Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 18(9): e1010851, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36174087

RESUMO

During infection, Bacillus anthracis bacilli encounter potent antimicrobial peptides (AMPs) such as defensins. We examined the role that B. anthracis capsule plays in protecting bacilli from defensins and other cationic AMPs by comparing their effects on a fully virulent encapsulated wild type (WT) strain and an isogenic capsule-deficient capA mutant strain. We identified several human defensins and non-human AMPs that were capable of killing B. anthracis. The human alpha defensins 1-6 (HNP-1-4, HD-5-6), the human beta defensins 1-4 (HBD-1-4), and the non-human AMPs, protegrin, gramicidin D, polymyxin B, nisin, and melittin were all capable of killing both encapsulated WT and non-encapsulated capA mutant B. anthracis. However, non-encapsulated capA mutant bacilli were significantly more susceptible than encapsulated WT bacilli to killing by nearly all of the AMPs tested. We demonstrated that purified capsule bound HBD-2, HBD-3, and HNP-1 in an electrophoretic mobility shift assay. Furthermore, we determined that the capsule layer enveloping WT bacilli bound and trapped HBD-3, substantially reducing the amount reaching the cell wall. To assess whether released capsule might also play a protective role, we pre-incubated HBD-2, HBD-3, or HNP-1 with purified capsule before their addition to non-encapsulated capA mutant bacilli. We found that free capsule completely rescued the capA mutant bacilli from killing by HBD-2 and -3 while killing by HNP-1 was reduced to the level observed with WT bacilli. Together, these results suggest an immune evasion mechanism by which the capsule, both that enveloping the bacilli and released fragments, contributes to virulence by binding to and inhibiting the antimicrobial activity of cationic AMPs.


Assuntos
Bacillus anthracis , Nisina , alfa-Defensinas , beta-Defensinas , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Peptídeos Antimicrobianos , Defensinas/genética , Defensinas/farmacologia , Gramicidina , Humanos , Meliteno , Polimixina B , alfa-Defensinas/farmacologia
2.
J Am Assoc Lab Anim Sci ; 56(6): 792-801, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29256375

RESUMO

We used a continuous-monitoring digital telemetry system to investigate temperature response in New Zealand White rabbits after inhalation or subcutaneous challenge with Bacillus anthracis. Two spore preparations of B. anthracis Ames A2084 were evaluated by using a nose-only inhalation model, and 2 strains, B. anthracis Ames A2084 and B. anthracis UT500, were evaluated in a subcutaneous model. Animal body temperature greater than 3 SD above the mean baseline temperature was considered a significant increase in body temperature (SIBT). All rabbits that exhibited SIBT after challenge by either route of infection or bacterial strain eventually died or were euthanized due to infection, and all rabbits that died or were euthanized due to infection exhibited SIBT during the course of disease. The time at onset of SIBT preceded clinical signs of disease in 94% of the rabbits tested by as long as 2 days. In addition, continuous temperature monitoring facilitated discrimination between the 2 B. anthracis strains with regard to the time interval between SIBT and death. These data suggest that for the New Zealand White rabbit anthrax model, SIBT is a reliable indicator of infection, is predictive of experimental outcome in the absence of treatment, and is measurable prior to the appearance of more severe signs of disease. The use of digital telemetry to monitor infectious disease course in animal models of anthrax can potentially be used in conjunction with other clinical score metrics to refine endpoint euthanasia criteria.


Assuntos
Antraz/veterinária , Coelhos , Telemetria/veterinária , Aerossóis , Animais , Animais de Laboratório , Antraz/microbiologia , Antraz/fisiopatologia , Bacillus anthracis/química , Bacillus anthracis/classificação , Bacillus anthracis/patogenicidade , Temperatura Corporal , Modelos Animais de Doenças , Febre/veterinária , Inalação
3.
Biochemistry ; 51(6): 1199-212, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22257032

RESUMO

γ-Glutamyl transpeptidase (GGT) is a two-substrate enzyme that plays a central role in glutathione metabolism and is a potential target for drug design. GGT catalyzes the cleavage of γ-glutamyl donor substrates and the transfer of the γ-glutamyl moiety to an amine of an acceptor substrate or water. Although structures of bacterial GGT have revealed details of the protein-ligand interactions at the donor site, the acceptor substrate site is relatively undefined. The recent identification of a species-specific acceptor site inhibitor, OU749, suggests that these inhibitors may be less toxic than glutamine analogues. Here we investigated the donor and acceptor substrate preferences of Bacillus anthracis GGT (CapD) and applied computational approaches in combination with kinetics to probe the structural basis of the enzyme's substrate and inhibitor binding specificities and compare them with human GGT. Site-directed mutagenesis studies showed that the R432A and R520S variants exhibited 6- and 95-fold decreases in hydrolase activity, respectively, and that their activity was not stimulated by the addition of the l-Cys acceptor substrate, suggesting an additional role in acceptor binding and/or catalysis of transpeptidation. Rat GGT (and presumably HuGGT) has strict stereospecificity for L-amino acid acceptor substrates, while CapD can utilize both L- and D-acceptor substrates comparably. Modeling and kinetic analysis suggest that R520 and R432 allow two alternate acceptor substrate binding modes for L- and D-acceptors. R432 is conserved in Francisella tularensis, Yersinia pestis, Burkholderia mallei, Helicobacter pylori and Escherichia coli, but not in human GGT. Docking and MD simulations point toward key residues that contribute to inhibitor and acceptor substrate binding, providing a guide to designing novel and specific GGT inhibitors.


Assuntos
Proteínas de Bactérias/química , gama-Glutamiltransferase/química , Animais , Bacillus anthracis/enzimologia , Cápsulas Bacterianas/química , Cápsulas Bacterianas/genética , Cápsulas Bacterianas/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Domínio Catalítico , Biologia Computacional/métodos , Glutationa/metabolismo , Humanos , Ligantes , Camundongos , Ligação Proteica , Ratos , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Sulfonamidas/metabolismo , Sulfonamidas/farmacologia , Suínos , Tiadiazóis/metabolismo , Tiadiazóis/farmacologia , gama-Glutamiltransferase/antagonistas & inibidores , gama-Glutamiltransferase/genética
4.
Infect Immun ; 79(4): 1512-25, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21300775

RESUMO

The Burkholderia pseudomallei K96243 genome encodes six type VI secretion systems (T6SSs), but little is known about the role of these systems in the biology of B. pseudomallei. In this study, we purified recombinant Hcp proteins from each T6SS and tested them as vaccine candidates in the BALB/c mouse model of melioidosis. Recombinant Hcp2 protected 80% of mice against a lethal challenge with K96243, while recombinant Hcp1, Hcp3, and Hcp6 protected 50% of mice against challenge. Hcp6 was the only Hcp constitutively produced by B. pseudomallei in vitro; however, it was not exported to the extracellular milieu. Hcp1, on the other hand, was produced and exported in vitro when the VirAG two-component regulatory system was overexpressed in trans. We also constructed six hcp deletion mutants (Δhcp1 through Δhcp6) and tested them for virulence in the Syrian hamster model of infection. The 50% lethal doses (LD(50)s) for the Δhcp2 through Δhcp6 mutants were indistinguishable from K96243 (<10 bacteria), but the LD(50) for the Δhcp1 mutant was >10(3) bacteria. The hcp1 deletion mutant also exhibited a growth defect in RAW 264.7 macrophages and was unable to form multinucleated giant cells in this cell line. Unlike K96243, the Δhcp1 mutant was only weakly cytotoxic to RAW 264.7 macrophages 18 h after infection. The results suggest that the cluster 1 T6SS is essential for virulence and plays an important role in the intracellular lifestyle of B. pseudomallei.


