Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-30150481

RESUMO

Alpha toxin (AT) is a cytolytic pore-forming toxin that plays a key role in Staphylococcus aureus pathogenesis; consequently, extensive research was undertaken to understand the AT mechanism of action and its utility as a target for novel prophylaxis and treatment strategies against S. aureus infections. MEDI4893 (suvratoxumab) is a human anti-AT IgG1 monoclonal antibody (MAb) that targets AT and is currently in phase 2 clinical development. As shown previously, the MEDI4893-binding epitope on AT is comprised of the highly conserved amino acid regions 177 to 200 and 261 to 271, suggesting these amino acids are important for AT function. To test this hypothesis and gain insight into the effect of mutations in the epitope on AT neutralization by MEDI4893, nine MEDI4893 contact residues in AT were individually mutated to alanine. Consistent with our hypothesis, 8 out of 9 mutants exhibited >2-fold loss in lytic activity resulting from a defect in cell binding and pore formation. MEDI4893 binding affinity was reduced >2-fold (2- to 27-fold) for 7 out of 9 mutants, and no binding was detected for the W187A mutant. MEDI4893 effectively neutralized all of the lytic mutants in vitro and in vivo When the defective mutants were introduced into an S. aureus clinical isolate, the mutant-expressing strains exhibited less severe disease in mouse models and were effectively neutralized by MEDI4893. These results indicate the MEDI4893 epitope is highly conserved due in part to its role in AT pore formation and bacterial fitness, thereby decreasing the likelihood for the emergence of MAb-resistant variants.


Assuntos
Alanina/genética , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Toxinas Bacterianas/genética , Mutagênese/genética , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Células A549 , Animais , Antibacterianos/farmacologia , Anticorpos Monoclonais Humanizados , Anticorpos Amplamente Neutralizantes , Epitopos/genética , Epitopos/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia
2.
Artigo em Inglês | MEDLINE | ID: mdl-28584141

RESUMO

Secreted alpha-toxin and surface-localized clumping factor A (ClfA) are key virulence determinants in Staphylococcus aureus bloodstream infections. We previously demonstrated that prophylaxis with a multimechanistic monoclonal antibody (MAb) combination against alpha-toxin (MEDI4893*) and ClfA (11H10) provided greater strain coverage and improved efficacy in an S. aureus lethal bacteremia model. Subsequently, 11H10 was found to exhibit reduced affinity and impaired inhibition of fibrinogen binding to ClfA002 expressed by members of a predominant hospital-associated methicillin-resistant S. aureus (MRSA) clone, ST5. Consequently, we identified another anti-ClfA MAb (SAR114) from human tonsillar B cells with >100-fold increased affinity for three prominent ClfA variants, including ClfA002, and potent inhibition of bacterial agglutination by 112 diverse clinical isolates. We next constructed bispecific Abs (BiSAbs) comprised of 11H10 or SAR114 as IgG scaffolds and grafted anti-alpha-toxin (MEDI4893*) single-chain variable fragment to the amino or carboxy terminus of the anti-ClfA heavy chains. Although the BiSAbs exhibited in vitro potencies similar to those of the parental MAbs, only 11H10-BiSAb, but not SAR114-BiSAb, showed protective activity in murine infection models comparable to the respective MAb combination. In vivo activity with SAR114-BiSAb was observed in infection models with S. aureus lacking ClfA. Our data suggest that high-affinity binding to ClfA sequesters the SAR114-BiSAb to the bacterial surface, thereby reducing both alpha-toxin neutralization and protection in vivo These results indicate that a MAb combination targeting ClfA and alpha-toxin is more promising for future development than the corresponding BiSAb.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico , Bacteriemia/tratamento farmacológico , Toxinas Bacterianas/imunologia , Coagulase/imunologia , Proteínas Hemolisinas/imunologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Infecções Estafilocócicas/tratamento farmacológico , Animais , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/uso terapêutico , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais Humanizados , Anticorpos Neutralizantes/imunologia , Bacteriemia/microbiologia , Anticorpos Amplamente Neutralizantes , Feminino , Staphylococcus aureus Resistente à Meticilina/imunologia , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/imunologia , Fatores de Virulência
3.
Br J Dermatol ; 177(1): 63-71, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27779765

RESUMO

Atopic dermatitis (AD) is a complex, chronic inflammatory skin disorder affecting more than 10% of U.K. children and is a major cause of occupation-related disability. A subset of patients, particularly those with severe AD, are persistently colonized with Staphylococcus aureus and exacerbation of disease is commonly associated with this bacterium by virtue of increased inflammation and allergic sensitization, aggravated by skin barrier defects. Understanding the complex biology of S. aureus is an important factor when developing new drugs to combat infection. Staphylococcus aureus generates exoproteins that enable invasion and dissemination within the host skin but can also damage the skin and activate the host immune system. Antibiotics are often used by dermatologists to aid clearance of S. aureus; however, these are becoming less effective and chronic usage is discouraged with the emergence of multiple antibiotic-resistant strains. New ways to target S. aureus using monoclonal antibodies and vaccines are now being developed. This review will attempt to evaluate the key biology of S. aureus, current treatment of S. aureus infections in AD and recent advances in developing new anti-S. aureus therapies that have potential in severe AD.


Assuntos
Dermatite Atópica/tratamento farmacológico , Infecções Cutâneas Estafilocócicas/tratamento farmacológico , Staphylococcus aureus , Antibacterianos/uso terapêutico , Citocinas/metabolismo , Dermatite Atópica/microbiologia , Previsões , Humanos , Queratinócitos/microbiologia , Queratinócitos/fisiologia
4.
Antimicrob Agents Chemother ; 59(8): 4526-32, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25987629

RESUMO

Immunocompromised individuals are at increased risk of Staphylococcus aureus pneumonia. Neutralization of alpha-toxin (AT) with the monoclonal antibody (MAb) MEDI4893* protects normal mice from S. aureus pneumonia; however, the effects of the MAb in immunocompromised mice have not been reported. In this study, passive immunization with MEDI4893* increased survival rates and reduced bacterial numbers in the lungs in an immunocompromised murine S. aureus pneumonia model. Lungs from infected mice exhibited alveolar epithelial damage, protein leakage, and bacterial overgrowth, whereas lungs from mice passively immunized with MEDI4893* retained a healthy architecture, with an intact epithelial barrier. Adjunctive therapy or prophylaxis with a subtherapeutic MEDI4893* dose combined with subtherapeutic doses of vancomycin or linezolid improved survival rates, compared with the monotherapies. Furthermore, coadministration of MEDI4893* with vancomycin or linezolid extended the antibiotic treatment window. These data suggest that MAb-mediated neutralization of AT holds promise in strategies for prevention and adjunctive therapy among immunocompromised patients.


Assuntos
Antibacterianos/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Hospedeiro Imunocomprometido/efeitos dos fármacos , Pneumonia Estafilocócica/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Animais , Anticorpos Monoclonais Humanizados , Anticorpos Amplamente Neutralizantes , Feminino , Linezolida/farmacologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Taxa de Sobrevida , Vancomicina/farmacologia
5.
Antimicrob Agents Chemother ; 58(2): 1108-17, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24295977

RESUMO

Alpha-toxin (AT) is a major virulence factor in the disease pathogenesis of Staphylococcus aureus. We previously identified a monoclonal antibody (MAb) against AT that reduced disease severity in a mouse dermonecrosis model. Here, we evaluate the activity of an affinity-optimized variant, LC10, in a mouse model of S. aureus pneumonia. Passive immunization with LC10 increased survival and reduced bacterial numbers in the lungs and kidneys of infected mice and showed protection against diverse S. aureus clinical isolates. The lungs of S. aureus-infected mice exhibited bacterial pneumonia, including widespread inflammation, whereas the lungs of mice that received LC10 exhibited minimal inflammation and retained healthy architecture. Consistent with reduced immune cell infiltration, LC10-treated animals had significantly lower (P < 0.05) proinflammatory cytokine and chemokine levels in the bronchoalveolar lavage fluid than did those of the control animals. This reduction in inflammation and damage to the LC10-treated animals resulted in reduced vascular protein leakage and CO2 levels in the blood. LC10 was also assessed for its therapeutic activity in combination with vancomycin or linezolid. Treatment with a combination of LC10 and vancomycin or linezolid resulted in a significant increase (P < 0.05) in survival relative to the monotherapies and was deemed additive to synergistic by isobologram analysis. Consistent with improved survival, the lungs of animals treated with antibiotic plus LC10 exhibited less inflammatory tissue damage than those that received monotherapy. These data provide insight into the mechanisms of protection provided by AT inhibition and support AT as a promising target for immunoprophylaxis or adjunctive therapy against S. aureus pneumonia.


Assuntos
Antibacterianos/farmacologia , Anticorpos Monoclonais/farmacologia , Toxinas Bacterianas/antagonistas & inibidores , Proteínas Hemolisinas/antagonistas & inibidores , Pulmão/efeitos dos fármacos , Pneumonia Estafilocócica/tratamento farmacológico , Acetamidas/farmacologia , Animais , Toxinas Bacterianas/imunologia , Líquido da Lavagem Broncoalveolar/química , Quimiocinas/antagonistas & inibidores , Quimiocinas/biossíntese , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Proteínas Hemolisinas/imunologia , Imunização Passiva , Rim/efeitos dos fármacos , Rim/imunologia , Rim/microbiologia , Linezolida , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Oxazolidinonas/farmacologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/mortalidade , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Análise de Sobrevida , Vancomicina/farmacologia
6.
Immunooncol Technol ; 6: 9-17, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35757236

RESUMO

Immunotherapies have drastically improved clinical outcomes in a wide range of malignancies. Nevertheless, patient responses remain highly variable, and reliable biomarkers that predict responses accurately are not yet fully understood. Compelling evidence from preclinical studies and observational data from clinical cohorts have shown that commensal microorganisms that reside in the human gastrointestinal tract, collectively termed the 'microbiome', can actively modify responses to chemotherapeutic agents and immunotherapies by influencing host immunosurveillance. Notably, microbial correlates are largely context specific, and response signatures may vary by patient population, geographic location and type of anticancer treatment. Therefore, the incongruence of beneficial microbiome signatures across studies, along with an emerging understanding of the mechanisms underlying the interactions between the microbiome, metabolome and host immune system, highlight a critical need for additional comprehensive and standardized multi-omics studies. Future research should consider key host factors, such as diet and use of medication, in both preclinical animal models and large-scale, multicenter clinical trials. In addition, there is a strong rationale to evaluate the microbiome as a tumor-extrinsic biomarker of clinical outcomes and to test the therapeutic potential of derived microbial products (e.g. defined microbial consortia), with the eventual goal of improving the efficacy of existing anticancer treatments. This review discusses the importance of the microbiome from the perspective of cancer immunotherapies, and outlines future steps that may contribute to wide-ranging clinical and translational benefits that may improve the health and quality of life of patients with cancer.

7.
Science ; 292(5517): 695-7, 2001 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-11326092

RESUMO

The protective antigen moiety of anthrax toxin translocates the toxin's enzymic moieties to the cytosol of mammalian cells by a mechanism that depends on its ability to heptamerize and insert into membranes. We identified dominant-negative mutants of protective antigen that co-assemble with the wild-type protein and block its ability to translocate the enzymic moieties across membranes. These mutants strongly inhibited toxin action in cell culture and in an animal intoxication model, suggesting that they could be useful in therapy of anthrax.


Assuntos
Antraz/tratamento farmacológico , Antígenos de Bactérias , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/genética , Mutação , Animais , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Células CHO , Membrana Celular/metabolismo , Cricetinae , Endocitose , Genes Dominantes , Masculino , Transporte Proteico , Ratos , Ratos Endogâmicos F344 , Receptores de Peptídeos/metabolismo
8.
Nat Biotechnol ; 19(10): 958-61, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11581662

RESUMO

Screening peptide libraries is a proven strategy for identifying inhibitors of protein-ligand interactions. Compounds identified in these screens often bind to their targets with low affinities. When the target protein is present at a high density on the surface of cells or other biological surfaces, it is sometimes possible to increase the biological activity of a weakly binding ligand by presenting multiple copies of it on the same molecule. We isolated a peptide from a phage display library that binds weakly to the heptameric cell-binding subunit of anthrax toxin and prevents the interaction between cell-binding and enzymatic moieties. A molecule consisting of multiple copies of this nonnatural peptide, covalently linked to a flexible backbone, prevented assembly of the toxin complex in vitro and blocked toxin action in an animal model. This result demonstrates that protein-protein interactions can be inhibited by a synthetic, polymeric, polyvalent inhibitor in vivo.


Assuntos
Antígenos de Bactérias , Toxinas Bacterianas/antagonistas & inibidores , Peptídeos/farmacologia , Resinas Acrílicas , Inibidores de Adenilil Ciclases , Adenilil Ciclases/metabolismo , Animais , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidade , Células CHO , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/metabolismo , Cricetinae , Desenho de Fármacos , Ensaio de Imunoadsorção Enzimática , Biblioteca de Peptídeos , Peptídeos/síntese química , Peptídeos/isolamento & purificação , Peptídeos/metabolismo , Ligação Proteica , Ratos , Ratos Endogâmicos F344
9.
mBio ; 7(3)2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27353753

RESUMO

UNLABELLED: Staphylococcus aureus produces numerous virulence factors, each contributing different mechanisms to bacterial pathogenesis in a spectrum of diseases. Alpha toxin (AT), a cytolytic pore-forming toxin, plays a key role in skin and soft tissue infections and pneumonia, and a human anti-AT monoclonal antibody (MAb), MEDI4893*, has been shown to reduce disease severity in dermonecrosis and pneumonia infection models. However, interstrain diversity and the complex pathogenesis of S. aureus bloodstream infections suggests that MEDI4893* alone may not provide adequate protection against S. aureus sepsis. Clumping factor A (ClfA), a fibrinogen binding protein, is an important virulence factor facilitating S. aureus bloodstream infections. Herein, we report on the identification of a high-affinity anti-ClfA MAb, 11H10, that inhibits ClfA binding to fibrinogen, prevents bacterial agglutination in human plasma, and promotes opsonophagocytic bacterial killing (OPK). 11H10 prophylaxis reduced disease severity in a mouse bacteremia model and was dependent on Fc effector function and OPK. Additionally, prophylaxis with 11H10 in combination with MEDI4893* provided enhanced strain coverage in this model and increased survival compared to that obtained with the individual MAbs. The MAb combination also reduced disease severity in murine dermonecrosis and pneumonia models, with activity similar to that of MEDI4893* alone. These results indicate that an MAb combination targeting multiple virulence factors provides benefit over a single MAb neutralizing one virulence mechanism by providing improved efficacy, broader strain coverage, and protection against multiple infection pathologies. IMPORTANCE: Alternative strategies to broad-spectrum antibiotics are required to combat the antibiotic resistance epidemic. Previous attempts at active or passive immunization against Staphylococcus aureus targeting single antigens have failed in clinical trials despite positive preclinical data. To provide broad disease and isolate coverage, an effective immunization strategy likely must target multiple virulence mechanisms of the pathogen. Herein, we tested a multimechanistic MAb combination targeting alpha toxin (AT) and clumping factor A (ClfA) that neutralizes AT-mediated cytotoxicity, blocks fibrinogen binding by ClfA, prevents bacterial agglutination, targets the bacteria for opsonophagocytic killing, and provides broad isolate coverage in a lethal-bacteremia model. Although each MAb alone was effective in bacteremia against some individual isolates, the MAb combination provided improved protection against other isolates. These results illustrate the importance of targeting multiple virulence mechanisms and highlight the potential for an MAb combination targeting AT and ClfA to effectively prevent S. aureus disease.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Toxinas Bacterianas/imunologia , Coagulase/imunologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/imunologia , Fatores de Virulência/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/uso terapêutico , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Neutralizantes/uso terapêutico , Carga Bacteriana , Modelos Animais de Doenças , Células HL-60 , Humanos , Imunização Passiva/métodos , Camundongos , Fagocitose , Infecções Estafilocócicas/sangue , Infecções Estafilocócicas/microbiologia
10.
Clin Vaccine Immunol ; 19(3): 377-85, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22237895

RESUMO

Staphylococcus aureus alpha toxin (AT) is an important virulence determinant and may be a valid target for immunoprophylaxis against staphylococcal disease. Here we report the identification of potent inhibitory anti-AT monoclonal antibodies (MAbs) derived using B-cell hybridoma technology from VelocImmune mice engineered to produce IgG with a human variable domain. A small panel of inhibitory MAbs blocked AT-mediated lysis of rabbit red blood cells, A549 human lung epithelial cells, and THP-1 human monocytic cells, in a dose-dependent manner. Binding studies indicated that these MAbs recognize a similar epitope on AT and exhibit dissociation constants (K(D)) ranging from 0.50 to 15 nM. In an S. aureus dermonecrosis model, mice passively immunized with anti-AT inhibitory MAbs exhibited significant reductions of lesion size relative to mice treated with an irrelevant IgG control. Interestingly, there was a correlation between MAb affinity for a single epitope, the 50% inhibitory concentration (IC(50)) in the AT hemolytic assay, and lesion size reduction in the dermonecrosis model. A representative high-affinity MAb, 2A3.1, was demonstrated to significantly reduce lesion size following infection with three different clinical isolates (USA300, CC30, and CC5). Taken together, these results indicate that in vitro potency of anti-AT MAbs predicts in vivo potency in this model, supporting their continued preclinical evaluation as molecules for immunoprophylaxis against staphylococcal skin and soft tissue infections caused by diverse clinical isolates.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antitoxinas/administração & dosagem , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/toxicidade , Proteínas Hemolisinas/antagonistas & inibidores , Proteínas Hemolisinas/toxicidade , Necrose/prevenção & controle , Infecções Cutâneas Estafilocócicas/prevenção & controle , Animais , Anticorpos Monoclonais/farmacologia , Antitoxinas/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Relação Dose-Resposta Imunológica , Células Epiteliais/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Feminino , Humanos , Imunização Passiva , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/efeitos dos fármacos , Necrose/patologia , Coelhos , Infecções Cutâneas Estafilocócicas/patologia
11.
Mol Microbiol ; 25(3): 429-40, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9302006

RESUMO

Clostridium septicum alpha toxin is activated by a proteolytic cleavage at Arg-398 in its carboxy terminus, which yields a 41.3-kDa cytolytically active toxin and a 5.1-kDa propeptide. Studies were performed to determine when the propeptide dissociated from the toxin after proteolytic activation of the protoxin (AT(pro)) and to demonstrate the chaperone activity of the propeptide. The propeptide was found to remain associated with the toxin after activation with trypsin (AT(act)) when analysed by gel filtration or affinity chromatography of a polyhistidine-tagged derivative that contained the polyhistidine tag on the propeptide. The affinity of the propeptide for the toxin was decreased significantly when a mutation was introduced in which Val-400 was converted to a cysteine residue. This mutation destabilized the interaction of the propeptide with the toxin and the propeptide was found to dissociate from the toxin under the same gel-filtration conditions used for the wild-type toxin. The separation of the propeptide in the V400C mutant did not affect the cytolytic activity of the toxin and therefore the propeptide was not necessary for cytolytic activity. These data suggested that the propeptide did not dissociate from the main body of the toxin after proteolysis. Further analysis demonstrated that purified propeptide was a potent inhibitor of alpha toxin activity, which inhibited the oligomerization of alpha toxin into a functional pore. These data suggest that the propeptide does not participate in the final oligomerized complex and that oligomerization appears to displace the propeptide from AT(act). The importance of the propeptide to the solution stability of alpha toxin was also demonstrated. When AT(pro) was activated in solution with trypsin a significant level (approximately 50%) of inactive aggregate formed. This aggregate, which could be removed by centrifugation at 14,000 x g, was made up of both SDS-sensitive and -resistant aggregates, suggesting that a variety of inactive aggregates formed when the monomers interacted in solution. Significantly higher levels of haemolytic activity (approximately 16-fold) were observed when alpha toxin was proteolytically activated after membrane binding instead of in solution. These results support the role of the propeptide as an intramolecular chaperone that stabilizes the monomeric AT(pro) and shuttles it to the membrane where it is activated by protease, oligomerizes into a pre-pore complex and forms a pore. The data suggest that oligomerization of the toxin displaces the propeptide from the monomer form of alpha toxin and that the propeptide does not participate in, and is not necessary to, the final cytolytic complex.


Assuntos
Toxinas Bacterianas/metabolismo , Clostridium/metabolismo , Chaperonas Moleculares/metabolismo , Toxinas Bacterianas/antagonistas & inibidores , Toxinas Bacterianas/genética , Sequência de Bases , Clonagem Molecular , Clostridium/genética , Primers do DNA/genética , Membrana Eritrocítica/efeitos dos fármacos , Escherichia coli/genética , Hemólise/efeitos dos fármacos , Humanos , Técnicas In Vitro , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
J Biol Chem ; 276(11): 8371-6, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11113126

RESUMO

The protective antigen (PA) moiety of anthrax toxin delivers the toxin's enzymatic moieties to the cytosol of mammalian cells by a mechanism associated with its ability to heptamerize and form a transmembrane pore. Here we report that mutations in Lys-397, Asp-425, or Phe-427 ablate killing of CHO-K1 cells by a cytotoxic PA ligand. These mutations blocked PA's ability to mediate pore formation and translocation in cells but had no effect on its receptor binding, proteolytic activation, or ability to oligomerize and bind the toxin's enzymatic moieties. The mutation-sensitive residues lie in the 2beta(7)-2beta(8) and 2beta(10)-2beta(11) loops of domain 2 and are distant both in primary structure and topography from the 2beta(2)-2beta(3) loop, which is believed to participate in formation of a transmembrane beta-barrel. These results suggest that Lys-397, Asp-425, and Phe-427 participate in conformational rearrangements of a heptameric pore precursor that are necessary for pore formation and translocation. Identification of these residues will aid in elucidating the mechanism of translocation and may be useful in developing therapeutic and prophylactic agents against anthrax.


Assuntos
Antígenos de Bactérias , Toxinas Bacterianas/química , Animais , Toxinas Bacterianas/toxicidade , Transporte Biológico , Células CHO , Cricetinae , Concentração de Íons de Hidrogênio , Mutação Puntual , Conformação Proteica , Relação Estrutura-Atividade
13.
Mol Microbiol ; 23(3): 551-8, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9044288

RESUMO

Low-temperature inhibition of the cytolytic activity of alpha toxin has facilitated the identification of an important step in the cytolytic mechanism of this toxin. When alpha toxin-dependent haemolysis was measured on erythrocytes at various temperatures it was clear that at temperatures < or = 15 degrees C the haemolysis rate was significantly inhibited with little or no haemolysis occurring at 4 degrees C. Alpha toxin appeared to bind to and oligomerize on erythrocyte membranes with similar kinetics at 4 degrees C and 37 degrees C. The slight differences in these two processes at 4 degrees C and 37 degrees C could not account for the loss of cytolytic activity at low temperature. At 4 degrees C alpha toxin neither stimulated potassium release from erythrocytes nor formed pores in planar membranes. In contrast, at temperatures > or = 25 degrees C both processes proceeded rapidly. Pores that were opened in osmotically stabilized erythrocytes could not be closed by low temperature. Therefore, low temperature appeared to prevent the oligomerized complex from forming a pore in the membrane. These data support the hypothesis that alpha toxin oligomerizes into a membrane-bound, pre-pore complex prior to formation of a pore in a lipid bilayer.


Assuntos
Toxinas Bacterianas/farmacologia , Membrana Eritrocítica/efeitos dos fármacos , Proteínas Hemolisinas/farmacologia , Proteínas Nucleares/metabolismo , Membrana Eritrocítica/química , Membrana Eritrocítica/metabolismo , Hemólise/efeitos dos fármacos , Hemólise/fisiologia , Humanos , Proteínas de Membrana/metabolismo
14.
Mol Microbiol ; 31(3): 785-94, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10048023

RESUMO

Aerolysin is a bilobal channel-forming toxin secreted by Aeromonas hydrophila. The alpha toxin produced by Clostridium septicum is homologous to the large lobe of aerolysin. However, it does not contain a region corresponding to the small lobe of the Aeromonas toxin, leading us to ask what the function of the small lobe is. We fused the small lobe of aerolysin to alpha toxin, producing a hybrid protein that should structurally resemble aerolysin. Unlike aerolysin, the hybrid was not secreted when expressed in Aeromonas salmonicida. The purified hybrid was activated by proteolytic processing in the same way as both parent proteins and, after activation, it formed oligomers that corresponded to the aerolysin heptamer. Like aerolysin, the hybrid was far more active than alpha toxin against human erythrocytes and mouse T lymphocytes. Both aerolysin and the hybrid bound to human glycophorin, and both were inhibited by preincubation with this erythrocyte glycoprotein, whereas alpha toxin was unaffected. We conclude that aerolysin contains two receptor binding sites, one for glycosyl-phosphatidylinositol-anchored proteins that is located in the large lobe and is also found in alpha toxin, and a second site, located in the small lobe, that binds a surface carbohydrate determinant.


Assuntos
Toxinas Bacterianas/química , Clostridium/química , Clostridium/fisiologia , Receptores de Superfície Celular , Ribonucleases , Fosfolipases Tipo C/química , Aeromonas/química , Animais , Toxinas Bacterianas/análise , Proteínas Sanguíneas/farmacologia , Encéfalo/metabolismo , Células CHO , Proteínas de Transporte/fisiologia , Moléculas de Adesão Celular Neuronais/farmacologia , Sobrevivência Celular , Contactinas , Cricetinae , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Proteínas Granulares de Eosinófilos , Eritrócitos/metabolismo , Receptores de Folato com Âncoras de GPI , Proteínas Hemolisinas , Humanos , Camundongos , Proteínas Citotóxicas Formadoras de Poros , Ratos , Proteínas Recombinantes de Fusão/isolamento & purificação , Antígenos Thy-1/farmacologia , Fatores de Tempo , Distribuição Tecidual , Fosfolipases Tipo C/análise , Fosfolipases Tipo C/fisiologia , Glicoproteínas Variantes de Superfície de Trypanosoma/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA