Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Lancet Reg Health Eur ; 4: 100084, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33842908

RESUMO

BACKGROUND: Coronavirus disease 19 (COVID-19) is spreading globally and treatment options remain limited. A formulation of niclosamide, a potent anti-SARS-CoV-2 agent and a broad-spectrum antiviral treatment candidate, optimized for inhalation and intranasal administration (UNI91104) was developed. METHODS: We conducted a randomized, placebo-controlled, double-blind, single-centre, dose-ascending Phase 1 trial to assess the safety of UNI91104 in Denmark (NCT04576312). Healthy volunteers were randomly assigned to a ascending single dose in cohort 1-4 and five doses over 2.5 days in cohort 5. Inclusion criteria included a minimum 80% of predicted lung function. Exclusion criteria included severe, clinically significant allergies and current acute or chronic condition especially airway diseases. Safety was evaluated through adverse events (AEs) and pulmonary function tests including forced expiratory volume in one second (FEV1) and fractional exhaled nitric oxide (FeNO) tests. The primary endpoints were defined as the frequency of reported AEs and the change of safety variables relative to pre-dose. Data from all enroled healthy volunteers receiving any amount of IMP was included in the primary analyses. The pharmacokinetics of UNI91104 was determined. FINDINGS: The trial was conducted between 29 June 2020 and 08 August 2020. Thirty-four healthy volunteers received UNI91104 and ten placebo. No serious AEs or discontinuation were reported. Mild irritation in the upper respiratory tract following inhalation of UNI91104 was reported as most frequent AE (45 events in 26 healthy volunteers, 59% of all healthy volunteers). Nasal application was well-tolerated. There was no evidence of difference in the change of mean levels of pulmonary function tests between active and placebo group across all cohorts. Five healthy volunteers (11.4%) (1 on placebo) had signs of increased transient FeNO and 4 on active (9.1%) experienced asymptomatic drops in FEV1, which resolved spontaneously or were reversible with a ß2-agonist. Niclosamide exhibited dose-proportional pharmacokinetics following inhalation and intranasal administration. INTERPRETATION: UNI91104, a promising candidate for inhalation and intranasal therapy against COVID-19 and other viral respiratory tract infections is well-tolerated in healthy volunteers and warrants further testing in patient trials. FUNDING: The study was funded by Innovationsfonden Denmark and UNION therapeutics.

2.
Lancet Respir Med ; 6(7): 511-525, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29792288

RESUMO

BACKGROUND: Tralokinumab is an anti-interleukin-13 human monoclonal antibody developed for the treatment of severe, uncontrolled asthma. These clinical trials aimed to assess the efficacy and safety of tralokinumab in this population. METHODS: STRATOS 1 and STRATOS 2 were randomised, double-blind, parallel-group, placebo-controlled, phase 3 clinical trials that enrolled participants aged 12-75 years with severe asthma that was inadequately controlled despite use of inhaled corticosteroids (≥500 µg per day fluticasone or equivalent) and a long-acting ß2 agonist (but not oral corticosteroids). STRATOS 1 was done at 246 sites in 14 countries, and STRATOS 2 was done at 242 sites in 13 countries. In STRATOS 1, participants were randomly assigned (2:1) to receive tralokinumab 300 mg or matching placebo subcutaneously every 2 weeks or every 4 weeks for 52 weeks. In STRATOS 2, participants were randomly assigned (1:1) to receive tralokinumab 300 mg or matching placebo subcutaneously every 2 weeks for 52 weeks. STRATOS 1 attempted to identify a biomarker-positive population with enhanced tralokinumab benefit, which was then tested in STRATOS 2. The primary endpoint was the annualised asthma exacerbation rate (AAER) reduction at week 52 in the all-comers population for STRATOS 1 and in the biomarker-positive population for STRATOS 2. All efficacy analyses for both trials were done on the full analysis set by an intention-to-treat approach. The safety analysis set comprised any participant who received the investigational drug and was categorised by treatment received. These trials are registered with ClinicalTrials.gov, numbers NCT02161757 (STRATOS 1) and NCT02194699 (STRATOS 2), and with the EU Clinical Trials Register, EudraCT 2013-005614-35 (STRATOS 1) and EudraCT 2013-005615-27 (STRATOS 2). FINDINGS: STRATOS 1 was done between June 13, 2014, and Feb 28, 2017. 1207 participants were randomly assigned and 1202 treated as follows: tralokinumab every 2 weeks (n=398), tralokinumab every 4 weeks (n=404), or placebo (n=400). STRATOS 2 was done between Oct 30, 2014, and Sept 21, 2017. 856 participants were randomly assigned and 849 treated as follows: tralokinumab every 2 weeks (n=427) and placebo every 2 weeks (n=422). In the STRATOS 1 all-comers population, tralokinumab every 2 weeks did not significantly reduce AAER compared with placebo (7·0% reduction [95% CI -20·8 to 28·4]; rate ratio 0·93 [95% CI 0·72 to 1·21]; p=0·59). Baseline fractional exhaled nitric oxide (FENO) 37 ppb or greater was identified as the preferred biomarker in STRATOS 1; in FENO-high participants, tralokinumab every 2 weeks (n=97) reduced AAER by 44·0% (95% CI 6·0 to 66·0; rate ratio 0·56 [95% CI 0·34 to 0·94]; p=0·028) compared with placebo (n=102). In the STRATOS 2 FENO-high population, tralokinumab every 2 weeks (n=108) did not significantly improve AAER (15·8% reduction [95% CI -33·7 to 47·0]; rate ratio 0·84 [95% CI 0·53 to 1·34]; p=0·47) compared with placebo (n=121). The safety profile was consistent with that of previous tralokinumab trials. INTERPRETATION: Tralokinumab reduced AAER in participants with severe asthma with baseline FENO 37 ppb or higher in STRATOS 1, but not in STRATOS 2. These inconsistent effects on AAER do not support a key role for interleukin 13 in severe asthma exacerbations. FUNDING: AstraZeneca.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Asma/tratamento farmacológico , Adolescente , Adulto , Idoso , Criança , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Resultado do Tratamento , Adulto Jovem
3.
BMC Pulm Med ; 14: 52, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24669829

RESUMO

BACKGROUND: AZD9164 has demonstrated potential as an inhaled, long-acting, muscarinic antagonist (LAMA) bronchodilator. However, in patients with COPD, but not in healthy subjects, a transient initial drop in FEV1 was observed following inhalation of nebulised doses of AZD9164 in citrate buffer.Two additional studies were conducted to further assess the safety and tolerability of multiple ascending doses of AZD9164 in 27 white and 18 Japanese healthy subjects and in 4 patients with COPD. In these studies, AZD9164 was inhaled via Turbuhaler™. METHODS: These were Phase I, randomised, double-blind, placebo-controlled, multiple ascending dose (MAD) studies conducted in Sweden and UK. Healthy subjects (mean age 25.9 yrs) and patients with COPD (mean age 66 yrs, mean post-bronchodilator FEV1 60.1% predicted normal value) were randomised 2:1 to active treatment (400, 1000 or 2800 µg delivered doses of AZD9164) or placebo. RESULTS: No safety or tolerability concerns were identified in the healthy subjects at doses up to and including 2800 µg and both studies confirmed the bronchodilator effect of AZD9164. However, the first 3 patients in the COPD cohort who received AZD9164 (1000 µg) experienced a transient fall in FEV1 5 to 15 minutes after inhalation of AZD9164 while the patient receiving placebo did not. The study safety review process then resulted in cessation of further activities on AZD9164. Retrospective analysis showed that two healthy subjects had also had transient falls in FEV1 shortly after inhalation of AZD9164 400 and 2800 µg respectively, although neither reported any related respiratory symptoms or other AEs. CONCLUSIONS: These results show that transient paradoxical bronchoconstriction can occur in some healthy subjects, in addition to patients with COPD, following inhalation of AZD9164 and that the citrate buffer used in the nebulised formulation cannot have been the only cause of the drop in FEV1 in previous studies. As preclinical data do not provide an explanation, the reasons for this brief post-dose drop in FEV1 remain unclear. However, these results highlight the importance of monitoring lung function immediately post-dose when investigating novel inhaled treatments, even when a rapid onset of effect is not expected. TRIAL REGISTRATION: Clinicaltrials.gov NCT01016951 and NCT01096563.


Assuntos
Espasmo Brônquico/induzido quimicamente , Broncodilatadores/efeitos adversos , Antagonistas Muscarínicos/efeitos adversos , Piperidinas/efeitos adversos , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Quinuclidinas/efeitos adversos , Administração por Inalação , Adulto , Broncodilatadores/uso terapêutico , Método Duplo-Cego , Feminino , Humanos , Masculino , Antagonistas Muscarínicos/uso terapêutico , Piperidinas/uso terapêutico , Quinuclidinas/uso terapêutico , Estudos Retrospectivos , Adulto Jovem
4.
J Invest Dermatol ; 130(4): 1167-77, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19727116

RESUMO

We examined the importance of injury for the epidermal innate immune response in human skin wounds. We found that injury, independent of infiltrating inflammatory cells, generated prominent chemotactic activity toward neutrophils in injured skin because of IL-8 production. Furthermore, injury was a major inducer of the expression of antimicrobial (poly)peptides (AMPs) in skin wounds. In human skin, these injury-induced innate immune responses were mediated by activation of the epidermal growth factor receptor (EGFR). Consequently, inhibition of the EGFR blocked both the chemotactic activity generated in injured skin and the expression of the majority of the AMPs. The importance of injury was confirmed in mouse experiments in vivo, in which injury independent of infection was a potent inducer of AMPs in skin wounds. To our knowledge, these data thereby provide a previously unreported molecular link between injury and neutrophil accumulation and identify the molecular background for the vast expression of IL-8 and AMPs in wounded epidermis. Conceptually, these data show that the growth factor response elicited by injury is important for the recruitment of neutrophils in skin wounds.


Assuntos
Epiderme/imunologia , Epiderme/lesões , Queratinócitos/imunologia , Neutrófilos/imunologia , Cicatrização/imunologia , Monofosfato de Adenosina/metabolismo , Animais , Biópsia , Células Cultivadas , Quimiotaxia/imunologia , Epiderme/microbiologia , Receptores ErbB/metabolismo , Humanos , Interleucina-8/metabolismo , Interleucinas/metabolismo , Queratinócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Técnicas de Cultura de Órgãos , Especificidade da Espécie , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes
5.
Lancet ; 374(9691): 712-9, 2009 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-19716963

RESUMO

BACKGROUND: Concern is continuing about increased risk of pneumonia in patients with chronic obstructive pulmonary disease (COPD) who use inhaled corticosteroids. We aimed to establish the effects of inhaled budesonide on the risk of pneumonia in such patients. METHODS: We pooled patient data from seven large clinical trials of inhaled budesonide (320-1280 mug/day), with or without formoterol, versus control regimen (placebo or formoterol alone) in patients with stable COPD and at least 6 months of follow-up. The primary analysis compared treatment groups for the risk of pneumonia as an adverse event or serious adverse event during the trial or within 15 days of the trial end. Cox proportional hazards regression was used to analyse the data on an intention-to-treat basis. Data were adjusted for patients' age, sex, smoking status, body-mass index, and postbronchodilator percent of predicted forced expiratory volume in 1 s (FEV(1)). FINDINGS: We analysed data from 7042 patients, of whom 3801 were on inhaled budesonide and 3241 were on control treatment, with 5212 patient-years of exposure to treatment. We recorded no significant difference between treatment groups for the occurrence of pneumonia as an adverse event (3% [n=122 patients] vs 3% [n=103]; adjusted hazard ratio 1.05, 95% CI 0.81-1.37) or a serious adverse event (1% [n=53] vs 2% [n=50]; 0.92, 0.62-1.35), or for time to pneumonia as an adverse event (log-rank test 0.94) or a serious adverse event (0.61). Increasing age and decreasing percent of predicted FEV(1) were the only two variables that were significantly associated with occurrence of pneumonia as an adverse event or a serious adverse event. INTERPRETATION: Budesonide treatment for 12 months does not increase the risk of pneumonia in patients with COPD during that time and therefore is safe for clinical use in such patients. FUNDING: Michael Smith Foundation for Health Research.


Assuntos
Anti-Inflamatórios/efeitos adversos , Broncodilatadores/efeitos adversos , Budesonida/efeitos adversos , Pneumonia/induzido quimicamente , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Administração por Inalação , Etanolaminas/efeitos adversos , Feminino , Seguimentos , Volume Expiratório Forçado , Fumarato de Formoterol , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Pneumonia/epidemiologia , Modelos de Riscos Proporcionais , Ensaios Clínicos Controlados Aleatórios como Assunto , Medição de Risco , Fatores de Risco , Segurança , Fumar/epidemiologia , Fatores de Tempo
6.
FASEB J ; 22(9): 3146-53, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18511548

RESUMO

A common mammalian defense mechanism employed to prevent systemic dissemination of invasive bacteria involves occlusion of local microvasculature and encapsulation of bacteria within fibrin networks. Acquisition of plasmin activity at the bacterial cell surface circumvents this defense mechanism, allowing invasive disease initiation. To facilitate this process, S. pyogenes secretes streptokinase, a plasminogen-activating protein. Streptokinase polymorphism exhibited by S. pyogenes isolates is well characterized. However, the functional differences displayed by these variants and the biological significance of this variation has not been elucidated. Phylogenetic analysis of ska sequences from 28 S. pyogenes isolates revealed 2 main sequence clusters (clusters 1 and 2). All strains secreted streptokinase, as determined by Western blotting, and were capable of acquiring cell surface plasmin activity after incubation in human plasma. Whereas culture supernatants from strains containing cluster 1 ska alleles also displayed soluble plasminogen activation activity, supernatants from strains containing cluster 2 ska alleles did not. Furthermore, plasminogen activation activity in culture supernatants from strains containing cluster 2 ska alleles could only be detected when plasminogen was prebound with fibrinogen. This study indicates that variant streptokinase proteins secreted by S. pyogenes isolates display differing plasminogen activation characteristics and may therefore play distinct roles in disease pathogenesis.


Assuntos
Plasminogênio/metabolismo , Streptococcus pyogenes/enzimologia , Estreptoquinase/genética , Ativação Enzimática/fisiologia , Variação Genética , Humanos , Filogenia , Streptococcus pyogenes/genética
7.
Contrib Microbiol ; 14: 83-100, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17684334

RESUMO

Endotoxin, or lipopolysaccharide (LPS), is a constituent of the outer cell membrane of Gram-negative bacteria. LPS is a highly potent proinflammatory substance, that, when inhaled, dose-dependently causes fever, chills, and bronchoconstriction. These symptoms are accompanied by a proinflammatory response in sputum and bronchoalveolar lavage fluid with elevation of neutrophils, macrophages and certain cytokines/chemokines. This response can be partially modified with certain drugs. Similar inflammatory changes are observed both in the stable state of chronic obstructive lung disease (COPD) and during exacerbations of this disease. Cigarette smoke, which contains bioactive LPS, is the most common cause of COPD and may also precipitate exacerbations. In addition, the presence of Gram-negative bacteria in the lower airways is a distinguishing feature both of stable COPD and of exacerbations. Based on this knowledge we argue here that inhaled LPS provocation of healthy volunteers can be used as a model or COPD as well as for exacerbations of this disease.


Assuntos
Testes de Provocação Brônquica/métodos , Lipopolissacarídeos/imunologia , Doença Pulmonar Obstrutiva Crônica/etiologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/imunologia , Humanos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/metabolismo
8.
Mol Microbiol ; 65(5): 1147-57, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17662041

RESUMO

Disturbed haemostasis is a central finding in severe Streptococcus pyogenes infection. In particular, microthrombi are found both at the local site of infection and at distant sites. Platelets are responsible for maintaining vascular function and haemostasis. We report here that M1 protein of S. pyogenes triggers immune-mediated platelet activation and thrombus formation. M1 protein is released from the bacterial surface and forms complexes with plasma fibrinogen. These complexes bind to the fibrinogen receptor on resting platelets. When these complexes also contain immunoglobulin G (IgG) against M1 protein, this will engage the Fc receptor on the platelets and activation will occur. Activation of the platelets leads to platelet aggregation and the generation of platelet-rich thrombi. Neutrophils and monocytes are in turn activated by the platelets. Platelet thrombi are deposited in the microvasculature, and aggregated platelets, IgG and M1 protein colocalize in biopsies from patients diagnosed with S. pyogenes toxic shock syndrome. This chain of events results in a pro-coagulant and pro-inflammatory state typical of severe S. pyogenes infection.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/metabolismo , Ativação Plaquetária , Infecções Estreptocócicas/imunologia , Streptococcus pyogenes/imunologia , Trombose/sangue , Animais , Fibrinogênio/metabolismo , Humanos , Imunoglobulina G/imunologia , Monócitos/citologia , Monócitos/imunologia , Neutrófilos/citologia , Neutrófilos/imunologia , Infecções Estreptocócicas/fisiopatologia , Streptococcus pyogenes/patogenicidade
9.
J Clin Invest ; 116(7): 1878-85, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16778986

RESUMO

We found that sterile wounding of human skin induced epidermal expression of the antimicrobial (poly)peptides human beta-defensin-3, neutrophil gelatinase-associated lipocalin, and secretory leukocyte protease inhibitor through activation of the epidermal growth factor receptor. After skin wounding, the receptor was activated by heparin-binding epidermal growth factor that was released by a metalloprotease-dependent mechanism. Activation of the epidermal growth factor receptor generated antimicrobial concentrations of human beta-defensin-3 and increased the activity of organotypic epidermal cultures against Staphylococcus aureus. These data demonstrate that sterile wounding initiates an innate immune response that increases resistance to overt infection and microbial colonization.


Assuntos
Receptores ErbB/metabolismo , Imunidade Inata , Pele/imunologia , Pele/lesões , Ativação Transcricional , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Células Cultivadas , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Lipocalina-2 , Lipocalinas , Camundongos , Proteínas Secretadas Inibidoras de Proteinases , Proteínas/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Pele/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , beta-Defensinas/genética , beta-Defensinas/metabolismo
10.
Thromb Haemost ; 95(6): 982-90, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16732377

RESUMO

Streptococcus pyogenes is an important Gram-positive pathogen that is strictly limited to infections in humans. Here we report that streptolysin O (SLO), a cytolytic exotoxin secreted by S. pyogenes, activates human polymorphonuclear neutrophils (PMNs) by perforating these cells. This appears to be followed by an influx of Ca(2+) and p38 MAPK activation. As a consequence, PMNs secrete heparin-binding protein, a potent inducer of vascular leakage, and neutrophil-borne proteins, including LL-37, alpha-defensins, and elastase. The results of the present work therefore suggest that the interaction between SLO and PMNs evokes an exaggerated host response which may contribute to the pathogenesis of local and generalized S. pyogenes infections.


Assuntos
Degranulação Celular , Mediadores da Inflamação/metabolismo , Neutrófilos/fisiologia , Streptococcus pyogenes/enzimologia , Estreptolisinas/metabolismo , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas Sanguíneas/metabolismo , Cálcio/metabolismo , Proteínas de Transporte/metabolismo , Quelantes/farmacologia , Ácido Egtázico/farmacologia , Eritrócitos/metabolismo , Hemólise , Humanos , Imidazóis/farmacologia , Mutação , Neutrófilos/efeitos dos fármacos , Neutrófilos/microbiologia , Neutrófilos/ultraestrutura , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Transdução de Sinais , Streptococcus pyogenes/genética , Streptococcus pyogenes/isolamento & purificação , Estreptolisinas/genética , Transfecção , alfa-Defensinas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Catelicidinas
11.
Eur J Immunol ; 35(5): 1472-81, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15832298

RESUMO

During phagocytosis, surface receptors on neutrophils interact with pathogens opsonized with complement factor C3b/iC3b and in some cases with antibodies. In human immune sera antibodies directed against surface-bound M proteins mediated killing of Streptococcus pyogenes by neutrophils. Surprisingly, blocking of the Fc receptors had little effect on the killing. In contrast, inhibition of C3b/iC3b generation, or blocking of the major neutrophil iC3b receptor CD11b/CD18, enabled S. pyogenes to grow efficiently in immune sera. Inhibition of CD11b/CD18, but not of CD32, the major neutrophil signaling Fc receptor, prevented Streptococcus-induced NADPH oxidase-dependent respiratory burst, and blocking of C3b/iC3b formation inhibited Streptococcus-induced activation of Cdc42, a small GTPase critically involved in transmitting pro-inflammatory signals to the cytoskeleton. Consequently, ligation of CD11b/CD18 by bacteria-bound iC3b is necessary for inducing a neutrophil response leading to elimination of S. pyogenes in immune human serum.


Assuntos
Antígeno CD11b/imunologia , Neutrófilos/imunologia , Receptores de Complemento 3b/imunologia , Receptores Fc/imunologia , Streptococcus pyogenes/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Complemento C3b/imunologia , Citometria de Fluxo , Glicoproteínas/imunologia , Humanos , Fagocitose/imunologia , Proteínas Recombinantes/imunologia
12.
Science ; 305(5688): 1283-6, 2004 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-15333838

RESUMO

Group A streptococci, a common human pathogen, secrete streptokinase, which activates the host's blood clot-dissolving protein, plasminogen. Streptokinase is highly specific for human plasminogen, exhibiting little or no activity against other mammalian species, including mouse. Here, a transgene expressing human plasminogen markedly increased mortality in mice infected with streptococci, and this susceptibility was dependent on bacterial streptokinase expression. Thus, streptokinase is a key pathogenicity factor and the primary determinant of host species specificity for group A streptococcal infection. In addition, local fibrin clot formation may be implicated in host defense against microbial pathogens.


Assuntos
Plasminogênio/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/enzimologia , Streptococcus pyogenes/patogenicidade , Estreptoquinase/metabolismo , Ancrod/farmacologia , Animais , Anticoagulantes/farmacologia , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Contagem de Colônia Microbiana , Suscetibilidade a Doenças , Fibrina/metabolismo , Fibrinolisina/metabolismo , Fibrinólise , Deleção de Genes , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Plasminogênio/genética , Pele/irrigação sanguínea , Pele/microbiologia , Especificidade da Espécie , Baço/microbiologia , Streptococcus pyogenes/crescimento & desenvolvimento , Estreptoquinase/genética , Transgenes , Virulência
13.
Biochemistry ; 43(9): 2445-57, 2004 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-14992582

RESUMO

Protein L is a bacterial surface protein with 4-5 immunoglobulin (Ig)-binding domains (B1-B5), each of which appears to have two binding sites for Ig, corresponding to the two edges of its beta-sheet. To verify these sites biochemically and to probe their relative contributions to the protein L-Ig kappa light chain (kappa) interaction, we compared the binding of PLW (the Y47W mutant of the B1 domain) to that of mutants designed to disrupt binding to sites 1 and 2, using gel filtration, BIAcore surface plasmon resonance, fluorescence titration, and solid-phase radioimmunoassays. Gel filtration experiments show that PLW binds kappa both in 1:1 complexes and multivalently, consistent with two binding sites. Covalent dimers of the A20C and V51C mutants of PLW were prepared to eliminate site 1 and site 2 binding, respectively; both the A20C and V51C dimers bind kappa in 1:1 complexes and multivalently, indicating that neither site 1 nor site 2 is solely responsible for kappa binding. The A20R mutant was designed computationally to eliminate site 1 binding while preserving site 2 binding; consistent with this design, the A20R mutant binds kappa in 1:1 complexes but not multivalently. To probe the contributions of amino acid side chains to binding, we prepared 75 point mutants spanning nearly every residue of PLW; BIAcore studies of these mutants revealed two binding-energy "hot spots" consistent with sites 1 and 2. These data indicate that PLW binds kappa at both sites with similar affinities (high nanomolar), with the strongest contributions to the binding energy from Tyr34 (site 2) and Tyr36 (site 1). Compared to other protein-protein complexes, the binding is insensitive to amino acid substitutions at these sites, consistent with the large number of main chain interactions relative to side chain interactions. The strong binding of protein L to Ig kappa light chains of various species may result from the ambidextrous binding of the B1-B5 domains and the unimportance of specific side chain interactions.


Assuntos
Substituição de Aminoácidos , Proteínas de Bactérias , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Cadeias kappa de Imunoglobulina/química , Cadeias kappa de Imunoglobulina/metabolismo , Alanina/genética , Substituição de Aminoácidos/genética , Sítios de Ligação/genética , Cromatografia em Gel , Biologia Computacional/métodos , Simulação por Computador , Proteínas de Ligação a DNA/genética , Dimerização , Dissulfetos/química , Humanos , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/metabolismo , Cinética , Mutagênese Sítio-Dirigida , Ligação Proteica/genética , Radioimunoensaio , Espectrometria de Fluorescência , Ressonância de Plasmônio de Superfície , Termodinâmica , Fatores de Tempo , Triptofano/genética , Tirosina/genética
14.
J Immunol ; 172(6): 3798-807, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15004185

RESUMO

Group A streptococci (GAS) are common human pathogens that express major surface-associated virulence factors designated M proteins. In this study, we explored directly the cellular mechanisms behind their supposed ability to prevent phagocytosis. Isolated human neutrophils killed an M-negative GAS mutant (DeltaM5), but not the wild-type parent strain (M5). After 3 h, 3-4 times as many DeltaM5 as M5 bacteria were associated with the neutrophils, and more DeltaM5 than M5 bacteria were ingested. However, there was no statistically significant difference between DeltaM5 and M5 bacteria in regard to the percentage of the neutrophil-associated bacteria that were ingested, indicating that M5 protein prevents an adhesion receptor-dependent association with neutrophils and not the phagocytic machinery per se. Different Abs against CD11b/CD18 (CR3) blocked adhesion and killing of DeltaM5 bacteria, whereas the blocking of two other complement receptors, CD11c/CD18 (CR4) and CD35 (CR1), did not. The CD11b/CD18-mediated killing of DeltaM5 bacteria resulted in protein tyrosine phosphorylations and Cdc42 activation. Furthermore, inhibition of CD11b/CD18 receptor engagement or tyrosine kinase activity blocked the DeltaM5-induced activation of Cdc42 as well as the killing of these bacteria. We conclude that M5 protein interferes with the CD11b/CD18-dependent association between GAS and neutrophils, and thereby blocks subsequent ingestion of the bacteria.


Assuntos
Antígenos de Bactérias/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Antígeno CD11b/fisiologia , Antígenos CD18/fisiologia , Proteínas de Transporte/fisiologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Fagocitose/imunologia , Transdução de Sinais/imunologia , Streptococcus pyogenes/imunologia , Anticorpos Bloqueadores/farmacologia , Antígenos de Bactérias/genética , Aderência Bacteriana/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Antígeno CD11b/imunologia , Antígenos CD18/imunologia , Proteínas de Transporte/genética , Ativação do Complemento/imunologia , Humanos , Imunossupressores/farmacologia , Neutrófilos/metabolismo , Fosforilação , Proteínas Tirosina Quinases/fisiologia , Receptores de Complemento/antagonistas & inibidores , Receptores de Complemento/fisiologia , Streptococcus pyogenes/genética , Tirosina/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteína RAC2 de Ligação ao GTP
15.
Eur J Biochem ; 270(10): 2303-11, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12752450

RESUMO

Several microbial pathogens have been reported to interact with glycosaminoglycans (GAGs) on cell surfaces and in the extracellular matrix. Here we demonstrate that M protein, a major surface-expressed virulence factor of the human bacterial pathogen, Streptococcus pyogenes, mediates binding to various forms of GAGs. Hence, S. pyogenes strains expressing a large number of different types of M proteins bound to dermatan sulfate (DS), highly sulfated fractions of heparan sulfate (HS) and heparin, whereas strains deficient in M protein surface expression failed to interact with these GAGs. Soluble M protein bound DS directly and could also inhibit the interaction between DS and S. pyogenes. Experiments with M protein fragments and with streptococci expressing deletion constructs of M protein, showed that determinants located in the NH2-terminal part as well as in the C-repeat region of the streptococcal proteins are required for full binding to GAGs. Treatment with ABC-chondroitinase and HS lyase that specifically remove DS and HS chains from cell surfaces, resulted in significantly reduced adhesion of S. pyogenes bacteria to human epithelial cells and skin fibroblasts. Together with the finding that exogenous DS and HS could inhibit streptococcal adhesion, these data suggest that GAGs function as receptors in M protein-mediated adhesion of S. pyogenes.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/química , Glicosaminoglicanos/metabolismo , Streptococcus pyogenes/metabolismo , Fatores de Virulência/química , Proteínas de Bactérias/metabolismo , Adesão Celular , Dermatan Sulfato/química , Dermatan Sulfato/metabolismo , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Células Epiteliais/citologia , Deleção de Genes , Glicosaminoglicanos/química , Humanos , Ligação Proteica , Células Tumorais Cultivadas
16.
Microbiology (Reading) ; 148(Pt 12): 3933-3945, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12480897

RESUMO

Primary infection by group A streptococci (GAS) takes place at either the throat or skin of the human host, often leading to pharyngitis or impetigo, respectively. Many GAS strains differ in their preference for throat and skin tissue sites. Previous epidemiological findings show that many of the strains displaying strong tropism for the skin have a high-affinity binding site for plasminogen, located within M protein (PAM), a prominent surface fibril. Plasminogen bound by PAM interacts with streptokinase, a plasminogen activator secreted by GAS, to yield bacterial-bound plasmin activity. In this study, PAM and streptokinase were tested for their roles in infection using an experimental model that closely mimics human impetigo. Inactivation of genes encoding either PAM or streptokinase led to a partial, but significant, loss of virulence in vivo, as measured by net growth of the bacteria and pathological alterations. The relative loss in virulence in vivo was greater for the streptokinase mutant than for the PAM mutant. However, the PAM mutant, but not the streptokinase mutant, displayed a partial loss in resistance to phagocytosis in vitro. The combined experimental and epidemiological data provide evidence that PAM and streptokinase play a key role in mediating skin-specific infection by GAS. In addition, secreted cysteine proteinase activity due to SpeB leads to degradation of streptokinase in stationary phase broth cultures. Since SpeB is also a determinant of tissue-specific GAS infection at the skin, direct interactions between these two proteolytic pathways may constitute an important pathogenic mechanism. An integrated model for superficial infection at the skin is presented.


Assuntos
Antígenos de Bactérias , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Impetigo/fisiopatologia , Plasminogênio/metabolismo , Streptococcus pyogenes/patogenicidade , Estreptoquinase/metabolismo , Animais , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/genética , Proteínas de Transporte/genética , Modelos Animais de Doenças , Fibrinolisina/metabolismo , Humanos , Impetigo/microbiologia , Camundongos , Camundongos SCID , Ativadores de Plasminogênio/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus pyogenes/genética , Streptococcus pyogenes/crescimento & desenvolvimento , Estreptoquinase/genética , Virulência
17.
Infect Immun ; 70(12): 6646-51, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12438337

RESUMO

The human pathogen Streptococcus pyogenes primarily infects the upper respiratory tract and skin, but occasionally it disseminates and causes severe invasive disease with high mortality. This study revealed that the activity of extracellular EndoS, which hydrolyzes the functionally important N-linked oligosaccharides on opsonizing immunoglobulin G (IgG), contributes to increased survival of S. pyogenes in human blood ex vivo. The inability to kill the bacteria is due to reduced binding of IgG to Fc receptors and impaired classical pathway-mediated activation of complement. In addition, the activity of extracellular SpeB, which cleaves IgG into Fc and Fab fragments, also increases bacterial survival. This suggests that S. pyogenes expresses two enzymes, EndoS and SpeB, which modulate IgG by different mechanisms in order to evade the adaptive immune system.


Assuntos
Cisteína Endopeptidases/metabolismo , Glicosídeo Hidrolases/metabolismo , Imunoglobulina G/metabolismo , Fagocitose/imunologia , Streptococcus pyogenes/enzimologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sangue/microbiologia , Ativação do Complemento , Cisteína Endopeptidases/genética , Glicosídeo Hidrolases/genética , Humanos , Proteínas Opsonizantes , Receptores Fc/metabolismo , Streptococcus pyogenes/patogenicidade
18.
Blood ; 100(13): 4470-7, 2002 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12393652

RESUMO

We have characterized 2 distinct mechanisms through which infectious agents may promote platelet adhesion and thrombus formation in flowing blood, thus contributing to the progression of disease. In one case, the process initiates when the integrin alpha(IIb)beta(3) mediates platelet arrest onto immobilized bacterial constituents that have bound plasma fibrinogen. If blood contains antibodies against the bacteria, immunoglobulin (Ig) G may cluster on the same surface and activate adherent platelets through the Fc(gamma)RIIA receptor, leading to thrombus growth. As an alternative, bacteria that cannot bind fibrinogen may attach to substrates, such as immobilized plasma proteins or components of the extracellular matrix, which also support platelet adhesion. As a result of this colocalization, IgG bound to bacteria can activate neighboring platelets and induce thrombus growth regardless of their ability to initiate platelet-surface contact. Our results demonstrate that intrinsic constituents of infectious agents and host proteins play distinct but complementary roles in recruiting platelets into thrombi, possibly contributing to complications of acute and chronic infections.


Assuntos
Antígenos de Bactérias , Plaquetas/microbiologia , Adesividade Plaquetária , Staphylococcus aureus/fisiologia , Streptococcus pyogenes/fisiologia , Trombose/microbiologia , Adulto , Anticorpos Antibacterianos/imunologia , Anticorpos Antibacterianos/metabolismo , Antígenos CD/metabolismo , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Fibrinogênio/metabolismo , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Modelos Biológicos , Ativação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ligação Proteica , Receptores de IgG/metabolismo , Streptococcus pyogenes/imunologia , Trombose/etiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...