Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 23(8): 2521-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19276173

RESUMO

Vibrio cholerae cytolysin (VCC) forms SDS-stable heptameric beta-barrel transmembrane pores in mammalian cell membranes. In contrast to structurally related pore formers of gram-positive organisms, no oligomeric prepore stage of assembly has been detected to date. In the present study, disulfide bonds were engineered to tie the pore-forming amino acid sequence to adjacent domains. In their nonreduced form, mutants were able to bind to rabbit erythrocytes and to native erythrocyte membranes suspended in PBS solution and form SDS-labile oligomers. These remained nonfunctional and represented the long-sought VCC prepores. Disulfide bond reduction in these oligomers released the pore-forming sequence from its locked position, and subsequent membrane insertion led to formation of SDS-stable pores and hemolysis. Addition of increasing amounts of an inactive mutant to wild-type toxin resulted in the formation of mixed oligomers with progressively reduced SDS stability on membranes. Membrane insertion of active monomers in these hybrid oligomers was still observed, but the functional pore diameter was reduced. These findings indicate that formation of an oligomeric prepore precedes membrane insertion of the pore-forming amino acid sequence and demonstrate that pore formation by VCC follows the same archetypical pathway as beta-barrel cytolysins of gram-positive organisms such as staphylococcal alpha-toxin.


Assuntos
Citotoxinas/química , Proteínas Citotóxicas Formadoras de Poros/química , Vibrio cholerae/citologia , Animais , Cisteína/química , Citotoxinas/genética , Citotoxinas/toxicidade , Membrana Eritrocítica/efeitos dos fármacos , Bactérias Gram-Positivas/química , Bactérias Gram-Positivas/patogenicidade , Técnicas In Vitro , Modelos Biológicos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/toxicidade , Estrutura Quaternária de Proteína , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/toxicidade , Vibrio cholerae/genética , Vibrio cholerae/patogenicidade
2.
Biol Chem ; 389(9): 1201-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18713007

RESUMO

Abstract Escherichia coli hemolysin is a pore-forming protein belonging to the RTX toxin family. Cysteine scanning mutagenesis was performed to characterize the putative pore-forming domain of the molecule. A single cysteine residue was introduced at 48 positions within the sequence spanning residues 170-400 and labeled with the polarity-sensitive dye badan. Spectrofluorimetric analyses indicated that several amino acids in this domain are inserted into the lipid bilayer during pore formation. An amphipathic alpha-helix spanning residues 272-298 was identified that may line the aqueous pore. The importance of this sequence was highlighted by the introduction of two prolines at positions 284 and 287. Disruption of the helix structure did not affect binding properties, but totally abolished the hemolytic activity of the molecule.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Porinas/metabolismo , Motivos de Aminoácidos , Animais , Linhagem Celular Tumoral , Membrana Eritrocítica , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Estrutura Secundária de Proteína , Coelhos
3.
J Biol Chem ; 281(36): 26014-21, 2006 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-16829693

RESUMO

High susceptibility of rabbit erythrocytes toward the pore-forming action of staphylococcal alpha-toxin correlates with the presence of saturable, high affinity binding sites. All efforts to identify a protein or glycolipid receptor have failed, and the fact that liposomes composed solely of phosphatidylcholine are efficiently permeabilized adds to the enigma. A novel concept is advanced here to explain the puzzle. We propose that low affinity binding moieties can assume the role of high affinity binding sites due to their spatial arrangement in the membrane. Evidence is presented that phosphocholine head groups of sphingomyelin, clustered in sphingomyelin-cholesterol microdomains, serve this function for alpha-toxin. Clustering is required so that oligomerization, which is prerequisite for stable attachment of the toxin to the membrane, can efficiently occur. Outside these clusters, binding to phosphocholine is too transient for toxin monomers to find each other. The principle of membrane targeting in the absence of any genuine, high affinity receptor may also underlie the assembly of other lipid-inserted oligomers including cytotoxic peptides, protein toxins, and immune effector molecules.


Assuntos
Toxinas Bacterianas , Proteínas Hemolisinas , Microdomínios da Membrana , Fosforilcolina , Estrutura Quaternária de Proteína , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Sítios de Ligação , Linhagem Celular , Colesterol/metabolismo , Eritrócitos/química , Eritrócitos/citologia , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Humanos , Lipossomos/química , Lipossomos/metabolismo , Microdomínios da Membrana/química , Microdomínios da Membrana/metabolismo , Fosforilcolina/química , Fosforilcolina/metabolismo , Ligação Proteica , Coelhos , Esfingomielina Fosfodiesterase/metabolismo , Esfingomielinas/química , Esfingomielinas/metabolismo
4.
FASEB J ; 20(7): 973-5, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16597673

RESUMO

Escherichia coli alpha-hemolysin (HlyA), archetype of a bacterial pore-forming toxin, has been reported to deregulate physiological Ca2+ channels, thus inducing periodic low-frequency Ca2+ oscillations that trigger transcriptional processes in mammalian cells. The present study was undertaken to delineate the mechanisms underlying the Ca2+ oscillations. Patch-clamp experiments were combined with single cell measurements of intracellular Ca2+ and with flowcytometric analyses. Application of HlyA at subcytocidal concentrations provoked Ca2+ oscillations in human renal and endothelial cells. However, contrary to the previous report, the phenomenon could not be inhibited by the Ca2+ channel blocker nifedipine and Ca2+ oscillations showed no constant periodicity at all. Ca2+ oscillations were dependent on the pore-forming activity of HlyA: application of a nonhemolytic but bindable toxin had no effect. Washout experiments revealed that Ca2+ oscillations could not be maintained in the absence of toxin in the medium. Analogously, propidium iodide flux into cells occurred in the presence of HlyA, but cells rapidly became impermeable toward the dye after toxin washout, indicating resealing or removal of the membrane lesions. Finally, patch-clamp experiments revealed temporal congruence between pore formation and Ca2+ influx. We conclude that the nonperiodic Ca2+ oscillations induced by HlyA are not due to deregulation of physiological Ca2+ channels but derive from pulsed influxes of Ca2+ as a consequence of formation and rapid closure of HlyA pores in mammalian cell membranes.


Assuntos
Cálcio/metabolismo , Proteínas de Escherichia coli/farmacologia , Escherichia coli/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Proteínas Hemolisinas , Humanos , Rim/citologia , Rim/efeitos dos fármacos , Nifedipino/farmacologia
5.
Methods Mol Biol ; 315: 393-403, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16110172

RESUMO

This chapter provides protocols to measure the reversible permeabilization of mast cells by streptolysin O (SLO) and to follow SLO-induced activation of mast cells by monitoring degranulation, activation of mitogen-activated protein kinases, and production of tumor necrosis factor-alpha. A method that uses SLO to deliver molecules into the cytosol of living cells also is described. Furthermore, we outline a procedure to measure the activation of nuclear factor-kappaB by lipopolysaccharide and ionomycin using transfection of mast cells with reporter genes by electroporation. These protocols should be widely applicable in mast cell research.


Assuntos
Lipopolissacarídeos/imunologia , Mastócitos/imunologia , Estreptolisinas/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Proteínas de Bactérias/imunologia , Células da Medula Óssea/imunologia , Permeabilidade da Membrana Celular , Eletroporação/métodos , Ativação Enzimática , Ionomicina/metabolismo , Ionóforos/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
6.
Mol Microbiol ; 57(1): 124-31, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15948954

RESUMO

Vibrio cholerae cytolysin (VCC) is an oligomerizing pore-forming toxin that is related to cytolysins of many other Gram-negative organisms. VCC contains six cysteine residues, of which two were found to be present in free sulphydryl form. The positions of two intramolecular disulphide bonds were mapped, and one was shown to be essential for correct folding of protoxin. Mutations were created in which the two free cysteines were deleted, so that single cysteine substitution mutants could be generated for site-specific labelling. Employment of polarity-sensitive fluorophores identified amino acid side-chains that formed part of the pore-forming domain of VCC. The sequence commenced at residue 311, and was deduced to form a beta-barrel in the assembled oligomer with the subsequent odd-numbered residues facing the lipid bilayer and even-numbered residues facing the lumen. Pro328/Lys329 were tentatively identified as the position at which the sequence turns back into the membrane and where the antiparallel beta-strand commences. This was deduced from fluorimetric analyses combined with experiments in which the pore was reversibly occluded by derivatization of sulphydryl groups with a bulky moiety. Our data support computer-based predictions that the membrane-permeabilizing amino acid sequence of VCC is homologous to the beta-barrel-forming sequence of staphylococcal cytolysins and identify the beta-barrel as a membrane-perforating structure that is highly conserved in evolution.


Assuntos
Membrana Celular/metabolismo , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , 2-Naftilamina/análogos & derivados , 2-Naftilamina/química , Sequência de Aminoácidos , Animais , Sequência Conservada , Cisteína/química , Dissulfetos/química , Corantes Fluorescentes/química , Glicoproteínas de Membrana/genética , Modelos Moleculares , Dados de Sequência Molecular , Perforina , Mutação Puntual , Proteínas Citotóxicas Formadoras de Poros , Conformação Proteica , Estrutura Terciária de Proteína , Coelhos , Compostos de Sulfidrila/química , Vibrio cholerae
7.
Infect Immun ; 71(11): 6171-7, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14573633

RESUMO

Streptolysin O (SLO), a major virulence factor of pyogenic streptococci, binds to cholesterol in the membranes of eukaryotic cells and oligomerizes to form large transmembrane pores. While high toxin doses are rapidly cytocidal, low doses are tolerated because a limited number of lesions can be resealed. Here, we report that at sublethal doses, SLO activates primary murine bone marrow-derived mast cells to degranulate and to rapidly induce or enhance the production of several cytokine mRNAs, including tumor necrosis factor alpha (TNF-alpha). Mast cell-derived TNF-alpha plays an important protective role in murine models of acute inflammation, and the production of this cytokine was analyzed in more detail. Release of biologically active TNF-alpha peaked approximately 4 h after stimulation with SLO. Production of TNF-alpha was blunted upon depletion of protein kinase C by pretreatment of the cells with phorbol-12 myristate-13 acetate. Transient permeabilization of mast cells with SLO also led to the activation of the stress-activated protein kinases p38 mitogen-activated protein (MAP) kinase and c-jun N-terminal kinase (JNK), and inhibition of p38 MAP kinase markedly reduced production of TNF-alpha. In contrast, secretion of preformed granule constituents triggered by membrane permeabilization was not dependent on p38 MAP kinase or on protein kinase C. Thus, transcriptional activation of mast cells following transient permeabilization might contribute to host defense against infections via the beneficial effects of TNF-alpha. However, hyperstimulation of mast cells might also lead to overproduction of TNF-alpha, which would then promote the development of toxic streptococcal syndromes.


Assuntos
Mastócitos/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteína Quinase C/fisiologia , Estreptolisinas/toxicidade , Fator de Necrose Tumoral alfa/biossíntese , Animais , Proteínas de Bactérias , Relação Dose-Resposta a Droga , Mastócitos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/análise , Ativação Transcricional , Fator de Necrose Tumoral alfa/genética , Proteínas Quinases p38 Ativadas por Mitógeno
8.
FASEB J ; 16(2): 237-9, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11744625

RESUMO

Streptolysin O (SLO), archetype of a cholesterol-binding bacterial cytolysin, forms large pores in the plasma membrane of mammalian cells. We have recently reported that when a limited number of pores are generated in a cell, they can be sealed in a Ca++-dependent process. Here, we show that resealing is followed by the release of IL-6 and IL-8 from keratinocytes and from endothelial cells, both relevant targets for SLO attack. Production of cytokines by these cells was preceded by activation of transcription factor nuclear factor kappaB, which thus emerges as a common denominator of stress responses to various pore-forming agents, including alpha-toxin of Staphylococcus aureus and complement. Furthermore, we show that activation and cytokine release in response to an agent that forms a pore in the plasma membrane do not depend on paracrine effects, because supernatants of cells perforated by SLO did not activate bystander cells. The study provides definitive evidence that a transient transmembrane pore suffices to trigger productive transcriptional activation in a target cell.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Estreptolisinas/farmacologia , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias , Linhagem Celular , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , NF-kappa B/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...