Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 837
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Anaerobe ; 65: 102265, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32860931

RESUMO

Clostridium perfringens type A is the causative agent of clostridial myonecrosis, and α-toxin has been reported to be responsible for the pathogenesis. Recently, it was reported that regeneration of skeletal muscle after C. perfringens-induced muscle disorders is delayed, but the detailed mechanisms have not been elucidated. Here, we tested whether α-toxin impairs the differentiation of C2C12 myoblasts, a useful cell line to study muscle growth, maturation, and regeneration in vitro. α-Toxin dose-dependently inhibited myotube formation in C2C12 cultures after induction of their differentiation by horse serum. Also, immunoblot analysis revealed that α-toxin dose-dependently decreases the expressions of two skeletal muscle differentiation markers, myogenic differentiation 1 (MyoD) and myogenin. These results demonstrate that α-toxin impairs the myogenic differentiation of C2C12 myoblasts. To reveal the mechanism behind α-toxin-mediated impairment of myogenic differentiation, we focused on ceramide production since α-toxin is known to promote the formation of ceramide by its sphingomyelinase activity. Immunofluorescent analysis revealed that ceramide production is accelerated by treatment with α-toxin. Furthermore, a synthetic cell-permeable ceramide analog, C2-ceramide, inhibited myotube formation in C2C12 cells and decreased the expressions of MyoD and myogenin, suggesting that accelerated ceramide production is involved in the α-toxin-mediated blockage of myogenic differentiation. Together, our results illustrate that the impairment of myogenic differentiation by α-toxin might be crucial for the pathogenesis of C. perfringens to delay regeneration of severely damaged skeletal muscles.


Assuntos
Toxinas Bacterianas/farmacologia , Proteínas de Ligação ao Cálcio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Fosfolipases Tipo C/farmacologia , Animais , Biomarcadores , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Camundongos , Desenvolvimento Muscular , Proteína MyoD/metabolismo , Mioblastos/metabolismo , Miogenina/metabolismo
2.
Exp Parasitol ; 204: 107731, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31374185

RESUMO

Neospora caninum is an obligate intracellular parasite related to cases of abortion and fertility impairment in cattle. The control of the parasite still lacks an effective protective strategy and the understanding of key mechanisms for host infection might be crucial for identification of specific targets. There are many proteins related to important mechanisms in the host cell infection cycle such as adhesion, invasion, proliferation and immune evasion. The surface proteins, especially SRS (Surface Antigen Glycoprotein - Related Sequences), have been demonstrated to have a pivotal role in the adhesion and invasion processes, making them potential anti-parasite targets. However, several predicted surface proteins were not described concerning their function and importance in the parasite life cycle. As such, a novel SRS protein, NcSRS57, was described. NcSRS57 antiserum was used to detect SRS proteins by immunofluorescence in parasites treated or not with phosphatidylinositol-specific phospholipase C (PI-PLC). The treatment with PI-PLC also allowed the identification of NcSRS29B and NcSRS29C, which were the most abundant SRS proteins in the soluble fraction. Our data indicated that SRS proteins in N. caninum shared a high level of sequence similarity and were susceptible to PI-PLC. In addition, the description of the SRS members, regarding abundance, function and immunogenicity will be useful in guiding specific methods to control the mechanism of adhesion and invasion mediated by these surface proteins.


Assuntos
Antígenos de Protozoários/metabolismo , Antígenos de Superfície/metabolismo , Neospora/química , Fosfoinositídeo Fosfolipase C/farmacologia , Proteínas de Protozoários/metabolismo , Animais , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Chlorocebus aethiops , Clonagem Molecular , DNA de Protozoário/isolamento & purificação , Eletroforese em Gel Bidimensional , Eletroforese em Gel de Poliacrilamida , Soros Imunes/imunologia , Soros Imunes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Neospora/efeitos dos fármacos , Neospora/genética , Neospora/imunologia , Fosfoinositídeo Fosfolipase C/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Espectrometria de Massas em Tandem , Fosfolipases Tipo C/metabolismo , Fosfolipases Tipo C/farmacologia , Células Vero
3.
Microb Pathog ; 118: 1-8, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29524545

RESUMO

We previously developed a stable and marker-free Lactobacillus casei strain (PPαT Δupp) that contained a chromosomally integrated expression cassette (PPαT) that enabled the surface expression of the Clostridium perfringens alpha toxin. To measure immune responses against the alpha toxin, specific-pathogen-free BALB/c mice were inoculated with L. casei PPαT Δupp by oral gavage. Then, specific immunoglobulin A (IgA) and immunoglobulin G (IgG) antibodies and cytokines were measured by enzyme-linked immunosorbent assay (ELISA) and flow cytometry (FCM). The results showed that alpha toxin-specific IgA and IgG antibodies and cytokines were markedly increased following immunization. Natural alpha toxin challenge and neutralization tests were performed. The results showed that immunized mice can fully resist 1.5 minimum lethal doses of toxin. These results indicated that the immunized mice can produce not only humoral immunity, but also cellular immunity. These results provide a new pathway for the development of a safe, effective, and food-grade vaccine.


Assuntos
Toxinas Bacterianas/biossíntese , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/farmacologia , Vacinas Bacterianas/imunologia , Vacinas Bacterianas/farmacologia , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/imunologia , Proteínas de Ligação ao Cálcio/farmacologia , Imunização , Lacticaseibacillus casei/imunologia , Lacticaseibacillus casei/metabolismo , Fosfolipases Tipo C/biossíntese , Fosfolipases Tipo C/imunologia , Fosfolipases Tipo C/farmacologia , Administração Oral , Animais , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/imunologia , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/genética , Vacinas Bacterianas/genética , Proteínas de Ligação ao Cálcio/genética , Proliferação de Células , Infecções por Clostridium/imunologia , Infecções por Clostridium/microbiologia , Infecções por Clostridium/prevenção & controle , Clostridium perfringens/genética , Clostridium perfringens/imunologia , Citocinas/sangue , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Regulação Bacteriana da Expressão Gênica , Vetores Genéticos , Instabilidade Genômica , Imunidade Celular , Imunidade Humoral , Imunoglobulina A/análise , Imunoglobulina A/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Lacticaseibacillus casei/genética , Dose Letal Mediana , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Organismos Livres de Patógenos Específicos , Fosfolipases Tipo C/genética , Vacinação , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
4.
Biol Pharm Bull ; 39(10): 1694-1700, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27725448

RESUMO

Clostridium perfringens type A, a Gram-positive, anaerobic bacterium, causes gas gangrene. Recently, we reported that C. perfringens α-toxin blocked neutrophil differentiation in an enzyme activity-dependent manner to impair host innate immunity, which should be crucial for the pathogenesis of C. perfringens. However, the detailed mechanism remains unclear. Lipid rafts have been reported to be platforms for signaling molecules involved in the regulation of cell differentiation in many different cell types. In this study, we found that cell surface expression of a lipid raft marker, GM1 ganglioside, decreased in association with neutrophil differentiation by flow cytometry analysis and morphological observation. In vitro treatment of isolated mouse bone marrow cells with α-toxin or an α-toxin variant lacking phospholipase C and sphingomyelinase activities revealed that α-toxin increased the cell surface expression of GM1 ganglioside in an enzyme activity-dependent manner. C. perfringens infection also increased GM1 ganglioside levels in bone marrow myeloid cells. Moreover, treatment of bone marrow cells with methyl-ß-cyclodextrin, a lipid raft-disrupting agent, impaired neutrophil differentiation. Together, our results suggest that the integrity of lipid rafts should be properly maintained during granulopoiesis, and α-toxin might perturb lipid raft integrity leading to the impairment of neutrophil differentiation.


Assuntos
Toxinas Bacterianas/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Proteínas de Ligação ao Cálcio/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Fosfolipases Tipo C/farmacologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Gangliosídeo G(M1)/metabolismo , Camundongos Endogâmicos C57BL , Neutrófilos/citologia , Neutrófilos/metabolismo , beta-Ciclodextrinas/farmacologia
5.
Biochim Biophys Acta ; 1822(10): 1581-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22721959

RESUMO

A characteristic feature of gas gangrene with Clostridium perfringens (C. perfringens) is the absence of neutrophils within the infected area and the massive accumulation of neutrophils at the vascular endothelium around the margins of the necrotic region. Intravenous injection of C. perfringens alpha-toxin into mice resulted in the accumulation of neutrophils at the vascular endothelium in lung and liver, and release of GRO/KC, a member of the CXC chemokine family with homology to human interleukin-8 (IL-8). Alpha-toxin triggered activation of signal transduction pathways causing mRNA expression and production of IL-8, which activates migration and binding of neutrophils, in A549 cells. K252a, a tyrosine kinase A (TrkA) inhibitor, and siRNA for TrkA inhibited the toxin-induced phosphorylation of TrkA and production of IL-8. In addition, K252a inhibited the toxin-induced phosphorylation of extracellular regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase (MAPK). PD98059, an ERK1/2 inhibitor, depressed phosphorylation of ERK1/2 and nuclear translocation of nuclear factor kappa B (NF-κB) p65, but SB203580, a p38 MAPK inhibitor, did not. On the other hand, PD98059 and SB203580 suppressed the toxin-induced production of IL-8. Treatment of the cells with PD98059 resulted in inhibition of IL-8 mRNA expression induced by the toxin and that with SB203580 led to a decrease in the stabilization of IL-8 mRNA. These results suggest that alpha-toxin induces production of IL-8 through the activation of two separate pathways, the ERK1/2/NF-κB and p38 MAPK pathways.


Assuntos
Toxinas Bacterianas/farmacologia , Proteínas de Ligação ao Cálcio/farmacologia , Interleucina-8/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fosfolipases Tipo C/farmacologia , Animais , Carbazóis/farmacologia , Linhagem Celular Tumoral , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Alcaloides Indólicos/farmacologia , Interleucina-8/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , NF-kappa B/genética , NF-kappa B/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , RNA Mensageiro/genética , Transdução de Sinais/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Nihon Rinsho ; 70(8): 1313-7, 2012 Aug.
Artigo em Japonês | MEDLINE | ID: mdl-22894064

RESUMO

Clostridium perfringens causes gas gangrene with inflammatory myopathies and infrequently septicemia associated with massive intravascular hemolysis. The microorganism is known to produce a variety of toxins and enzymes that are responsible for severe myonecrotic lesions. Notably, alpha-toxin, which possesses hemolytic, necrotic and lethal activities, and phospholipase C and sphingomyelinase activities, is an important agent for the diseases. The cytokine storm induced by alpha-toxin, mainly the release of TNF-alpha, plays an important role in the death and massive hemolysis. The toxin-induced release of TNF-alpha from neutrophils and macrophages is dependent on the activation of ERK1/2 signal transduction via TrkA receptor. In addition, 14- and 15-membered macrolides specifically block the toxin-induced events through the activation of neutrophils and macrophages.


Assuntos
Toxinas Bacterianas , Proteínas de Ligação ao Cálcio , Clostridium perfringens/patogenicidade , Gangrena Gasosa/microbiologia , Fosfolipases Tipo C , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Toxinas Bacterianas/efeitos adversos , Toxinas Bacterianas/farmacologia , Proteínas de Ligação ao Cálcio/efeitos adversos , Proteínas de Ligação ao Cálcio/farmacologia , Gangrena Gasosa/tratamento farmacológico , Hemólise , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Macrolídeos/farmacologia , Macrolídeos/uso terapêutico , Macrófagos/metabolismo , Neutrófilos/metabolismo , Fosforilação/efeitos dos fármacos , Receptor trkA/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Fosfolipases Tipo C/efeitos adversos , Fosfolipases Tipo C/farmacologia
7.
Neurotox Res ; 40(3): 791-802, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35438391

RESUMO

Chlorpromazine, an antipsychotic medication, is conventionally applied to cope with the psychotic disorder such as schizophrenia. In cellular studies, chlorpromazine exerts many different actions through calcium ion (Ca2+) signaling, but the underlying pathways are elusive. This study explored the effect of chlorpromazine on viability, Ca2+ signaling pathway and their relationship in glial cell models (GBM 8401 human glioblastoma cell line and Gibco® Human Astrocyte (GHA)). First, chlorpromazine between 10 and 40 µM induced cytotoxicity in GBM 8401 cells but not in GHA cells. Second, in terms of Ca2+ homeostasis, chlorpromazine (10-30 µM) increased intracellular Ca2+ concentrations ([Ca2+]i) rises in GBM 8401 cells but not in GHA cells. Ca2+ removal reduced the signal by approximately 55%. Furthermore, chelation of cytosolic Ca2+ with BAPTA-AM reduced chlorpromazine (10-40 µM)-induced cytotoxicity in GBM 8401 cells. Third, in Ca2+-containing medium of GBM 8401 cells, chlorpromazine-induced Ca2+ entry was inhibited by the modulators of store-operated Ca2+ channel (2-APB and SKF96365). Lastly, in Ca2+-free medium of GBM 8401 cells, treatment with the endoplasmic reticulum Ca2+ pump inhibitor thapsigargin completely inhibited chlorpromazine-increased [Ca2+]i rises. Conversely, treatment with chlorpromazine abolished thapsigargin-increased [Ca2+]i rises. Inhibition of phospholipase C (PLC) with U73122 abolished chlorpromazine-increased [Ca2+]i rises. Together, in GBM 8401 cells but not in GHA cells, chlorpromazine increased [Ca2+]i rises by Ca2+ influx via store-operated Ca2+ entry and PLC-dependent Ca2+ release from the endoplasmic reticulum. Moreover, the Ca2+ chelator BAPTA-AM inhibited cytotoxicity in chlorpromazine-treated GBM 8401 cells. Therefore, Ca2+ signaling was involved in chlorpromazine-induced cytotoxicity in GBM 8401 cells.


Assuntos
Antipsicóticos , Sinalização do Cálcio , Antipsicóticos/toxicidade , Apoptose , Cálcio/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Quelantes , Clorpromazina/farmacologia , Humanos , Neuroglia/metabolismo , Tapsigargina/farmacologia , Fosfolipases Tipo C/metabolismo , Fosfolipases Tipo C/farmacologia
8.
Biochem Biophys Res Commun ; 411(2): 241-6, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21740889

RESUMO

Alpha-toxin-induced phosphorylation of PDK1 via the tyrosine kinase A (TrkA) receptor signaling pathway plays an important role in the activation of rabbit neutrophils. The relation between the toxin and TrkA, however, remains poorly understood. Here, we show that the toxin-induced phosphorylation of TrkA is closely related to the induction of neurite-outgrowth in PC12 cells. The toxin induced neurite-outgrowth and phosphorylation of TrkA in the cells in a dose-dependent manner. K252a, a TrkA inhibitor, and shRNA for TrkA inhibited the toxin-induced neurite-outgrowth, and phosphorylation of TrkA and ERK1/2. PD98059, an inhibitor of the ERK1/2 cascade, inhibited phosphorylation of ERK1/2 and the neurite-outgrowth induced by alpha-toxin. The wild-type toxin induced the formation of diacylglycerol, and neurite-outgrowth, but H148G, a variant toxin which binds to cell membranes and has lost the enzymatic activity did not. We demonstrated that the phosphorylation of TrkA through the phospholipid metabolism induced by the toxin synergistically play a key role in neurite-outgrowth.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Neuritos/fisiologia , Fosfolipídeos/metabolismo , Receptor trkA/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Toxinas Bacterianas/farmacologia , Proteínas de Ligação ao Cálcio/farmacologia , Flavonoides/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Neuritos/efeitos dos fármacos , Neuritos/enzimologia , Células PC12 , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/genética , Ratos , Receptor trkA/genética , Fosfolipases Tipo C/farmacologia
9.
Cereb Cortex ; 20(4): 982-96, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19666830

RESUMO

Cholinergic neurotransmission in the medial prefrontal cortex (mPFC) is critical for normal processing of cue detection and cognitive performance. However, the mechanism by which cholinergic system modifies mPFC synaptic function remains unclear. Here we show that activation of muscarinic acetylcholine receptors (mAChRs) by carbamoylcholine (CCh) induces long-term depression (CCh-LTD) of excitatory synaptic transmission on mPFC layer V pyramidal neurons. The induction of CCh-LTD is dependent on M(1) mAChR activation but does not require N-methyl-D-aspartate receptor activation or coincident synaptic stimulation. Activation of phospholipase C (PLC), protein kinase C (PKC), and postsynaptic Ca(2+) release from inositol 1,4,5-triphosphate (IP(3)) receptor-sensitive internal stores are required for CCh-LTD induction. The expression of CCh-LTD is likely to be presynaptic because it is accompanied by a decrease in 1/(coefficient of variance)(2) and an increase in synaptic failure and paired-pulse ratio of synaptic responses. CCh-LTD is blocked by nitric oxide (NO) synthase inhibitors, soluble guanylyl cyclase (sGC) inhibitor, and protein kinase G (PKG) inhibitor. Synaptic stimulation of M(1) mAChRs with prolonged paired-pulse low-frequency stimulation also triggers LTD. These results suggest that activation of M(1) mAChRs can induce LTD on mPFC layer V pyramidal neurons through the activation of postsynaptic PLC/PKC/IP(3) receptor- and subsequently presynaptic NO/sGC/PKG-dependent signaling processes.


Assuntos
Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/fisiologia , Neurotransmissores/farmacologia , Óxido Nítrico/farmacologia , Córtex Pré-Frontal/fisiologia , Receptores Muscarínicos/metabolismo , Animais , Biofísica , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Interações Medicamentosas/fisiologia , Estimulação Elétrica/métodos , Inibidores Enzimáticos/farmacologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Técnicas In Vitro , Inositol 1,4,5-Trifosfato/farmacologia , Masculino , Técnicas de Patch-Clamp/métodos , Córtex Pré-Frontal/citologia , Proteína Quinase C/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fosfolipases Tipo C/farmacologia
10.
Lett Appl Microbiol ; 53(2): 238-43, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21671964

RESUMO

AIMS: To investigate the influence of subinhibitory concentrations of luteolin on the production of α-toxin in Staphylococcus aureus. METHODS AND RESULTS: The minimal inhibitory concentrations (MICs) were determined using a broth microdilution method, and the MICs of luteolin against the tested Staph. aureus strains ranged from 16 to 64 µg ml(-1). Haemolysis, Western blot and real-time reverse transcription (RT)-PCR assays were used to evaluate the effect of luteolin on Staph. aureusα-toxin secretion and on the level of gene expression, respectively. The data indicated that subinhibitory concentrations of luteolin dose dependently decreased the production of α-toxin in both meticillin-sensitive Staph. aureus (MSSA) and meticillin-resistant Staph. aureus (MRSA). Furthermore, the transcriptional levels of agr (accessory gene regulator) in Staph. aureus were also inhibited by luteolin. CONCLUSIONS: Luteolin decreases the production and/or secretion of α-toxin in Staph. aureus; the reduced production may be dependent, in part, upon the luteolin-induced inhibition of the agr locus. SIGNIFICANCE AND IMPACT OF THE STUDY: The findings indicate that luteolin may be used as a basis for the development of antimicrobial agents aimed at bacterial virulence factors.


Assuntos
Anti-Infecciosos/farmacologia , Toxinas Bacterianas/biossíntese , Luteolina/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Anti-Infecciosos/química , Luteolina/química , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/metabolismo , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo , Fosfolipases Tipo C/farmacologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Fatores de Virulência/farmacologia
11.
J Exp Med ; 164(4): 1338-43, 1986 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-3489808

RESUMO

The mechanism by which the rat T cell alloantigen, RT-6.2, is attached to the membrane was investigated. Treatment of rat lymph node and T-hybridoma cells with phosphatidylinositol-specific phospholipase C (PI-PLC) caused a substantial reduction in the amount of RT-6.2 on the cell surface. No significant release of a rat T helper marker (visualized by the mAb W3/25) was observed in response to PI-PLC treatment. This is in sharp contrast to the effects of trypsin, which removes most of the T helper marker but had little effect on RT-6.2. SDS-PAGE analysis of the RT-6.2 released by PI-PLC indicated that the Mr was not significantly changed by this treatment. Phase separation of the released RT-6.2 in Triton X-114 showed that the PI-PLC had converted it from an amphiphilic membrane form to a water-soluble form, apparently by removing its hydrophobic membrane anchoring domain. These results strongly suggest that RT-6.2, in common with Thy-1 and several other cell surface proteins, is anchored in the membrane by the 1,2-diacylglycerol moiety of a covalently attached phosphatidylinositol molecule.


Assuntos
Antígenos de Superfície/análise , Isoantígenos/análise , Fosfatidilinositóis/metabolismo , Linfócitos T/imunologia , Fosfolipases Tipo C/farmacologia , Animais , Ratos
12.
J Exp Med ; 180(3): 945-58, 1994 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-8064242

RESUMO

Macrophages from mice bearing large D1-DMBA-3 mammary tumors have a decreased capacity to kill tumor targets. This effect is due to an impaired ability to produce nitric oxide (NO) in response to lipopolysaccharide (LPS) stimulation. Here we report that the DA-3 tumor cell line, derived from the in vivo adenocarcinoma D1-DMBA-3, produces a factor that inhibits both NO production/release and cytotoxicity of LPS-activated peritoneal exudate macrophages (PEM). However, other complex macrophage functions such as phagocytosis, superoxide production, mitochondrial dehydrogenase activity, and synthesis of proteins were not reduced by this factor. The NO inhibitor has been found to be lipid in nature. Lipid extracts from DA-3 cell culture supernatants were purified by repeated silica gel column chromatography. The active molecule was unambiguously characterized as phosphatidyl serine (PS) by fast atom bombardment tandem mass spectrometry. Preliminary results indicate a lack of induced NO synthase (iNOS) activity in the lysates of LPS-activated PEM pretreated with PS. The ubiquity of PS in the inner leaflet of biological membranes and its NO inhibitory property, suggest that this phospholipid may be one of the long elusive molecules responsible for regulating physiological levels of NO in the host and hence preventing cellular dysfunction and/or tissue damage. Furthermore, the possible overexpression and shedding of PS by DA-3 tumor cells may represent a novel mechanism to impair macrophage cytotoxicity, a host function that contributes to the protection against developing neoplasms.


Assuntos
Neoplasias Mamárias Experimentais/química , Óxido Nítrico/antagonistas & inibidores , Fosfatidilserinas/isolamento & purificação , Animais , Feminino , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/biossíntese , Fosfatidilserinas/farmacologia , Fosfolipases Tipo C/farmacologia
13.
J Exp Med ; 170(2): 481-97, 1989 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-2526846

RESUMO

A low affinity receptor for IgG immune complexes, Fc gamma RIII(CD16), is expressed on human NK cells as an integral membrane glycoprotein anchored through a transmembrane peptide; on polymorphonuclear neutrophils (PMN) the receptor is anchored through a phosphatidylinositol (PI) linkage. The protein on NK cells has a molecular mass 6-10 kD larger than that on PMN, and, unlike the latter, is resistant to PI-specific phospholipase C (PI-PLC). Fc gamma RIII(CD16) transcripts isolated from PMN and NK cells of single donors revealed multiple single nucleotide differences, one of which converts an in frame UGA termination codon to a CGA codon. The resulting open reading frame encodes a longer cytoplasmic domain for Fc gamma RIII(CD16) in NK cells, contributing to its transmembrane anchor. Two nearly identical, linked genes that encode these transcripts have been cloned for Fc gamma RIII(CD16), one of which (III-1) is allelic for NA-1 and NA-2. The allelic sites have been mapped to two single nucleotides in the extracellular domain. These genes are transcribed in a cell type-specific fashion to generate the alternatively anchored forms of this receptor.


Assuntos
Antígenos de Diferenciação/genética , Células Matadoras Naturais/fisiologia , Neutrófilos/fisiologia , Receptores Fc/genética , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , DNA/genética , Genes , Glicosídeo Hidrolases/farmacologia , Humanos , Glicoproteínas de Membrana/genética , Dados de Sequência Molecular , Sondas de Oligonucleotídeos , Terminação Traducional da Cadeia Peptídica , Fosfatidilinositóis/fisiologia , RNA Mensageiro/genética , Receptores de IgG , Fosfolipases Tipo C/farmacologia
14.
J Exp Med ; 175(2): 437-45, 1992 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-1346270

RESUMO

Optimal induction of clonal expansion by normal CD4 T cells requires a ligand that can engage the T cell receptor as well as functionally defined costimulatory activity on the same antigen-presenting cell surface. While the presence of effective costimulation induces proliferation, T cell receptor ligation in its absence renders T cells inactive or anergic. The molecular basis of this costimulatory activity remains to be defined. Here we describe a monoclonal antibody that can block the costimulatory activity of splenic accessory cells. Treatment with this antibody not only blocks the proliferation of CD4 T cells to a T cell receptor ligand, but also induces T cell nonresponsiveness to subsequent stimulation. Sequence analysis of the antigen recognized by this antibody indicates that it recognizes a protein that is identical to heat-stable antigen. Gene transfer experiments directly demonstrate that this protein has costimulatory activity. Thus, heat-stable antigen meets the criteria for a costimulator of T cell clonal expansion.


Assuntos
Antígenos CD , Antígenos de Diferenciação/imunologia , Linfócitos T CD4-Positivos/imunologia , Glicoproteínas de Membrana , Animais , Anticorpos Monoclonais/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Linfócitos B/imunologia , Antígeno CD24 , Complexo CD3 , Células CHO , Cricetinae , Glicolipídeos/metabolismo , Glicosilfosfatidilinositóis , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Fosfatidilinositóis/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores Fc/imunologia , Transfecção , Fosfolipases Tipo C/farmacologia
15.
J Neurochem ; 111(1): 160-70, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19656262

RESUMO

The synaptic vesicle accumulation and subsequent morphological remodeling of axon terminals are characteristic features of presynaptic differentiation of zebrafish olfactory sensory neurons. The synaptic vesicle accumulation and axon terminal remodeling are regulated by protein kinase A and calcineurin signaling, respectively. To investigate upstream signals of presynaptic differentiation, we focused on Ca(2+) signaling as Ca(2+)/calmodulin is required for the activation of both calcineurin and some adenylyl cyclases. We here showed that application of Ca(2+)/calmodulin inhibitor or olfactory sensory neuron-specific expression of calmodulin inhibitory peptide suppressed both synaptic vesicle accumulation and axon terminal remodeling. Thus, the trigger of presynaptic differentiation could be Ca(2+) release from intracellular stores or Ca(2+) influx. Application of a phospholipase C inhibitor or olfactory sensory neuron-specific expression of inositol 1,4,5-trisphosphate (IP(3)) 5-phosphatase suppressed synaptic vesicle accumulation, but not morphological remodeling. In contrast, application of a voltage-gated Ca(2+) channel blocker or expression of Kir2.1 inward rectifying potassium channel prevented the morphological remodeling. We also provided evidence that IP(3) signaling acted upstream of protein kinase A signaling. Our results suggest that IP(3)-mediated Ca(2+)/calmodulin signaling stimulates synaptic vesicle accumulation and subsequent neuronal activity-dependent Ca(2+)/calmodulin signaling induces the morphological remodeling of axon terminals.


Assuntos
Sinalização do Cálcio/fisiologia , Condutos Olfatórios/citologia , Terminações Pré-Sinápticas/metabolismo , Sinapses/fisiologia , Vesículas Sinápticas/fisiologia , Análise de Variância , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Calmodulina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Embrião não Mamífero , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Inositol Polifosfato 5-Fosfatases , Microinjeções , Proteína de Marcador Olfatório/genética , Proteína de Marcador Olfatório/metabolismo , Condutos Olfatórios/embriologia , Neurônios Receptores Olfatórios , Monoéster Fosfórico Hidrolases/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , Transfecção/métodos , Fosfolipases Tipo C/farmacologia , Proteína 2 Associada à Membrana da Vesícula/genética , Proteína 2 Associada à Membrana da Vesícula/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
16.
J Cell Biol ; 105(3): 1377-86, 1987 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3654757

RESUMO

Qualitative and quantitative changes in neural cell adhesion molecule (N-CAM) protein and mRNA forms were measured during myogenesis in G8-1 and C2 cell lines. Indirect immunofluorescence assay showed that N-CAM was constitutively expressed by myoblasts in culture and that myotubes appeared to be stained more strongly. These changes were quantified using a dot blot assay. N-CAM levels increased almost 4-fold in G8-1 cells and 15-fold in C2 cells during myogenesis. The kinetics of accumulation of N-CAM were not coordinate with other muscle markers such as creatine kinase or acetylcholine receptor levels, since N-CAM accumulated significantly ahead of these markers. Immunoblotting showed that myogenesis was not associated with changes in the extent of sialylation of N-CAM. However, distinct changes in desialo forms were observed after neuraminidase treatment. Myogenesis was accompanied by increases in 125- and 155-kD desialo forms with minor changes in 120- and 145-kD forms. Biosynthetic labeling studies showed that myoblasts specifically expressed a transmembrane isoform of 145 kD that was phosphorylated and was down-regulated in myotubes. Pulse-chase analysis of myotubes showed that the 120-kD isoform and an isoform of 145 kD that co-migrated with, but was distinct from, the 145 kD transmembrane isoform of myoblasts were precursors of the 125- and 155-kD isoforms, respectively, that accumulated in myotubes. The 125- and 155-kD isoforms in myotubes are linked to the cell membrane via phosphatidylinositol linkage and can be released by phospholipase C. Indirect immunofluorescence analysis showed that phosphatidylinositol specific phospholipase C specifically released N-CAM from the myotube membrane generating N-CAM-free myotubes, while myoblasts were unaffected by this treatment. Three N-CAM mRNA species were observed in mouse muscle cell lines. Myoblasts were characterized by their expression of 6.7- and 5.2-kb transcripts while myotubes express 5.2- and 2.9-kb transcripts. Thus, myogenesis is qualitatively associated with a down regulation of the 6.7-kb transcript and an up regulation of the 5.2- and 2.9-kb transcript.


Assuntos
Antígenos de Superfície/genética , RNA Mensageiro/genética , Antígenos de Superfície/isolamento & purificação , Adesão Celular , Moléculas de Adesão Celular , Linhagem Celular , Cinética , Músculos/citologia , Processamento de Proteína Pós-Traducional , Tunicamicina/farmacologia , Fosfolipases Tipo C/farmacologia
17.
J Cell Biol ; 109(4 Pt 1): 1779-86, 1989 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2793939

RESUMO

During myogenesis myoblasts fuse to form multinucleate cells that express muscle-specific proteins. A specific cell-cell adhesion process precedes lipid bilayer union during myoblast fusion (Knudsen, K. A., and A. F. Horwitz. 1977. Dev. Biol. 58:328-338) and is mediated by cell surface glycoproteins (Knudsen, K. A., 1985. J. Cell Biol. 101:891-897). In this paper we show that myoblast adhesion and myotube formation are inhibited by treating fusion-competent myoblasts with phosphatidylinositol-specific phospholipase C (PI-PLC). The effect of PI-PLC on myoblast adhesion is dose dependent and inhibited by D-myo-inositol 1-monophosphate and the effect on myotube formation is reversible, suggesting a specific, nontoxic effect on myogenesis by the enzyme. A soluble form of adhesion-related glycoproteins is released from fusion-competent myoblasts by treatment with PI-PLC as evidenced by (a) the ability of phospholipase C (PLC)-released material to block the adhesion-perturbing activity of a polyclonal antiserum to intact myoblasts; and (b) the ability of PLC-released glycoprotein to stimulate adhesion-perturbing antisera when injected into mice. PI-PLC treatment of fusion-competent myoblasts releases an isoform of N-CAM into the supernate, suggesting that N-CAM may participate in mediating myoblast interaction during myogenesis.


Assuntos
Glicoproteínas de Membrana/fisiologia , Lipídeos de Membrana/fisiologia , Músculos/fisiologia , Fosfatidilinositóis/fisiologia , Animais , Adesão Celular/efeitos dos fármacos , Agregação Celular/efeitos dos fármacos , Fusão Celular , Embrião de Galinha , Eletroforese em Gel de Poliacrilamida , Soros Imunes , Immunoblotting , Glicoproteínas de Membrana/isolamento & purificação , Músculos/citologia , Fosfolipases Tipo C/farmacologia
18.
J Cell Biol ; 123(6 Pt 2): 1789-96, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8276897

RESUMO

alpha-Thrombin induced a change in the cell morphology of IIC9 fibroblasts from a semiround to an elongated form, accompanied by an increase in stress fibers. Incubation of the cells with phospholipase D (PLD) from Streptomyces chromofuscus and exogenous phosphatidic acid (PA) caused similar morphological changes, whereas platelet-derived growth factor (PDGF) and phorbol 12-myristate 13-acetate (PMA) induced different changes, e.g., disruption of stress fibers and cell rounding. alpha-Thrombin, PDGF, and exogenous PLD increased PA by 20-40%, and PMA produced a smaller increase. alpha-Thrombin and exogenous PLD produced rapid increases in the amount of filamentous actin (F-actin) that were sustained for at least 60 min. However, PDGF produced a transient increase of F-actin at 1 min and PMA caused no significant change. Dioctanoylglycerol was ineffective except at 50 micrograms/ml. Phospholipase C from Bacillus cereus, which increased diacylglycerol (DAG) but not PA, did not change F-actin content. Down-regulation of protein kinase C (PKC) did not block actin polymerization induced by alpha-thrombin. H-7 was also ineffective. Exogenous PA activated actin polymerization with a significant effect at 0.01 microgram/ml and a maximal increase at 1 microgram/ml. No other phospholipids tested, including polyphosphoinositides, significantly activated actin polymerization. PDGF partially inhibited PA-induced actin polymerization after an initial increase at 1 min. PMA completely or largely blocked actin polymerization induced by PA or PLD. These results show that PC-derived PA, but not DAG or PKC, activates actin polymerization in IIC9 fibroblasts, and indicate that PDGF and PMA have inhibitory effects on PA-induced actin polymerization.


Assuntos
Actinas/metabolismo , Ácidos Fosfatídicos/metabolismo , Ácidos Fosfatídicos/farmacologia , Fosfatidilcolinas/metabolismo , Actinas/ultraestrutura , Animais , Bacillus cereus/enzimologia , Linhagem Celular , Cricetinae , Cricetulus , Diglicerídeos/farmacologia , Embrião de Mamíferos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Cinética , Fosfolipase D/farmacologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteína Quinase C , Streptomyces/enzimologia , Acetato de Tetradecanoilforbol/farmacologia , Trombina/farmacologia , Fosfolipases Tipo C/metabolismo , Fosfolipases Tipo C/farmacologia
19.
J Cell Biol ; 123(2): 293-302, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8408213

RESUMO

Extracellular Ca2+ triggers assembly and sealing of tight junctions (TJs) in MDCK cells. These events are modulated by G-proteins, phospholipase C, protein kinase C (PKC), and calmodulin. In the present work we observed that 1,2-dioctanoylglycerol (diC8) promotes the assembly of TJ in low extracellular Ca2+, as evidenced by translocation of the TJ-associated protein ZO-1 to the plasma membrane, formation of junctional fibrils observed in freeze-fracture replicas, decreased permeability of the intercellular space to [3H]mannitol, and reorganization of actin filaments to the cell periphery, visualized by fluorescence microscopy using rhodamine-phalloidin. In contrast, diC8 in low Ca2+ did not induce redistribution of the Ca-dependent adhesion protein E-cadherin (uvomorulin). Extracellular antibodies to E-cadherin block junction formation normally induced by adding Ca2+. diC8 counteracted this inhibition, suggesting that PKC may be in the signaling pathway activated by E-cadherin-mediated cell-cell adhesion. In addition, we found a novel phosphoprotein of 130 kD which coimmunoprecipitated with the ZO-1/ZO-2 complex. Although the assembly and sealing of TJs may involve the activation of PKC, the level of phosphorylation of ZO-1, ZO-2, and the 130-kD protein did not change after adding Ca2+ or a PKC agonist. The complex of these three proteins was present even in low extracellular Ca2+, suggesting that the addition of Ca2+ or diC8 triggers the translocation and assembly of preformed TJ subcomplexes.


Assuntos
Diglicerídeos/fisiologia , Junções Intercelulares/metabolismo , Actinas/análise , Actinas/metabolismo , Animais , Cálcio/farmacologia , Calmodulina/farmacologia , Linhagem Celular , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Permeabilidade da Membrana Celular/fisiologia , Citoesqueleto/química , Citoesqueleto/metabolismo , Citoesqueleto/ultraestrutura , Diglicerídeos/farmacologia , Cães , Técnica de Fratura por Congelamento , Proteínas de Ligação ao GTP/fisiologia , Junções Intercelulares/ultraestrutura , Rim/citologia , Rim/metabolismo , Rim/ultraestrutura , Manitol/farmacocinética , Proteínas de Membrana/análise , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Microscopia de Fluorescência , Fosfoproteínas/análise , Fosfoproteínas/metabolismo , Fosforilação , Testes de Precipitina , Proteína Quinase C/fisiologia , Fosfolipases Tipo C/farmacologia , Proteína da Zônula de Oclusão-1
20.
J Cell Biol ; 119(2): 451-61, 1992 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-1400585

RESUMO

Cadherins are a family of cell adhesion molecules that exhibit calcium-dependent, homophilic binding. Their function depends on both an HisAlaVal sequence in the first extracellular domain, EC1, and the interaction of a conserved cytoplasmic region with intracellular proteins. T-cadherin is an unusual member of the cadherin family that lacks the HisAlaVal motif and is anchored to the membrane through a glycosyl phosphatidylinositol moiety (Ranscht, B., and M. T. Dours-Zimmermann. 1991. Neuron. 7:391-402). To assay the function of T-cadherin in cell adhesion, we have transfected T-cadherin cDNA into CHO cells. Two proteins, mature T-cadherin and the uncleaved T-cadherin precursor, were produced from T-cadherin cDNA. The T-cadherin proteins differed from classical cadherins in several aspects. First, the uncleaved T-cadherin precursor was expressed, together with mature T-cadherin, on the surface of the transfected cells. Second, in the absence of calcium, T-cadherin was more resistant to proteolytic cleavage than other cadherins. Lastly, in contrast to classical cadherins, T-cadherin was not concentrated into cell-cell contacts between transfected cells in monolayer cultures. In cellular aggregation assays, T-cadherin induced calcium-dependent, homophilic adhesion which was abolished by treatment of T-cadherin-transfected cells with phosphatidylinositol-specific phospholipase C. These results demonstrate that T-cadherin is a functional cadherin that differs in several properties from classical cadherins. The function of T-cadherin in homophilic cell recognition implies that the mechanism of T-cadherin-induced adhesion is distinct from that of classical cadherins.


Assuntos
Caderinas/metabolismo , Cálcio/farmacologia , Adesão Celular/fisiologia , Glicosilfosfatidilinositóis/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Caderinas/efeitos dos fármacos , Caderinas/genética , Adesão Celular/efeitos dos fármacos , Membrana Celular/química , Sequência Conservada , Cricetinae , Citocalasina D/farmacologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/fisiologia , Dados de Sequência Molecular , Nocodazol/farmacologia , Precursores de Proteínas/efeitos dos fármacos , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Transfecção , Tripsina/farmacologia , Fosfolipases Tipo C/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA