais/análise , Neuroblastoma/química , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Biomarcadores/análise , Linhagem Celular Tumoral , Complexo I de Transporte de Elétrons/efeitos dos fármacos , Eletroforese em Gel Bidimensional/métodos , Espectrometria de Massas/métodos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurotoxinas/toxicidade , Proteoma/análise , Proteômica/métodos
10.
Biochem J ; 429(2): 261-71, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20441570

RESUMO

An extracellular form of the calcium-dependent protein-cross-linking enzyme TGase (transglutaminase) was demonstrated to be involved in the apical growth of Malus domestica pollen tube. Apple pollen TGase and its substrates were co-localized within aggregates on the pollen tube surface, as determined by indirect immunofluorescence staining and the in situ cross-linking of fluorescently labelled substrates. TGase-specific inhibitors and an anti-TGase monoclonal antibody blocked pollen tube growth, whereas incorporation of a recombinant fluorescent mammalian TGase substrate (histidine-tagged green fluorescent protein: His6-Xpr-GFP) into the growing tube wall enhanced tube length and germination, consistent with a role of TGase as a modulator of cell wall building and strengthening. The secreted pollen TGase catalysed the cross-linking of both PAs (polyamines) into proteins (released by the pollen tube) and His6-Xpr-GFP into endogenous or exogenously added substrates. A similar distribution of TGase activity was observed in planta on pollen tubes germinating inside the style, consistent with a possible additional role for TGase in the interaction between the pollen tube and the style during fertilization.


Assuntos
Malus/enzimologia , Transglutaminases/metabolismo , Espaço Extracelular/enzimologia , Germinação , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Malus/crescimento & desenvolvimento , Modelos Biológicos , Plantas Geneticamente Modificadas , Tubo Polínico/enzimologia , Tubo Polínico/crescimento & desenvolvimento , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Transglutaminases/antagonistas & inibidores
11.
Sci Rep ; 10(1): 2767, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-32066745

RESUMO

Current therapeutic strategies for Parkinson's disease (PD) aim to delay progression or replace damaged neurons by restoring the original neuronal structures. The poor regenerative capacity of neural tissue highlights the need for the development of cellular environments to model the pathogenesis of PD. In the current work, we have characterised the growth, survival and response to PD mimetics of human SH-SY5Y neuroblastoma and U-87MG glioblastoma cell lines cultured on polyacrylonitrile (PAN) and Jeffamine® doped polyacrylonitrile (PJ) nano-scaffolds. Differentiation induced by a range of agents was evaluated by immunoassays of neural protein biomarkers. PAN and PJ nanofibre scaffolds provided suitable three-dimensional (3D) environment to support the growth, differentiation and network formation of dopaminergic neuron- and astrocyte-like cell populations, respectively. The scaffolds selectively supported the survival and differentiation of both cell populations with prolonged neuronal survival when exposed to PD mimetics in the presence of astrocytes in a co-culture model. Such 3D nanoscaffold-based assays could aid our understanding of the molecular basis of PD mimetic-induced Parkinsonism and the discovery of neuroprotective agents.


Assuntos
Resinas Acrílicas/farmacologia , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , Astrócitos/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Humanos , Nanofibras/química , Degeneração Neural/patologia , Fármacos Neuroprotetores/química , Oxidopamina/farmacologia , Doença de Parkinson/patologia , Alicerces Teciduais
12.
Arch Toxicol ; 83(4): 373-80, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18633600

RESUMO

The aim of this study was to assess the neurotoxicity of diazinon oxon (DZO), a major in vivo metabolite of the phosphorothionate insecticide diazinon (DZ), on differentiating mouse N2a neuroblastoma cells. When used at concentrations of 1, 5 and 10 microM, DZO did not cause cell death but it impaired the outgrowth of axon-like processes after 24 h. Densitometric scanning of Western blots of lysates of N2a cells revealed that exposure to 5 or 10 microM DZO for 24 h increased the expression of phosphorylated neurofilament heavy chain (NFH) compared to controls, while there was no significant change in total NFH. By contrast, treatment of N2a cells with 1-10 microM DZO resulted in marked reductions in the expression of the axon growth-associated protein GAP-43. DZO-treated cells also showed an increased expression of the heat shock protein HSP-70 compared to controls. The above biochemical changes were not temporally related to inhibition of acetylcholinesterase (AChE). These data suggest that biologically relevant, subcytotoxic levels of DZO may exert neurotoxic effects on differentiating cells and that the mechanisms involved are different from those attributed to its parent compound.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Diazinon/análogos & derivados , Diazinon/toxicidade , Inseticidas/toxicidade , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Proteína GAP-43/efeitos dos fármacos , Proteína GAP-43/metabolismo , Proteínas de Choque Térmico HSP70/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/metabolismo , Camundongos , Neuroblastoma , Proteínas de Neurofilamentos/efeitos dos fármacos , Proteínas de Neurofilamentos/metabolismo , Neurônios/metabolismo , Neurônios/patologia
13.
Eur J Pharmacol ; 820: 113-129, 2018 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-29242118

RESUMO

NGF (nerve growth factor) and tissue transglutaminase (TG2) play important roles in neurite outgrowth and modulation of neuronal cell survival. In this study, we investigated the regulation of TG2 transamidase activity by NGF in retinoic acid-induced differentiating mouse N2a and human SH-SY5Y neuroblastoma cells. TG2 transamidase activity was determined using an amine incorporation and a peptide cross linking assay. In situ TG2 activity was assessed by visualising the incorporation of biotin-X-cadaverine using confocal microscopy. The role of TG2 in NGF-induced cytoprotection and neurite outgrowth was investigated by monitoring hypoxia-induced cell death and appearance of axonal-like processes, respectively. The amine incorporation and protein crosslinking activity of TG2 increased in a time and concentration-dependent manner following stimulation with NGF in N2a and SH-SY5Y cells. NGF mediated increases in TG2 activity were abolished by the TG2 inhibitors Z-DON (Z-ZON-Val-Pro-Leu-OMe; Benzyloxycarbonyl-(6-Diazo-5-oxonorleucinyl)-l-valinyl-l-prolinyl-l-leucinmethylester) and R283 (1,3,dimethyl-2[2-oxo-propyl]thio)imidazole chloride) and by pharmacological inhibition of extracellular signal-regulated kinases 1 and 2 (ERK1/2), protein kinase B (PKB) and protein kinase C (PKC), and removal of extracellular Ca2+. Fluorescence microscopy demonstrated NGF induced in situ TG2 activity. TG2 inhibition blocked NGF-induced attenuation of hypoxia-induced cell death and neurite outgrowth in both cell lines. Together, these results demonstrate that NGF stimulates TG2 transamidase activity via a ERK1/2, PKB and PKC-dependent pathway in differentiating mouse N2a and human SH-SY5Y neuroblastoma cells. Furthermore, NGF-induced cytoprotection and neurite outgrowth are dependent upon TG2. These results suggest a novel and important role of TG2 in the cellular functions of NGF.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neuroblastoma/patologia , Transglutaminases/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína 2 Glutamina gama-Glutamiltransferase , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
14.
Biochem Pharmacol ; 128: 55-73, 2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28065858

RESUMO

The PAC1 receptor and tissue transglutaminase (TG2) play important roles in neurite outgrowth and modulation of neuronal cell survival. In this study, we investigated the regulation of TG2 activity by the PAC1 receptor in retinoic acid-induced differentiating N2a neuroblastoma cells. TG2 transamidase activity was determined using an amine incorporation and a peptide cross linking assay. In situ TG2 activity was assessed by visualising the incorporation of biotin-X-cadaverine using confocal microscopy. TG2 phosphorylation was monitored via immunoprecipitation and Western blotting. The role of TG2 in PAC1 receptor-induced cytoprotection and neurite outgrowth was investigated by monitoring hypoxia-induced cell death and appearance of axonal-like processes, respectively. The amine incorporation and protein crosslinking activity of TG2 increased in a time and concentration-dependent manner following stimulation with pituitary adenylate cyclase-activating polypeptide-27 (PACAP-27). PACAP-27 mediated increases in TG2 activity were abolished by the TG2 inhibitors Z-DON and R283 and by pharmacological inhibition of protein kinase A (KT 5720 and Rp-cAMPs), protein kinase C (Ro 31-8220), MEK1/2 (PD 98059), and removal of extracellular Ca2+. Fluorescence microscopy demonstrated PACAP-27 induced in situ TG2 activity. TG2 inhibition blocked PACAP-27 induced attenuation of hypoxia-induced cell death and outgrowth of axon-like processes. TG2 activation and cytoprotection were also observed in human SH-SY5Y cells. Together, these results demonstrate that TG2 activity was stimulated downstream of the PAC1 receptor via a multi protein kinase dependent pathway. Furthermore, PAC1 receptor-induced cytoprotection and neurite outgrowth are dependent upon TG2. These results highlight the importance of TG2 in the cellular functions of the PAC1 receptor.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Neuritos/fisiologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Transglutaminases/metabolismo , Animais , Cálcio/metabolismo , Morte Celular , Diferenciação Celular , Hipóxia Celular , Linhagem Celular Tumoral , Neurônios Colinérgicos/fisiologia , Neurônios Colinérgicos/ultraestrutura , Proteínas de Ligação ao GTP/antagonistas & inibidores , Humanos , Camundongos , Fosforilação , Proteína 2 Glutamina gama-Glutamiltransferase , Inibidores de Proteínas Quinases/farmacologia , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/agonistas , Transglutaminases/antagonistas & inibidores
15.
Eur J Pharmacol ; 813: 105-121, 2017 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-28754379

RESUMO

Tissue transglutaminase 2 (TG2) is modulated by protein kinase A (PKA) mediated phosphorylation: however, the precise mechanism(s) of its modulation by G-protein coupled receptors coupled to PKA activation are not fully understood. In the current study we investigated the potential regulation of TG2 activity by the ß2-adrenoceptor in rat H9c2 cardiomyoblasts. Transglutaminase transamidation activity was assessed using amine-incorporating and protein cross-linking assays. TG2 phosphorylation was determined via immunoprecipitation and Western blotting. The long acting ß2-adrenoceptor agonist formoterol induced time- and concentration-dependent increases in TG2 transamidation. Increases in TG2 activity were reduced by the TG2 inhibitors Z-DON (Benzyloxycarbonyl-(6-Diazo-5-oxonorleucinyl)-L-valinyl-L-prolinyl-L-leucinmethylester) and R283 ((1,3,dimethyl-2[2-oxo-propyl]thio)imidazole chloride). Responses to formoterol were blocked by pharmacological inhibition of PKA, extracellular signal-regulated kinase 1 and 2 (ERK1/2), or phosphatidylinositol 3-kinase (PI-3K) signalling. Furthermore, the removal of extracellular Ca2+ also attenuated formoterol-induced TG2 activation. Fluorescence microscopy demonstrated TG2-induced biotin-X-cadaverine incorporation into proteins. Formoterol increased the levels of TG2-associated phosphoserine and phosphothreonine, which were blocked by inhibition of PKA, ERK1/2 or PI-3K signalling. Subsequent proteomic analysis identified known (e.g. lactate dehydrogenase A chain) and novel (e.g. Protein S100-A6) protein substrates for TG2. Taken together, the data obtained suggest that ß2-adrenoceptor-induced modulation of TG2 represents a novel paradigm in ß2-adrenoceptor cell signalling, expanding the repertoire of cellular functions responsive to catecholamine stimulation.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transglutaminases/metabolismo , Animais , Cálcio/metabolismo , Catecolaminas/farmacologia , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/enzimologia , Fosforilação/efeitos dos fármacos , Proteína 2 Glutamina gama-Glutamiltransferase , Ratos , Transdução de Sinais/efeitos dos fármacos , Transglutaminases/antagonistas & inibidores
16.
Biochem Pharmacol ; 71(8): 1240-7, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16499876

RESUMO

Sub-lethal concentrations of the organophosphate phenyl saligenin phosphate (PSP) inhibited the outgrowth of axon-like processes in differentiating mouse N2a neuroblastoma cells (IC(50) 2.5 microM). A transient rise in the phosphorylation state of neurofilament heavy chain (NFH) was detected on Western blots of cell extracts treated with 2.5 microM PSP for 4 h compared to untreated controls, as determined by a relative increase in reactivity with monoclonal antibody Ta51 (anti-phosphorylated NFH) compared to N52 (anti-total NFH). However, cross-reactivity of PSP-treated cell extracts was lower than that of untreated controls after 24 h exposure, as indicated by decreased reactivity with both antibodies. Indirect immunofluorescence analysis with these antibodies revealed the appearance of neurofilament aggregates in the cell bodies of treated cells and reduced axonal staining compared to controls. By contrast, there was no significant change in reactivity with anti-alpha-tubulin antibody B512 at either time point. The activation state of the MAP kinase ERK 1/2 increased significantly after PSP treatment compared to controls, particularly at 4 h, as indicated by increased reactivity with monoclonal antibody E-4 (anti-phosphorylated MAP kinase) but not with polyclonal antibody K-23 (anti-total MAP kinase). The observed early changes were concomitant with almost complete inhibition of the activity of neuropathy target esterase (NTE), one of the proposed early molecular targets in organophosphate-induced delayed neuropathy (OPIDN).


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Diferenciação Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neuritos/efeitos dos fármacos , Proteínas de Neurofilamentos/metabolismo , Compostos Organofosforados/farmacologia , Animais , Western Blotting , Linhagem Celular Tumoral , Camundongos , Neuritos/enzimologia , Neuritos/patologia , Neuroblastoma/patologia , Fosforilação
17.
Environ Toxicol Pharmacol ; 22(1): 70-4, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21783689

RESUMO

Phenyl saligenin phosphate (PSP) induces a characteristic neuropathy (OPIDN), the molecular basis of which has not been precisely defined. This study examined the in vitro effects of PSP on the phosphorylation of serine and threonine residues of proteins in porcine brain cytosol. Quantitative analysis of Western blots probed with antibodies recognizing phosphorylated serine residues demonstrated that 100µM PSP induced a significant increase in the phosphorylation of serine residues of a 50kDa protein. This protein was identified as the α- and ß-tubulin subunits by probing Western blots of extracts separated by two-dimensional polyacrylamide gel electrophoresis with anti-phosphoserine and anti-tubulin antibodies. By contrast, threonine phosphorylation of the 50kDa polypeptide and other proteins detected on Western blots probed with anti-phosphothreonine antibodies, was not significantly affected by PSP. These data indicate that PSP is able to induce increased phosphorylation of tubulin in serine residues, consistent with a possible role for this phenomenon in OPIDN induction.

18.
Toxicol In Vitro ; 32: 154-65, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26724415

RESUMO

The aims of this study were to examine the effects of CdCl2 on the viability, migration and cytoskeleton of cultured mouse TM4 Sertoli cells. Time- and concentration-dependent changes were exhibited by the cells but 1 µM CdCl2 was sub-cytotoxic at all time-points. Exposure to 1 and 12 µM CdCl2 for 4 h resulted in disruption of the leading edge, as determined by chemical staining. Cell migration was inhibited by both 1 and 12 µM CdCl2 in a scratch assay monitored by live cell imaging, although exposure to the higher concentration was associated with cell death. Western blotting and immunofluorescence staining indicated that CdCl2 caused a concentration dependent reduction in actin and tubulin levels. Exposure to Cd(2+) also resulted in significant changes in the levels and/or phosphorylation status of the microtubule and microfilament destabilising proteins cofilin and stathmin, suggesting disruption of cytoskeletal dynamics. Given that 1-12 µM Cd(2+) is attainable in vivo, our findings are consistent with the possibility that Cd(2+) induced impairment of testicular development and reproductive health may involve a combination of reduced Sertoli cell migration and impaired Sertoli cell viability depending on the timing, level and duration of exposure.


Assuntos
Cloreto de Cádmio/toxicidade , Movimento Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Células de Sertoli/efeitos dos fármacos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Masculino , Camundongos , Células de Sertoli/metabolismo , Células de Sertoli/fisiologia
19.
Biochem Pharmacol ; 107: 41-58, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-27005940

RESUMO

The regulation of tissue transglutaminase (TG2) activity by the GPCR family is poorly understood. In this study, we investigated the modulation of TG2 activity by the A1 adenosine receptor in cardiomyocyte-like H9c2 cells. H9c2 cells were lysed following stimulation with the A1 adenosine receptor agonist N(6)-cyclopentyladenosine (CPA). Transglutaminase activity was determined using an amine incorporating and a protein cross linking assay. TG2 phosphorylation was assessed via immunoprecipitation and Western blotting. The role of TG2 in A1 adenosine receptor-induced cytoprotection was investigated by monitoring hypoxia-induced cell death. CPA induced time and concentration-dependent increases in amine incorporating and protein crosslinking activity of TG2. CPA-induced increases in TG2 activity were attenuated by the TG2 inhibitors Z-DON and R283. Responses to CPA were blocked by PKC (Ro 31-8220), MEK1/2 (PD 98059), p38 MAPK (SB 203580) and JNK1/2 (SP 600125) inhibitors and by removal of extracellular Ca(2+). CPA triggered robust increases in the levels of TG2-associated phosphoserine and phosphothreonine, which were attenuated by PKC, MEK1/2 and JNK1/2 inhibitors. Fluorescence microscopy revealed TG2-mediated biotin-X-cadaverine incorporation into proteins and proteomic analysis identified known (Histone H4) and novel (Hexokinase 1) protein substrates for TG2. CPA pre-treatment reversed hypoxia-induced LDH release and decreases in MTT reduction. TG2 inhibitors R283 and Z-DON attenuated A1 adenosine receptor-induced cytoprotection. TG2 activity was stimulated by the A1 adenosine receptor in H9c2 cells via a multi protein kinase dependent pathway. These results suggest a role for TG2 in A1 adenosine receptor-induced cytoprotection.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Proteínas de Ligação ao GTP/agonistas , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mioblastos Cardíacos/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Proteínas de Ligação ao GTP/metabolismo , Hexoquinase/metabolismo , Histonas/metabolismo , Cinética , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Fosforilação/efeitos dos fármacos , Proteína 2 Glutamina gama-Glutamiltransferase , Inibidores de Proteínas Quinases/farmacologia , Ratos , Receptor A1 de Adenosina/sangue , Especificidade por Substrato , Transglutaminases/antagonistas & inibidores , Transglutaminases/metabolismo
20.
Toxicol Mech Methods ; 15(6): 405-10, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-20021063

RESUMO

The aim of this work was to study the neurodegenerative effects of the organophosphate (OP) pesticides chlorpyrifos (CPF) and chlorpyrifos-methyl (CHM) on cultured mouse N2a neuroblastoma cells. CPF or CHM, at a subcytotoxic concentration of 3 muM, were added to the cells either at the time of the induction of cell differentiation (codifferentiation) or 16 h after the induction of differentiation (postdifferentiation). CPF and CHM were similar in inhibiting significantly the outgrowth of axon-like processes from N2a cells after only 4 h exposure under both co- and postdifferentiation exposure conditions. Densitometric scanning of Western blots of extracts of cells treated with CPF or CHM for 4 h revealed significantly decreased cross-reactivity with a monoclonal antibody recognizing the protein GAP-43 under post- but not under codifferentiation exposure conditions. Exposure to CPF or CHM for 4 h under postdifferentiation conditions also resulted in reduced fluorescence of N2a cell body staining with anti-GAP-43. Cross-reactivity of Western blots with a monoclonal antibody recognizing alpha-tubulin was not significantly affected by OP treatment. These data indicate that a disturbance in GAP-43 may be important in the retraction of axons in predifferentiated N2a cells and support the notion that the mechanisms involved in CPF-and CHM-induced inhibition of axonal outgrowth may be different under co- and postdifferentiation exposure conditions.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA