Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 116(17): 8481-8486, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30948644

RESUMO

Despite the availability of antibiotics and vaccines, Neisseria meningitidis remains a major cause of meningitis and sepsis in humans. Due to its extracellular lifestyle, bacterial adhesion to host cells constitutes an attractive therapeutic target. Here, we present a high-throughput microscopy-based approach that allowed the identification of compounds able to decrease type IV pilus-mediated interaction of bacteria with endothelial cells in the absence of bacterial or host cell toxicity. Compounds specifically inhibit the PilF ATPase enzymatic activity that powers type IV pilus extension but remain inefficient on the ATPase that promotes pilus retraction, thus leading to rapid pilus disappearance from the bacterial surface and loss of pili-mediated functions. Structure activity relationship of the most active compound identifies specific moieties required for the activity of this compound and highlights its specificity. This study therefore provides compounds targeting pilus biogenesis, thereby inhibiting bacterial adhesion, and paves the way for a novel therapeutic option for meningococcal infections.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Fímbrias Bacterianas , Adenosina Trifosfatases/antagonistas & inibidores , Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Células Cultivadas , Fímbrias Bacterianas/efeitos dos fármacos , Fímbrias Bacterianas/metabolismo , Ensaios de Triagem em Larga Escala , Células Endoteliais da Veia Umbilical Humana , Humanos , Neisseria meningitidis/enzimologia , Neisseria meningitidis/patogenicidade
2.
Bioorg Med Chem ; 25(16): 4245-4252, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28687228

RESUMO

Membrane-active foldamers have recently emerged as potential mimics of antimicrobial peptides (AMPs). Amphiphilic cationic helical N,N'-linked oligoureas are one such class of AMP mimics with activities in vitro against a broad range of bacteria including Bacillus anthracis, a Gram-positive sporulating bacillus and causing agent of anthrax. Here we have used site-selective chemical modifications of the oligourea backbone to gain additional insight into the relationship between structure and function and modulate anthracidal activity. A series of analogues in which urea linkages at selected positions are replaced by thiourea and guanidium surrogates have been prepared on solid support and tested against different bacterial forms of B. anthracis (germinated spores and encapsulated bacilli). Urea→thiourea and urea→guanidinium replacements close to the negative end of the helix dipole led to analogues with increased potency and selectivity for B. anthracis versus mammalian cells.


Assuntos
Antibacterianos/farmacologia , Bacillus anthracis/efeitos dos fármacos , Guanidina/farmacologia , Tioureia/farmacologia , Ureia/farmacologia , Animais , Antibacterianos/síntese química , Antibacterianos/química , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Guanidina/química , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Células RAW 264.7 , Relação Estrutura-Atividade , Tioureia/química , Ureia/análogos & derivados , Ureia/química
3.
J Med Chem ; 59(18): 8221-32, 2016 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-27529632

RESUMO

The synthesis of bioinspired unnatural backbones leading to foldamers can provide effective peptide mimics with improved properties in a physiological environment. This approach has been applied to the design of structural mimics of membrane active antimicrobial peptides (AMPs) for which activities in vitro have been reported. Yet activities and pharmacokinetic properties in vivo in animal models have remained largely unexplored. Here, we report helical oligourea AMP mimics that are active in vitro against bacterial forms of Bacillus anthracis encountered in vivo, as well as in vivo in inhalational and cutaneous mouse models of B. anthracis infection. The pharmacokinetic profile and the tissue distribution were investigated by ß-radio imager whole-body mapping in mice. Low excretion and recovery of the native oligourea in the kidney following intravenous injection is consistent with high stability in vivo. Overall these results provide useful information that support future biomedical development of urea-based foldamer peptide mimics.


Assuntos
Antraz/tratamento farmacológico , Antibacterianos/uso terapêutico , Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Bacillus anthracis/efeitos dos fármacos , Peptidomiméticos/uso terapêutico , Ureia/uso terapêutico , Animais , Antibacterianos/química , Antibacterianos/farmacocinética , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacocinética , Peptídeos Catiônicos Antimicrobianos/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Peptidomiméticos/química , Peptidomiméticos/farmacocinética , Peptidomiméticos/farmacologia , Ureia/análogos & derivados , Ureia/farmacocinética , Ureia/farmacologia
4.
J Infect Dis ; 214(2): 281-7, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-26977051

RESUMO

The lung is the terminal target of Bacillus anthracis before death, whatever the route of infection (cutaneous, inhalational, or digestive). During a cutaneous infection in absence of toxins, we observed encapsulated bacteria colonizing the alveolar capillary network, bacteria and hemorrhages in alveolar and bronchiolar spaces, and hypoxic foci in the lung (endothelial cells) and brain (neurons and neuropil). Circulating encapsulated bacteria were as chains of approximately 13 µm in length. Bacteria of such size were immediately trapped within the lung capillary network, but bacteria of shorter length were not. Controlling lung-targeted pathology would be beneficial for anthrax treatment.


Assuntos
Antraz/microbiologia , Antraz/patologia , Bacillus anthracis/isolamento & purificação , Capilares/microbiologia , Pulmão/microbiologia , Animais , Capilares/patologia , Modelos Animais de Doenças , Histocitoquímica , Imuno-Histoquímica , Pulmão/patologia , Camundongos , Microscopia Eletrônica de Transmissão
5.
PLoS Negl Trop Dis ; 9(4): e0003455, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25830379

RESUMO

Emerging B. cereus strains that cause anthrax-like disease have been isolated in Cameroon (CA strain) and Côte d'Ivoire (CI strain). These strains are unusual, because their genomic characterisation shows that they belong to the B. cereus species, although they harbour two plasmids, pBCXO1 and pBCXO2, that are highly similar to the pXO1 and pXO2 plasmids of B. anthracis that encode the toxins and the polyglutamate capsule respectively. The virulence factors implicated in the pathogenicity of these B. cereus bv anthracis strains remain to be characterised. We tested their virulence by cutaneous and intranasal delivery in mice and guinea pigs; they were as virulent as wild-type B. anthracis. Unlike as described for pXO2-cured B. anthracis, the CA strain cured of the pBCXO2 plasmid was still highly virulent, showing the existence of other virulence factors. Indeed, these strains concomitantly expressed a hyaluronic acid (HA) capsule and the B. anthracis polyglutamate (PDGA) capsule. The HA capsule was encoded by the hasACB operon on pBCXO1, and its expression was regulated by the global transcription regulator AtxA, which controls anthrax toxins and PDGA capsule in B. anthracis. Thus, the HA and PDGA capsules and toxins were co-regulated by AtxA. We explored the respective effect of the virulence factors on colonisation and dissemination of CA within its host by constructing bioluminescent mutants. Expression of the HA capsule by itself led to local multiplication and, during intranasal infection, to local dissemination to the adjacent brain tissue. Co-expression of either toxins or PDGA capsule with HA capsule enabled systemic dissemination, thus providing a clear evolutionary advantage. Protection against infection by B. cereus bv anthracis required the same vaccination formulation as that used against B. anthracis. Thus, these strains, at the frontier between B. anthracis and B. cereus, provide insight into how the monomorphic B. anthracis may have emerged.


Assuntos
Antraz/microbiologia , Antígenos de Bactérias/metabolismo , Bacillus anthracis/metabolismo , Cápsulas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Fatores de Virulência/metabolismo , Animais , Antígenos de Bactérias/genética , Bacillus anthracis/genética , Bacillus anthracis/patogenicidade , Bacillus cereus/classificação , Bacillus cereus/genética , Bacillus cereus/metabolismo , Cápsulas Bacterianas/genética , Toxinas Bacterianas/genética , Genômica , Camundongos , Plasmídeos , Toxinas Biológicas , Virulência/genética , Fatores de Virulência/genética
6.
J Infect Dis ; 207(3): 450-7, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23148288

RESUMO

BACKGROUND: Germination is a key step for successful Bacillus anthracis colonization and systemic dissemination. Few data are available on spore germination in vivo, and the necessity of spore and host cell interactions to initiate germination is unclear. METHODS: To investigate the early interactions between B. anthracis spores and cutaneous tissue, spores were inoculated in an intraperitoneal cell-free device in guinea pigs or into the pinna of mice. Germination and bacterial growth were analyzed through colony-forming unit enumeration and electron microscopy. RESULTS: In the guinea pig model, germination occurred in vivo in the absence of cell contact. Similarly, in the mouse ear, germination started within 15 minutes after inoculation, and germinating spores were found in the absence of surrounding cells. Germination was not observed in macrophage-rich draining lymph nodes, liver, and spleen. Edema and lethal toxin production were not required for germination, as a toxin-deficient strain was as effective as a Sterne-like strain. B. anthracis growth was locally controlled for 6 hours. CONCLUSIONS: Spore germination involving no cell interactions can occur in vivo, suggesting that diffusible germinants or other signals appear sufficient. Different host tissues display drastic differences in germination-triggering capacity. Initial control of bacterial growth suggests a therapeutic means to exploit host innate defenses to hinder B. anthracis colonization.


Assuntos
Antraz/microbiologia , Bacillus anthracis/crescimento & desenvolvimento , Dermatopatias Bacterianas/microbiologia , Esporos Bacterianos/crescimento & desenvolvimento , Animais , Bacillus anthracis/ultraestrutura , Contagem de Colônia Microbiana , Modelos Animais de Doenças , Feminino , Cobaias , Interações Hospedeiro-Patógeno , Fígado/microbiologia , Tecido Linfoide/microbiologia , Camundongos , Baço/microbiologia , Esporos Bacterianos/ultraestrutura
7.
PLoS Pathog ; 8(1): e1002481, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22253596

RESUMO

NK cells are important immune effectors for preventing microbial invasion and dissemination, through natural cytotoxicity and cytokine secretion. Bacillus anthracis spores can efficiently drive IFN-γ production by NK cells. The present study provides insights into the mechanisms of cytokine and cellular signaling that underlie the process of NK-cell activation by B. anthracis and the bacterial strategies to subvert and evade this response. Infection with non-toxigenic encapsulated B. anthracis induced recruitment of NK cells and macrophages into the mouse draining lymph node. Production of edema (ET) or lethal (LT) toxin during infection impaired this cellular recruitment. NK cell depletion led to accelerated systemic bacterial dissemination. IFN-γ production by NK cells in response to B. anthracis spores was: i) contact-dependent through RAE-1-NKG2D interaction with macrophages; ii) IL-12, IL-18, and IL-15-dependent, where IL-12 played a key role and regulated both NK cell and macrophage activation; and iii) required IL-18 for only an initial short time window. B. anthracis toxins subverted both NK cell essential functions. ET and LT disrupted IFN-γ production through different mechanisms. LT acted both on macrophages and NK cells, whereas ET mainly affected macrophages and did not alter NK cell capacity of IFN-γ secretion. In contrast, ET and LT inhibited the natural cytotoxicity function of NK cells, both in vitro and in vivo. The subverting action of ET thus led to dissociation in NK cell function and blocked natural cytotoxicity without affecting IFN-γ secretion. The high efficiency of this process stresses the impact that this toxin may exert in anthrax pathogenesis, and highlights a potential usefulness for controlling excessive cytotoxic responses in immunopathological diseases. Our findings therefore exemplify the delicate balance between bacterial stimulation and evasion strategies. This highlights the potential implication of the crosstalk between host innate defences and B. anthracis in initial anthrax control mechanisms.


Assuntos
Bacillus anthracis/imunologia , Toxinas Bacterianas/farmacologia , Células Matadoras Naturais/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Esporos Bacterianos/imunologia , Animais , Células Cultivadas , Feminino , Homeostase/efeitos dos fármacos , Homeostase/imunologia , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor Cross-Talk/efeitos dos fármacos , Receptor Cross-Talk/imunologia , Esporos Bacterianos/fisiologia
8.
Am J Pathol ; 178(6): 2523-35, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21641378

RESUMO

Powerful noninvasive imaging technologies enable real-time tracking of pathogen-host interactions in vivo, giving access to previously elusive events. We visualized the interactions between wild-type Bacillus anthracis and its host during a spore infection through bioluminescence imaging coupled with histology. We show that edema toxin plays a central role in virulence in guinea pigs and during inhalational infection in mice. Edema toxin (ET), but not lethal toxin (LT), markedly modified the patterns of bacterial dissemination leading, to apparent direct dissemination to the spleen and provoking apoptosis of lymphoid cells. Each toxin alone provoked particular histological lesions in the spleen. When ET and LT are produced together during infection, a specific temporal pattern of lesion developed, with early lesions typical of LT, followed at a later stage by lesions typical of ET. Our study provides new insights into the complex spatial and temporal effects of B. anthracis toxins in the infected host, suggesting a greater role than previously suspected for ET in anthrax and suggesting that therapeutic targeting of ET contributes to protection.


Assuntos
Antraz/microbiologia , Antraz/patologia , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Diagnóstico por Imagem/métodos , Fatores de Virulência/imunologia , Animais , Antraz/prevenção & controle , Apoptose , Bacillus anthracis/patogenicidade , Feminino , Cobaias/microbiologia , Exposição por Inalação , Luminescência , Camundongos , Camundongos Endogâmicos BALB C , Nasofaringe/microbiologia , Nasofaringe/patologia , Testes de Neutralização , Pele/microbiologia , Pele/patologia , Baço/microbiologia , Baço/patologia , Fatores de Tempo
9.
J Infect Dis ; 200(9): 1381-9, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19785525

RESUMO

BACKGROUND: The Bacillus anthracis poly-gamma-D-glutamate capsule is essential for virulence. It impedes phagocytosis and protects bacilli from the immune system, thus promoting systemic dissemination. METHODS: To further define the virulence mechanisms brought into play by the capsule, we characterized the interactions between encapsulated nontoxinogenic B. anthracis and its host in vivo through histological analysis, perfusion, and competition experiments with purified capsule. RESULTS: Clearance of encapsulated bacilli from the blood was rapid (>90% clearance within 5 min), with 75% of the bacteria being trapped in the liver. Competition experiments with purified capsule polyglutamate inhibited this interaction. At the septicemic phase of cutaneous infection with spores, the encapsulated bacilli were trapped in the vascular spaces of the liver and interacted closely with the liver endothelium in the sinusoids and terminal and portal veins. They often grow as microcolonies containing capsular material shed by the bacteria. CONCLUSION: We show that, in addition to its inhibitory effect on the interaction with the immune system, the capsule surrounding B. anthracis plays an active role in mediating the trapping of the bacteria within the liver and may thus contribute to anthrax pathogenesis. Because other microorganisms produce polyglutamate, it may also represent a general mechanism of virulence or in vivo survival.


Assuntos
Antraz/microbiologia , Bacillus anthracis/patogenicidade , Cápsulas Bacterianas/fisiologia , Endotélio/microbiologia , Fígado/microbiologia , Animais , Feminino , Interações Hospedeiro-Patógeno , Camundongos , Ácido Poliglutâmico/fisiologia , Virulência
10.
Infect Immun ; 77(3): 1197-207, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19114543

RESUMO

Protective antigen (PA)-based anthrax vaccines acting on toxins are less effective than live attenuated vaccines, suggesting that additional antigens may contribute to protective immunity. Several reports indicate that capsule or spore-associated antigens may enhance the protection afforded by PA. Addition of formaldehyde-inactivated spores (FIS) to PA (PA-FIS) elicits total protection against cutaneous anthrax. Nevertheless, vaccines that are effective against cutaneous anthrax may not be so against inhalational anthrax. The aim of this work was to optimize immunization with PA-FIS and to assess vaccine efficacy against inhalational anthrax. We assessed the immune response to recombinant anthrax PA from Bacillus anthracis (rPA)-FIS administered by various immunization protocols and the protection provided to mice and guinea pigs infected through the respiratory route with spores of a virulent strain of B. anthracis. Combined subcutaneous plus intranasal immunization of mice yielded a mucosal immunoglobulin G response to rPA that was more than 20 times higher than that in lung mucosal secretions after subcutaneous vaccination. The titers of toxin-neutralizing antibody and antispore antibody were also significantly higher: nine and eight times higher, respectively. The optimized immunization elicited total protection of mice intranasally infected with the virulent B. anthracis strain 17JB. Guinea pigs were fully protected, both against an intranasal challenge with 100 50% lethal doses (LD(50)) and against an aerosol with 75 LD(50) of spores of the highly virulent strain 9602. Conversely, immunization with PA alone did not elicit protection. These results demonstrate that the association of PA and spores is very much more effective than PA alone against experimental inhalational anthrax.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Esporos Bacterianos/imunologia , Administração por Inalação , Administração Intranasal , Animais , Antraz/imunologia , Vacinas contra Antraz/administração & dosagem , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/administração & dosagem , Bacillus anthracis/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Cobaias , Injeções Subcutâneas , Pneumopatias/imunologia , Pneumopatias/microbiologia , Pneumopatias/prevenção & controle , Camundongos
11.
Infect Immun ; 75(10): 4754-61, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17635863

RESUMO

Bacillus anthracis is a spore-forming bacterium that causes anthrax. B. anthracis has three major virulence factors, namely, lethal toxin, edema toxin, and a poly-gamma-D-glutamic acid capsule. The toxins modulate host immune responses, and the capsule inhibits phagocytosis. With the goal of increasing safety, decreasing security concerns, and taking advantage of mammalian genetic tools and reagents, mouse models of B. anthracis infection have been developed using attenuated bacteria that produce toxins but no capsule. While these models have been useful in studying both toxinogenic infections and antitoxin vaccine efficacy, we questioned whether eliminating the capsule changed bacterial growth and dissemination characteristics. Thus, the progression of infection by toxinogenic noncapsulated B. anthracis was analyzed and compared to that by previously reported nontoxinogenic capsulated bacteria, using in vivo bioluminescence imaging. The influence of immunization with the toxin component protective antigen (PA) on the development of infection was also examined. The toxinogenic noncapsulated bacteria were initially confined to the cutaneous site of infection. Bacteria then progressed to the draining lymph nodes and, finally, late in the infection, to the lungs, kidneys, and frequently the gastrointestinal tract. There was minimal colonization of the spleen. PA immunization reduced bacterial growth from the outset and limited infection to the site of inoculation. These in vivo observations show that dissemination by toxinogenic noncapsulated strains differs markedly from that by nontoxinogenic capsulated strains. Additionally, PA immunization counters bacterial growth and dissemination in vivo from the onset of infection.


Assuntos
Antraz/imunologia , Antraz/microbiologia , Antígenos de Bactérias/biossíntese , Antígenos de Bactérias/imunologia , Bacillus anthracis/crescimento & desenvolvimento , Bacillus anthracis/imunologia , Cápsulas Bacterianas/imunologia , Toxinas Bacterianas/biossíntese , Toxinas Bacterianas/imunologia , Animais , Antraz/patologia , Bacillus anthracis/classificação , Cápsulas Bacterianas/biossíntese , Trato Gastrointestinal/microbiologia , Processamento de Imagem Assistida por Computador , Rim/microbiologia , Medições Luminescentes , Pulmão/microbiologia , Linfonodos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Pele/microbiologia , Especificidade da Espécie , Organismos Livres de Patógenos Específicos
12.
J Immunol ; 178(5): 2646-50, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17312104

RESUMO

Virulent strains of Bacillus anthracis produce immunomodulating toxins and an antiphagocytic capsule. The toxin component-protective Ag is a key target of the antianthrax immune response that induces production of toxin-neutralizing Abs. Coimmunization with spores enhances the antitoxin vaccine, and inactivated spores alone confer measurable protection. We aimed to identify the mechanisms of protection induced in inactivated-spore immunized mice that function independently of the toxin/antitoxin vaccine system. This goal was addressed with humoral and CD4 T lymphocyte transfer, in vivo depletion of CD4 T lymphocytes and IFN-gamma, and Ab-deficient (muMT(-/-)) or IFN-gamma-insensitive (IFN-gammaR(-/-)) mice. We found that humoral immunity did not protect from nontoxinogenic capsulated bacteria, whereas a cellular immune response by IFN-gamma-producing CD4 T lymphocytes protected mice. These results are the first evidence of protective cellular immunity against capsulated B. anthracis and suggest that future antianthrax vaccines should strive to augment cellular adaptive immunity.


Assuntos
Vacinas contra Antraz/imunologia , Antraz/imunologia , Bacillus anthracis/imunologia , Cápsulas Bacterianas/imunologia , Interferon gama/imunologia , Esporos Bacterianos/imunologia , Animais , Antraz/prevenção & controle , Anticorpos Antibacterianos/genética , Anticorpos Antibacterianos/imunologia , Formação de Anticorpos , Feminino , Imunidade Celular , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Interferon/deficiência , Receptores de Interferon/imunologia , Receptor de Interferon gama
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...