Assuntos
Perfilação da Expressão Gênica , Melioidose/microbiologia , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/metabolismo , Cricetinae , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Feminino , Imunofluorescência , Expressão Gênica , Genes Bacterianos , Humanos , Immunoblotting , Fígado/microbiologia , Fígado/patologia , Macrófagos/microbiologia , Macrófagos/patologia , Melioidose/genética , Melioidose/metabolismo , Mesocricetus , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Virulência/genética , Fatores de Virulência/genética
5.
Microbiology (Reading) ; 156(Pt 5): 1459-1467, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20110296

RESUMO

Capsule depolymerase (CapD) is a gamma-glutamyl transpeptidase and a product of the Bacillus anthracis capsule biosynthesis operon. In this study, we examined the effect of modulating capD expression on B. anthracis capsule phenotype, interaction with phagocytic cells and virulence in guinea pigs. Transcriptional fusions of capD were made to the genes encoding heat-shock protein 60 (hsp60) and elongation factor Tu (EFTu), and to capA, a B. anthracis capsule biosynthesis gene. Translation signals were altered to improve expression of capD, including replacing the putative ribosome-binding site with a consensus sequence and the TTG start codon with ATG. CapD was not detected by immunoblotting in lysates from wild-type B. anthracis Ames but was detected in strains engineered with a consensus ribosome-binding site for capD. Strains overexpressing capD at amounts detected by immunoblotting were found to have less surface-associated capsule and released primarily lower-molecular-mass capsule into culture supernatants. Overexpression of capD increased susceptibility to neutrophil phagocytic killing and adherence to macrophages and resulted in reduced fitness in a guinea pig model of infection. These data suggest that B. anthracis may have evolved weak capD expression resulting in optimized capsule-mediated virulence.


Assuntos
Bacillus anthracis/enzimologia , Bacillus anthracis/patogenicidade , Cápsulas Bacterianas/metabolismo , Glicosídeo Hidrolases/metabolismo , Animais , Bacillus anthracis/genética , Chaperonina 60/genética , Clonagem Molecular , Evolução Molecular , Glicosídeo Hidrolases/biossíntese , Glicosídeo Hidrolases/genética , Cobaias , Macrófagos/citologia , Macrófagos/metabolismo , Óperon , Fenótipo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Virulência/genética
6.
Antimicrob Agents Chemother ; 52(3): 1014-20, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18160516

RESUMO

Bacillus anthracis produces an antiphagocytic gamma-linked poly-D-glutamic acid capsule that is required for virulence. Capsule depolymerase (CapD) is a membrane-associated poly-gamma-glutamate-specific depolymerase encoded on the B. anthracis capsule plasmid, pX02, that is reported to contribute to virulence by anchoring the capsule to the peptidoglycan and partially degrading high-molecular-weight capsule from the bacterial surface. We previously demonstrated that treatment with CapD effectively removes the capsule from anthrax bacilli, rendering them susceptible to phagocytic killing in vitro. Here we report that CapD promoted in vivo phagocytic killing of B. anthracis bacilli by mouse peritoneal neutrophils and that parenteral administration of CapD protected mice in two models of anthrax infection. CapD conferred significant protection compared with controls when coinjected with encapsulated bacilli from fully virulent B. anthracis Ames or the nontoxigenic encapsulated strain Delta Ames and when injected 10 min after infection with encapsulated bacilli from B. anthracis Ames. Protection was also observed when CapD was administered 30 h after infection with B. anthracis Delta Ames spores, while significant protection could not be demonstrated following challenge with B. anthracis Ames spores. These data support the proposed role of capsule in B. anthracis virulence and suggest that strategies to target anthrax bacilli for neutrophil killing may lead to novel postexposure therapies.


Assuntos
Antraz/tratamento farmacológico , Bacillus anthracis/efeitos dos fármacos , Cápsulas Bacterianas/metabolismo , Glicosídeo Hidrolases/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Animais , Antraz/microbiologia , Bacillus anthracis/patogenicidade , Bacillus anthracis/fisiologia , Feminino , Glicosídeo Hidrolases/genética , Glicosídeo Hidrolases/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/imunologia , Fagocitose , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Esporos Bacterianos/fisiologia , Resultado do Tratamento , Virulência
7.
Antimicrob Agents Chemother ; 51(1): 215-22, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17074794

RESUMO

The poly-gamma-d-glutamic acid capsule confers antiphagocytic properties on Bacillus anthracis and is essential for virulence. In this study, we showed that CapD, a gamma-polyglutamic acid depolymerase encoded on the B. anthracis capsule plasmid, degraded purified capsule and removed the capsule from the surface of anthrax bacilli. Treatment with CapD induced macrophage phagocytosis of encapsulated B. anthracis and enabled human neutrophils to kill encapsulated organisms. A second glutamylase, PghP, a gamma-polyglutamic acid hydrolase encoded by Bacillus subtilis bacteriophage PhiNIT1, had minimal activity in degrading B. anthracis capsule, no effect on macrophage phagocytosis, and only minimal enhancement of neutrophil killing. Thus, the levels of both phagocytosis and killing corresponded to the degree of enzyme-mediated capsule degradation. The use of enzymes to degrade the capsule and enable phagocytic killing of B. anthracis offers a new approach to the therapy of anthrax.


Assuntos
Bacillus anthracis/metabolismo , Cápsulas Bacterianas/metabolismo , Ácido Poliglutâmico/metabolismo , gama-Glutamiltransferase/metabolismo , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Bacillus anthracis/efeitos dos fármacos , Bacillus anthracis/genética , Bacillus subtilis/efeitos dos fármacos , Bacillus subtilis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/farmacologia , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/farmacologia , Células Cultivadas , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Fagocitose/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , gama-Glutamil Hidrolase/metabolismo , gama-Glutamiltransferase/genética , gama-Glutamiltransferase/farmacologia
8.
Vaccine ; 23(1): 43-7, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15519706

RESUMO

Efficacy of a poly-gamma-D-glutamic acid anthrax capsule vaccine was assessed in a mouse model of infection. Capsule by itself was protective against lethal challenge with a toxin(-), capsule(+) Bacillus anthracis strain. Conjugation of capsule to bovine serum albumin resulted in enhanced IgG anti-capsule antibodies measured by ELISA, but completely abrogated the protection. The protective unconjugated capsule vaccine elicited significantly higher IgM titers and opsonic activity than did the non-protective capsule conjugate. When tested against a fully virulent toxin(+), capsule(+) B. anthracis strain, neither capsule nor protective antigen alone was protective. However, the combination of the two protected against a lethal challenge. These results suggest that capsule may enhance the protection afforded by protective antigen vaccines against anthrax if opsonizing antibodies are produced. Surprisingly, some protection was also observed when protective antigen was conjugated to itself.


Assuntos
Vacinas contra Antraz/administração & dosagem , Antraz/prevenção & controle , Bacillus anthracis/imunologia , Animais , Antraz/imunologia , Vacinas contra Antraz/química , Anticorpos Antibacterianos/biossíntese , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/química , Bacillus anthracis/química , Cápsulas Bacterianas , Toxinas Bacterianas/administração & dosagem , Toxinas Bacterianas/química , Toxinas Bacterianas/imunologia , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Camundongos , Ácido Poliglutâmico/administração & dosagem , Ácido Poliglutâmico/química , Ácido Poliglutâmico/imunologia , Vacinas Conjugadas/administração & dosagem , Vacinas Conjugadas/química
9.
Microb Pathog ; 37(3): 149-54, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15351038

RESUMO

A non-hemolytic enterotoxin (NHE) is one of the two enterotoxins thought to cause diarrhea produced by Bacillus cereus. We identified genes in Bacillus anthracis homologous to the B. cereus nheAB genes encoding proteins of the NHE complex. The NheA component was detected immunologically in culture supernatants from B. anthracis but not from a NheA(-) mutant, suggesting that B. anthracis produces and secretes the NheA subunit of NHE. A NheA deletion mutant was not attenuated in the guinea pig suggesting that NheA is not absolutely required for virulence.


Assuntos
Antraz/fisiopatologia , Bacillus anthracis/metabolismo , Bacillus cereus/metabolismo , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Enterotoxinas/metabolismo , Sequência de Aminoácidos , Animais , Antraz/microbiologia , Bacillus anthracis/genética , Bacillus anthracis/patogenicidade , Bacillus cereus/genética , Proteínas de Bactérias/genética , Toxinas Bacterianas/genética , Chlorocebus aethiops , Enterotoxinas/genética , Feminino , Regulação Bacteriana da Expressão Gênica , Cobaias , Dados de Sequência Molecular , Mutação , Células Vero , Virulência
10.
J Antimicrob Chemother ; 52(5): 790-5, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14563891

RESUMO

Pyrazinamide is an important sterilizing drug that shortens tuberculosis (TB) therapy. However, the mechanism of action of pyrazinamide is poorly understood because of its unusual properties. Here we show that pyrazinoic acid, the active moiety of pyrazinamide, disrupted membrane energetics and inhibited membrane transport function in Mycobacterium tuberculosis. The preferential activity of pyrazinamide against old non-replicating bacilli correlated with their low membrane potential and the disruption of membrane potential by pyrazinoic acid and acid pH. Inhibitors of membrane energetics increased the antituberculous activity of pyrazinamide. These findings shed new light on the mode of action of pyrazinamide and may help in the design of new drugs that shorten therapy.


Assuntos
Antituberculosos/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Pirazinamida/análogos & derivados , Pirazinamida/farmacologia , Azidas/farmacologia , Dicicloexilcarbodi-Imida/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Rotenona/farmacologia
11.
Microbiology (Reading) ; 143 ( Pt 10): 3367-3373, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9353938

RESUMO

The antituberculosis drug pyrazinamide (PZA) needs to be converted into pyrazinoic acid (POA) by the bacterial pyrazinamidase (PZase) in order to show bactericidal activity against Mycobacterium tuberculosis. M. avium is naturally resistant to PZA. To investigate whether this natural resistance to PZA is due to inability of the M. avium PZase to convert PZA to bactericidal POA, the M. avium PZase gene (pncA) was cloned by using the M. tuberculosis pncA gene as a probe. Sequence analysis showed that the M. avium pncA gene is 561 bp long, encoding a protein with a predicted size of about 19.8 kDa; but Western blotting showed that the M. avium PZase migrated as a 24 kDa band when expressed in M. bovis BCG and Escherichia coli. Sequence comparison revealed that M. avium PZase has 67.7% and 32.8% amino acid identity with the corresponding enzymes from M. tuberculosis and E. coli, respectively. Southern blot analysis with the M. avium pncA gene as a probe showed that M. terrae, M. gastri, M. marinum, M. fortuitum, M. xenopi, M. gordonae, M. szulgai, M. celatum and M. kansasii have close pncA homologues, whereas M. chelonae and M. smegmatis did not give significant hybridization signals. Transformation with the M. avium pncA gene conferred PZA susceptibility to PZA-resistant M. tuberculosis complex organisms, indicating that the nonsusceptibility of M. avium to PZA is not due to an ineffective PZase enzyme, but appears to be related to other factors such as transport of POA.


Assuntos
Amidoidrolases/genética , Genes Bacterianos , Complexo Mycobacterium avium/enzimologia , Complexo Mycobacterium avium/genética , Amidoidrolases/metabolismo , Sequência de Aminoácidos , Antituberculosos/metabolismo , Antituberculosos/farmacologia , Sequência de Bases , Transporte Biológico Ativo/genética , Clonagem Molecular , Primers do DNA/genética , Resistência Microbiana a Medicamentos/genética , Escherichia coli/genética , Humanos , Dados de Sequência Molecular , Mycobacterium/enzimologia , Mycobacterium/genética , Complexo Mycobacterium avium/efeitos dos fármacos , Pirazinamida/análogos & derivados , Pirazinamida/metabolismo , Pirazinamida/farmacologia , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...