Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 98(6): 3185-90, 2001 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-11248053

RESUMO

The pore-forming toxin streptolysin O (SLO) can be used to reversibly permeabilize adherent and nonadherent cells, allowing delivery of molecules with up to 100 kDa mass to the cytosol. Using FITC-labeled albumin, 10(5)-10(6) molecules were estimated to be entrapped per cell. Repair of toxin lesions depended on Ca(2+)-calmodulin and on intact microtubules, but was not sensitive to actin disruption or to inhibition of protein synthesis. Resealed cells were viable for days and retained the capacity to endocytose and to proliferate. The active domains of large clostridial toxins were introduced into three different cell lines. The domains were derived from Clostridium difficile B-toxin and Clostridium sordelli lethal toxin, which glycosylate small G-proteins, and from Clostridium botulinum C2 toxin, which ADP-ribosylates actin. After delivery with SLO, all three toxins disrupted the actin cytoskeleton to cause rounding up of the cells. Glucosylation assays demonstrated that G-proteins Rho and Ras were retained in the permeabilized cells and were modified by the respective toxins. Inactivation of these G-proteins resulted in reduced stimulus-dependent granule secretion, whereas ADP-ribosylation of actin by the C. botulinum C2-toxin resulted in enhanced secretion in cells. The presented method for introducing proteins into living cells should find multifaceted application in cell biology.


Assuntos
Permeabilidade da Membrana Celular/fisiologia , Proteínas/farmacocinética , Albuminas/metabolismo , Animais , Proteínas de Bactérias , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Transporte Biológico , Células COS , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Dextranos/metabolismo , Relação Dose-Resposta a Droga , Glicosilação , Humanos , Imunoglobulina G/metabolismo , Tamanho da Partícula , Ratos , Vesículas Secretórias , Estreptolisinas/farmacologia , Células Tumorais Cultivadas , Proteínas ras/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
2.
J Biol Chem ; 276(18): 14835-41, 2001 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-11279048

RESUMO

Staphylococcal alpha-toxin forms heptameric pores on eukaryotic cells. After binding to the cell membrane in its monomeric form, the toxin first assembles into a heptameric pre-pore. Subsequently, the pre-pore transforms into the final pore by membrane insertion of an amphipathic beta-barrel, which comprises the "central loop" domains of all heptamer subunits. The process of membrane insertion was analyzed here using a set of functionally altered toxin mutants. The results show that insertion may be initiated within an individual protomer when its NH2 terminus activates its central loop. The activated state is then shared with the central loops of the residual heptamer subunits, which results in cooperative membrane penetration. This cooperation of the central loops commences while these are still remote from the lipid bilayer. Nevertheless, it is subject to modulation by the target membrane, which therefore acts across a distance much like an allosteric effector. However, while allosteric transitions usually are reversible, membrane insertion of alpha-toxin is an irreversible event, and we show here that it can proceed to completion in a domino-like fashion when triggered by as little as a single foreign atom within the entire heptamer.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Regulação Alostérica , Animais , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Sequência de Bases , Membrana Celular/metabolismo , Primers do DNA , Fibroblastos/metabolismo , Proteínas Hemolisinas/química , Proteínas Hemolisinas/genética , Humanos , Mutagênese , Coelhos
3.
J Leukoc Biol ; 68(6): 865-72, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11129654

RESUMO

Cleavage of membrane-associated L-selectin regulates leukocyte rolling on vascular endothelium at sites of inflammation. We report that rapid and massive shedding of L-selectin occurs from granulocytes attacked by the pore-forming bacterial toxin streptolysin O (SLO). Shedding was not induced by an SLO mutant that retained binding capacity but lacked pore-forming activity. Cells permeabilized with SLO exhibited a 1.5-fold increase in the activity of neutral sphingomyelinase, which was accompanied by increased ceramide formation. L-selectin cleavage was inducible by treatment of cells with bacterial sphingomyelinase, and also through exogenous application of a cell-permeable ceramide analog. Our data identify a novel path to the shedding process and show that activation of neutral sphingomyelinase with the generation of ceramide is an important event underlying enhanced sheddase function in cells permeabilized by a pore-forming toxin.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Ceramidas/fisiologia , Granulócitos/efeitos dos fármacos , Isoenzimas/fisiologia , Selectina L/metabolismo , Esfingomielina Fosfodiesterase/fisiologia , Esfingosina/análogos & derivados , Estreptolisinas/farmacologia , Proteínas de Bactérias , Ceramidas/biossíntese , Dipeptídeos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Granulócitos/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , N-Formilmetionina Leucil-Fenilalanina/farmacologia , Fenantrolinas/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Esfingomielina Fosfodiesterase/classificação , Esfingosina/farmacologia , Estaurosporina/farmacologia
4.
J Immunol ; 164(10): 5120-4, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10799869

RESUMO

We report that potassium leakage from cells leads to activation of the Ca2+-independent phospholipase A2 (iPLA2), and the latter plays a pivotal role in regulating the cleavage of pro-IL-1 beta by the IL-converting enzyme caspase-1 in human monocytes. K+ efflux led to increases of cellular levels of glycerophosphocholine, an unambiguous indicator of phospholipase A2 activation. Both maturation of IL-1 beta and formation of glycerophosphocholine were blocked by bromoenol lactone, the specific iPLA2 inhibitor. Bromoenol lactone-dependent inhibition of IL-1 beta processing was not due to perturbation of the export machinery for pro-IL-1 beta and IL-1 beta or to caspase-1 suppression. Conspicuously, activation of Ca2+-dependent phospholipase A2 did not support but rather suppressed IL-1 beta processing. Thus, our findings reveal a specific role for iPLA2 activation in the sequence of events underlying IL-1 beta maturation.


Assuntos
Cálcio/fisiologia , Interleucina-1/metabolismo , Fosfolipases A/fisiologia , Potássio/fisiologia , Processamento de Proteína Pós-Traducional/imunologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/imunologia , Cálcio/metabolismo , Caspase 1/metabolismo , Inibidores de Caspase , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/imunologia , Humanos , Imunossupressores/farmacologia , Interleucina-1/antagonistas & inibidores , Líquido Intracelular/enzimologia , Líquido Intracelular/imunologia , Líquido Intracelular/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Monócitos/imunologia , Monócitos/metabolismo , Naftalenos/farmacologia , Fosfolipases A/antagonistas & inibidores , Fosfolipases A2 , Potássio/metabolismo , Pironas/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
5.
Mol Microbiol ; 36(2): 467-76, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10792732

RESUMO

Staphylococcal alpha-toxin forms heptameric pores that render membranes permeable for monovalent cations. The pore is formed by an amphipathic beta-barrel encompassing amino acid residues 118-140 of each subunit of the oligomer. Human fibroblasts are susceptible to alpha-toxin but are able to repair the membrane lesions. Thereby, toxin oligomers remain embedded in the plasma membrane and exposed to the extracellular medium. In this study, we sought to detect structural changes occurring in the pore-forming sequence during lesion repair. Single cysteine substitution mutants were labelled with the environmentally sensitive fluorochrome acrylodan and, after mixing with wild-type toxin, incorporated into hybrid heptamers on fibroblast membranes. Formation of the lipid-inserted beta-barrel was accompanied by characteristic fluorescence emission shifts. After lesion repair, the environment of the residues at the outer surface of the beta-barrel remained unchanged, indicating continued contact with lipids. However, the labelled residues oriented towards the channel lumen underwent a green to blue shift in fluorescence, indicating reduced exposure to water. Pore closure proceeded in the presence of calmodulin inhibitors and of microtubule disruptors; however, it was prevented by cytochalasin D and by inhibitors of lipid metabolism. Our findings reveal the existence of a novel mechanism of membrane repair that may consist in constriction of the inserted proteinaceous pore within the lipid bilayer.


Assuntos
Toxinas Bacterianas/toxicidade , Cálcio/metabolismo , Permeabilidade da Membrana Celular , Membrana Celular/efeitos dos fármacos , Proteínas Hemolisinas/toxicidade , Staphylococcus/metabolismo , Membrana Celular/fisiologia , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Humanos , Linfócitos/efeitos dos fármacos , Espectrometria de Fluorescência
6.
Eur J Clin Invest ; 29(10): 835-41, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10583425

RESUMO

BACKGROUND: Oxidatively modified low-density lipoproteins (LDLs) have been implicated in the pathogenesis of atherosclerosis and are found in human vascular lesions. There is increasing evidence that complement activation may also play a role in atherogenesis. Activated complement proteins have been demonstrated to be present in early atherosclerotic lesions, and lipids isolated from lesions have been shown to activate complement, hence their designation as lesion complement activator (LCA). The question now arose whether oxidized LDLs would also activate complement. MATERIAL AND METHODS: The complement-activating capacity of a lesion complement activator preparation and of minimally as well as heavily oxidized LDL was investigated by measuring SC5b-9 formation in normal human serum. In addition, C3 conversion was followed using two-dimensional immunoelectrophoresis. RESULTS: Minimally and heavily oxidized LDL generated small but significant amounts of SC5b-9 (7.9 microgram mL-1, SD 3.5, and 10.8 microgram mL-1, SD 1.2, respectively; n = 6) compared with native LDL (3.3 microgram mL-1, SD 1.4; P < 0.05), whereas LCA generated substantially larger amounts of the terminal complex (32.0 microgram mL-1, SD 3.2). Both oxidized LDL preparations caused only minor C3 conversion. CONCLUSIONS: These findings show that oxidation does not confer relevant complement-activating properties on LDL, suggesting that the lesion complement activator is not directly related to oxidized LDL. Oxidized LDL is probably of minor importance for complement activation in atherosclerotic lesions.


Assuntos
Arteriosclerose/metabolismo , Ativação do Complemento , Peroxidação de Lipídeos , Lipoproteínas LDL/metabolismo , Arteriosclerose/patologia , Colesterol/sangue , Complemento C3/metabolismo , Humanos , Lipoproteínas LDL/sangue , Malondialdeído/metabolismo , Oxirredução
7.
Biochemistry ; 38(46): 15204-11, 1999 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-10563803

RESUMO

Streptolysin O is a four-domain protein toxin that permeabilizes animal cell membranes. The toxin first binds as a monomer to membrane cholesterol and subsequently assembles into oligomeric transmembrane pores. Binding is mediated by a C-terminally located tryptophan-rich motif. In a previous study, conformational effects of membrane binding were characterized by introducing single mutant cysteine residues that were then thiol-specifically derivatized with the environmentally sensitive fluorophoracrylodan. Membrane binding of the labeled proteins was accompanied by spectral shifts of the probe fluorescence, suggesting that the toxin molecule had undergone a conformational change. Here we provide evidence that this change corresponds to an allosteric transition of the toxin monomer that is required for the subsequent oligomerization and pore formation. The conformational change is reversible with reversal of binding, and it is related to temperature in a fashion that closely parallels the temperature-dependency of oligomerization. Furthermore, we describe a point mutation (N402E) that, while compatible with membrane binding, abrogates the accompanying conformational change. At the same time, the N402E mutation also abolishes oligomerization. These findings corroborate the contention that the target membrane acts as an allosteric effector to activate the oligomerizing and pore-forming capability of streptolysin O.


Assuntos
Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Estreptolisinas/antagonistas & inibidores , Estreptolisinas/metabolismo , Alanina/genética , Regulação Alostérica , Animais , Asparagina/genética , Proteínas de Bactérias , Cisteína/genética , Membrana Eritrocítica/metabolismo , Ácido Glutâmico/genética , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Mutação Puntual , Ligação Proteica , Conformação Proteica , Coelhos , Ovinos , Espectrometria de Fluorescência , Estreptolisinas/química , Estreptolisinas/genética , Temperatura
8.
Infect Immun ; 67(2): 817-25, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9916096

RESUMO

Incubation of human epithelial cells with nanomolar concentrations of chromatographically purified Serratia marcescens hemolysin (ShlA) caused irreversible vacuolation and subsequent lysis of the cells. Vacuolation differed from vacuole formation by Helicobacter pylori VacA. Sublytic doses of ShlA led to a reversible depletion of intracellular ATP. Restoration to the initial ATP level was presumably due to the repair of the toxin damage and was inhibited by cycloheximide. Pores formed in epithelial cells and fibroblasts without disruption of the plasma membrane, and the pores appeared to be considerably smaller than those observed in artificial lipid membranes and in erythrocytes and did not allow the influx of propidium iodide or trypan blue. All cytotoxic effects induced by isolated recombinant ShlA were also obtained with exponentially growing S. marcescens cells. The previously suggested role of the hemolysin in the pathogenicity of S. marcescens is supported by these data.


Assuntos
Proteínas de Bactérias , Proteínas Hemolisinas/toxicidade , Serratia marcescens/metabolismo , Trifosfato de Adenosina/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Eucarióticas , Fibroblastos/fisiologia , Células HeLa , Proteínas Hemolisinas/metabolismo , Hemólise , Humanos , Oligossacarídeos , Osmose , Potássio/metabolismo , Células Tumorais Cultivadas
10.
Proc Natl Acad Sci U S A ; 94(21): 11607-11, 1997 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-9326657

RESUMO

Staphylococcal alpha-toxin is a 293-residue, single-chain polypeptide that spontaneously assembles into a heptameric pore in target cell membranes. To identify the pore-forming domain, substitution mutants have been produced in which single cysteine residues were introduced throughout the toxin molecule. By attaching the environmentally sensitive dye acrylodan to the sulfhydryl groups, the environment of individual amino acid side chains could be probed. In liposomes, a single 23-amino acid sequence (residues 118-140) was found to move from a polar to a nonpolar environment, indicating that this sequence forms the walls of the pore. However, periodicity in side chain environmental polarity could not be detected in the liposomal system. In the present study, the fluorimetric analyses were extended to physiological target cells. With susceptible cells such as rabbit erythrocytes and human lymphocytes, the 23 central amino acids 118-140 were again found to insert into the membrane; in contrast to the previous study with liposomes, the expected periodicity was now detected. Thus, every other residue in the sequence 126-140 entered a nonpolar environment in a striking display of an amphipathic transmembrane beta-barrel. In contrast, human granulocytes were found to bind alpha-toxin to a similar extent as lymphocytes, but the heptamers forming on these cells failed to insert their pore-forming domain into the membrane. As a consequence, nonfunctional heptamers assembled and the cells remained viable. The data resolve the molecular organization of a pore-forming toxin domain in living cells and reveal that resistant cells can prevent insertion of the functional domain into the bilayer.


Assuntos
Toxinas Bacterianas/farmacologia , Toxinas Bacterianas/toxicidade , Proteínas Hemolisinas/farmacologia , Proteínas Hemolisinas/toxicidade , Neutrófilos/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Trifosfato de Adenosina/sangue , Substituição de Aminoácidos , Animais , Toxinas Bacterianas/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Membrana Celular/ultraestrutura , Cisteína , Eritrócitos/efeitos dos fármacos , Eritrócitos/fisiologia , Citometria de Fluxo , Proteínas Hemolisinas/química , Humanos , Bicamadas Lipídicas , Mutagênese Sítio-Dirigida , Neutrófilos/fisiologia , Fagocitose , Potássio/sangue , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/toxicidade , Staphylococcus , Linfócitos T/fisiologia
11.
Infect Immun ; 65(4): 1293-8, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9119464

RESUMO

Many strains of Vibrio cholerae non-O1 and O1 El Tor that cause diarrhea do not harbor genes for a known secretogenic toxin. However, these strains usually elaborate a pore-forming toxin, hitherto characterized as a hemolysin and here designated V. cholerae cytolysin, whose action on intestinal cells has not yet been described. We report that V. cholerae cytolysin binds as a monomer to Intestine 407 cells and then assembles into detergent-stable oligomers that probably represent tetra- or pentamers. Oligomer formation is accompanied by generation of small transmembrane pores that allow rapid flux of K+ but not influx of Ca2+ or propidium iodide. Pore formation is followed by irreversible ATP depletion and cell death. Binding of fewer than 10(4) toxin molecules per cell in vitro is lethal. The possibility is raised that production of this toxin by bacteria that are in close contact with intestinal cells is rapidly cytocidal in vivo, and death of intestinal cells may be a cause of diarrhea.


Assuntos
Membrana Celular/efeitos dos fármacos , Citotoxinas/farmacologia , Intestinos/microbiologia , Vibrio cholerae , Morte Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Intestinos/patologia , Intestinos/ultraestrutura , Transporte de Íons/efeitos dos fármacos , Membrana Nuclear/efeitos dos fármacos
12.
Infect Immun ; 64(9): 3646-51, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8751912

RESUMO

Certain membrane-anchored proteins, including several cytokines and cytokine receptors, can be released into cell supernatants through the action of endogenous membrane-bound metalloproteinases. The shed molecules are then able to fulfill various biological functions; for example, soluble interleukin-6 receptor (sIL-6R) can bind to bystander cells, rendering these cells sensitive to the action of IL-6. Using IL-6R as a model substrate, we report that the metalloproteinase from Serratia marcescens mimics the action of the endogenous shedding proteinase. Treatment of human monocytes with the bacterial protease led to a rapid release of sIL-6R into the supernatant. This effect was inhibitable with TAPI [N-(D,L-[2-(hydroxyaminocarbonyl)methyl]-4-methylpentanoyl) L-3-(2' naphthyl)-alanyl-L-alanine, 2-aminoethyl amide], a specific inhibitor of the membrane-bound intrinsic metalloproteinase, but not with other conventional proteinase inhibitors. sIL-6R-liberating activity was also detected in culture supernatants of Staphylococcus aureus, Pseudomonas aeruginosa, and Listeria monocytogenes, organisms that are known to produce metalloproteinases. sIL-6R released through the action of S. marcescens metalloproteinase retained biological activity and rendered IL-6-unresponsive human hepatoma cells sensitive to stimulation with IL-6. This was shown by Northern (RNA) blot detection of haptoglobin mRNA and by quantitative measurements of de novo-synthesized haptoglobin in cell supernatants. Analysis of immunoprecipitated, radiolabeled sIL-6R revealed that the bacterial protease cleaved IL-6R at a site distinct from that utilized by the endogenous protease. These studies show that membrane-anchored proteins can be released in active form through cleavage at multiple sites, and they uncover a novel mechanism via which microbial proteases possibly provoke long-range biological effects in the host organism.


Assuntos
Antígenos CD/metabolismo , Infecções Bacterianas/enzimologia , Metaloendopeptidases/metabolismo , Monócitos/metabolismo , Receptores de Interleucina/metabolismo , Animais , Chlorocebus aethiops , Humanos , Listeria monocytogenes/imunologia , Proteínas de Membrana/metabolismo , Pseudomonas aeruginosa/imunologia , Receptores de Interleucina-6 , Proteínas Recombinantes , Serratia marcescens/enzimologia , Transdução de Sinais , Solubilidade , Staphylococcus aureus/enzimologia , Especificidade por Substrato
13.
Infect Immun ; 64(8): 2974-9, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8757823

RESUMO

The best-known activity of Staphylococcus aureus sphingomyelinase C, alias beta-toxin, is as a hemolysin that provokes hot-cold lysis of erythrocytes which contain substantial amounts of sphingomyelin in the plasma membrane. Sheep erythrocytes are most susceptible, and we found that one hemolytic unit, representing the toxin concentration that elicits 50% hemolysis of 2.5 X 10(8) erythrocytes per ml, corresponds to 0.05 enzyme units or to approximately 0.25 microg of sphingomyelinase per ml. The cytotoxic action of beta-toxin on nucleated cells has not been described in any detail before, and the present investigation was undertaken to fill this information gap. We now identify beta-toxin as a remarkably potent monocytocidal agent. At a concentration of 0.001 U/ml, corresponding to approximately 5 ng/ml, beta-toxin killed over 50% of human monocytes (10(6) cells per ml) within 60 min. By contrast, 1 to 5 microg of beta-toxin per ml had no cytocidal effects on human granulocytes, fibroblasts, lymphocytes, or erythrocytes. A selective monocytocidal action was also observed with sphingomyelinase C from Bacillus cereus and a Streptomyces sp., whereas phospholipase A2 and phospholipase D at 100 U/ml were without effect. Monocytes succumbing to the action of beta-toxin processed and released interleukin-1beta, soluble interleukin-6 receptor, and soluble CD14 into the supernatant. Thus, monocyte killing by beta-toxin is associated with cytokine-related events that are important for the initiation and progression of infectious disease. These findings uncover a potentially important role for sphingomyelinase as a determinant of microbial pathogenicity.


Assuntos
Toxinas Bacterianas/farmacologia , Citocinas/metabolismo , Citotoxinas/farmacologia , Exotoxinas/farmacologia , Monócitos/efeitos dos fármacos , Esfingomielina Fosfodiesterase/farmacologia , Antígenos CD/metabolismo , Morte Celular , Relação Dose-Resposta a Droga , Proteínas Hemolisinas , Humanos , Interleucina-1/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Monócitos/metabolismo , Receptores de Interleucina/metabolismo , Receptores de Interleucina-6 , Staphylococcus aureus/enzimologia , Fatores de Tempo
14.
Proc Natl Acad Sci U S A ; 93(15): 7882-7, 1996 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-8755571

RESUMO

Cleavage of membrane-associated proteins with the release of biologically active macromolecules is an emerging theme in biology. However, little is known about the nature and regulation of the involved proteases or about the physiological inducers of the shedding process. We here report that rapid and massive shedding of the interleukin 6 receptor (IL-6R) and the lipopolysaccharide receptor (CD14) occurs from primary and transfected cells attacked by two prototypes of pore-forming bacterial toxins, streptolysin O and Escherichia coli hemolysin. Shedding is not induced by an streptolysin O toxin mutant which retains cell binding capacity but lacks pore-forming activity. The toxin-dependent cleavage site of the IL-6R was mapped to a position close to, but distinct from, that observed after stimulation with phorbol myristate acetate. Soluble IL-6R that was shed from toxin-treated cells bound its ligand and induced an IL-6-specific signal in cells that primarily lacked the IL-6R. Transsignaling by soluble IL-6R and soluble CD14 is known to dramatically broaden the spectrum of host cells for IL-6 and lipopolysaccharide, and is thus an important mechanism underlying their systemic inflammatory effects. Our findings uncover a novel mechanism that can help to explain the long-range detrimental action of pore-forming toxins in the host organism.


Assuntos
Antígenos CD/efeitos dos fármacos , Proteínas Hemolisinas/farmacologia , Receptores de Lipopolissacarídeos/efeitos dos fármacos , Macrófagos/imunologia , Monócitos/imunologia , Receptores de Interleucina/efeitos dos fármacos , Estreptolisinas/farmacologia , Animais , Antígenos CD/biossíntese , Proteínas de Bactérias , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Escherichia coli , Haptoglobinas/biossíntese , Humanos , Cinética , Receptores de Lipopolissacarídeos/biossíntese , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Receptores de Interleucina/biossíntese , Receptores de Interleucina-6 , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/efeitos dos fármacos , Transdução de Sinais , Estaurosporina/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Transfecção , Células Tumorais Cultivadas
15.
Antimicrob Agents Chemother ; 40(5): 1116-20, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8723450

RESUMO

An important determinant of nephrotoxicity, which is the major complication of long-term amphotericin B treatment, is dysfunction of distal tubular epithelial cells. The underlying cause for this rather selective damage to the cells is unknown. In the present investigation, it was shown that kidney epithelial cells were initially damaged by amphotericin B at concentrations of 2.5 to 10 micrograms/ml, as demonstrable by a dramatic drop in cellular K+ levels. Cells could recover from the initial toxic action of the polyene if they were kept in medium of neutral pH, and cellular K+ levels returned to normal after 6 h. However, the recovery mechanisms failed at lower pHs of 5.6 to 6.0. At low pHs, cells became progressively depleted of ATP; they leaked lactate dehydrogenase and became irreversibly damaged after approximately 6 h. The possibility that the low pH characteristic of the distal tubulus lumen renders the renal epithelial cells particularly vulnerable to the toxic action of amphotericin B is raised. The concept is in line with an earlier report that alkalization ameliorates amphotericin B nephrotoxicity in rats.


Assuntos
Anfotericina B/toxicidade , Antifúngicos/toxicidade , Túbulos Renais Distais/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Epitélio/efeitos dos fármacos , Epitélio/patologia , Concentração de Íons de Hidrogênio , Túbulos Renais Distais/metabolismo , Túbulos Renais Distais/patologia , L-Lactato Desidrogenase/metabolismo , Macaca mulatta , Potássio/metabolismo
16.
Arch Microbiol ; 165(2): 73-9, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8593102

RESUMO

Staphylococcal alpha-toxin, streptolysin-O, and Escherichia coli hemolysin are well-studied prototypes of pore-forming bacterial cytotoxins. Each is produced as a water-soluble single-chain polypeptide that inserts into target membranes to form aqueous transmembrane pores. This review will compare properties of the three toxin prototypes, highlighting the similarities and also the differences in their structure, mode of binding, mechanism of pore formation, and the responses they elicit in target cells. Pore-forming toxins represent the most potent and versatile weapons with which invading microbes damage the host macroorganism.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Proteínas de Escherichia coli , Proteínas Hemolisinas/metabolismo , Estreptolisinas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidade , Membrana Celular/ultraestrutura , Proteínas Hemolisinas/química , Proteínas Hemolisinas/toxicidade , Humanos , Bicamadas Lipídicas , Dados de Sequência Molecular , Estreptolisinas/química , Estreptolisinas/toxicidade
18.
Arch Virol ; 141(10): 1999-2008, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8920831

RESUMO

After vaginal infections of mice with neuroinvasive strains of herpes simplex virus type 1 and 2 (HSV-1, HSV-2) virus replicates in the epithelium of the vagina, in the paravaginal ganglia, in the spinal cord and finally in the brain and in the adrenal glands. However, viral antigens could be demonstrated only in the medulla of the adrenal glands but not in the cortex, as assessed by immunohistochemistry (IHC). HSV could not be isolated from liver, spleen, uterus, and ovaries. This contrasts to the intraperitoneal (i.p) route of infection with replication in different visceral organs including the adrenal gland's cortex.


Assuntos
Medula Suprarrenal/virologia , Antígenos Virais/análise , Herpesvirus Humano 1/isolamento & purificação , Herpesvirus Humano 2/isolamento & purificação , Medula Suprarrenal/patologia , Animais , Córtex Cerebral/patologia , Córtex Cerebral/virologia , Chlorocebus aethiops , Feminino , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 2/imunologia , Herpesvirus Humano 2/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Medula Espinal/patologia , Medula Espinal/virologia , Vagina/patologia , Vagina/virologia , Células Vero , Replicação Viral
19.
J Exp Med ; 182(6): 1959-71, 1995 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-7500042

RESUMO

Combined treatment with trypsin, cholesterol esterase, and neuraminidase transforms LDL, but not HDL or VLDL, to particles with properties akin to those of lipid extracted from atherosclerotic lesions. Single or double enzyme modifications, or treatment with phospholipase C, or simple vortexing are ineffective. Triple enzyme treatment disrupts the ordered and uniform structure of LDL particles, and gives rise to the formation of inhomogeneous lipid droplets 10-200 nm in diameter with a pronounced net negative charge, but lacking significant amounts of oxidized lipid. Enzymatically modified LDL (E-LDL), but not oxidatively modified LDL (ox-LDL), is endowed with potent complement-activating capacity. As previously found for lipid isolated from atherosclerotic lesions, complement activation occurs to completion via the alternative pathway and is independent of antibody. E-LDL is rapidly taken up by human macrophages to an extent exceeding the uptake of acetylated LDL (ac-LDL) or oxidatively modified LDL. After 16 h, cholesteryl oleate ester formation induced by E-LDL (50 micrograms/ml cholesterol) was in the range of 6-10 nmol/mg protein compared with 3-6 nmol/mg induced by an equivalent amount of acetylated LDL. At this concentration, E-LDL was essentially devoid of direct cytotoxic effects. Competition experiments indicated that uptake of E-LDL was mediated in part by ox-LDL receptor(s). Thus, approximately 90% of 125I-ox-LDL degradation was inhibited by a 2-fold excess of unlabeled E-LDL. Uptake of 125I-LDL was not inhibited by E-LDL. We hypothesize that extracellular enzymatic modification may represent an important step linking subendothelial deposition of LDL to the initiation of atherosclerosis.


Assuntos
Arteriosclerose/etiologia , Ativação do Complemento , Lipoproteínas LDL/metabolismo , Proteínas de Membrana , Receptores de Lipoproteínas , Complemento C3/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento , Proteínas do Sistema Complemento/metabolismo , Células Espumosas/metabolismo , Glicoproteínas/metabolismo , Humanos , Metabolismo dos Lipídeos , Lipoproteínas LDL/química , Macrófagos/metabolismo , Neuraminidase/metabolismo , Receptores Imunológicos/metabolismo , Receptores Depuradores , Receptores Depuradores Classe B , Esterol Esterase/metabolismo , Tripsina/metabolismo
20.
Med Microbiol Immunol ; 184(1): 37-44, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-8538577

RESUMO

V. cholerae El Tor cytolysin is a secreted, water-soluble protein of M(r) 60,000 that may be relevant to the pathogenesis of acute diarrhea. In this communication, we demonstrate that the toxin binds to and oligomerizes in target membranes to form SDS-stable aggregates of M(r) 200,000-250,000 that generate small transmembrane pores. Pores formed in erythrocytes were approximately 0.7 nm in size, as demonstrated by osmotic protection experiments. Binding was shown to occur in a temperature-independent manner preceding the temperature-dependent oligomerization step. Pores were also shown to be formed in L929 and HEp-2 cells, human fibroblasts and keratinocytes, albeit with highly varying efficacy. At neutral pH and in the presence of serum, human fibroblasts were able to repair a limited number of lesions. The collective data identify V. cholerae El Tor cytolysin as an oligomerizing toxin that damages cells by creating small transmembrane pores.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Citotoxinas/farmacologia , Membrana Eritrocítica/metabolismo , Vibrio cholerae/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Células Cultivadas , Citotoxinas/química , Citotoxinas/metabolismo , Membrana Eritrocítica/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Hemólise , Humanos , Canais Iônicos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Dados de Sequência Molecular , Peso Molecular , Polímeros , Coelhos , Dodecilsulfato de Sódio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...