Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 154(5): 962-970, 2013 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-23993090

RESUMO

Cyclic dinucleotides (CDNs) have been previously recognized as important secondary signaling molecules in bacteria and, more recently, in mammalian cells. In the former case, they represent secondary messengers affecting numerous responses of the prokaryotic cell, whereas in the latter, they act as agonists of the innate immune response. Remarkable new discoveries have linked these two patterns of utilization of CDNs as secondary messengers and have revealed unexpected influences they likely had on shaping human genetic variation. This Review summarizes these recent insights and provides a perspective on future unanswered questions in this exciting field.


Assuntos
Imunidade Inata , Nucleotídeos Cíclicos/metabolismo , Animais , Bactérias/metabolismo , Citocinas/imunologia , Humanos , Proteínas de Membrana/metabolismo , Nucleotídeos Cíclicos/química
2.
Nature ; 567(7747): 194-199, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30787435

RESUMO

Cyclic dinucleotides (CDNs) have central roles in bacterial homeostasis and virulence by acting as nucleotide second messengers. Bacterial CDNs also elicit immune responses during infection when they are detected by pattern-recognition receptors in animal cells. Here we perform a systematic biochemical screen for bacterial signalling nucleotides and discover a large family of cGAS/DncV-like nucleotidyltransferases (CD-NTases) that use both purine and pyrimidine nucleotides to synthesize a diverse range of CDNs. A series of crystal structures establish CD-NTases as a structurally conserved family and reveal key contacts in the enzyme active-site lid that direct purine or pyrimidine selection. CD-NTase products are not restricted to CDNs and also include an unexpected class of cyclic trinucleotide compounds. Biochemical and cellular analyses of CD-NTase signalling nucleotides demonstrate that these cyclic di- and trinucleotides activate distinct host receptors and thus may modulate the interaction of both pathogens and commensal microbiota with their animal and plant hosts.


Assuntos
Proteínas de Bactérias/metabolismo , Nucleotídeos/biossíntese , Nucleotídeos/metabolismo , Nucleotidiltransferases/química , Nucleotidiltransferases/metabolismo , Animais , Cristalografia por Raios X , Fosfatos de Dinucleosídeos/biossíntese , Fosfatos de Dinucleosídeos/metabolismo , Células HEK293 , Humanos , Camundongos , Nucleotídeos/química , Nucleotidiltransferases/genética , Óperon/genética , Simbiose
3.
J Biol Chem ; 294(9): 3024-3036, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30593509

RESUMO

Upon host infection, Mycobacterium tuberculosis secretes the tuberculosis necrotizing toxin (TNT) into the cytosol of infected macrophages, leading to host cell death by necroptosis. TNT hydrolyzes NAD+ in the absence of any exogenous cofactor, thus classifying it as a ß-NAD+ glycohydrolase. However, TNT lacks sequence similarity with other NAD+ hydrolyzing enzymes and lacks the essential motifs involved in NAD+ binding and hydrolysis by these enzymes. In this study, we used NMR to examine the enzymatic activity of TNT and found that TNT hydrolyzes NADP+ as fast as NAD+ but does not cleave the corresponding reduced dinucleotides. This activity of TNT was not inhibited by ADP-ribose or nicotinamide, indicating low affinity of TNT for these reaction products. A selection assay for nontoxic TNT variants in Escherichia coli identified four of six residues in the predicted NAD+-binding pocket and four glycine residues that form a cradle directly below the NAD+-binding site, a conserved feature in the TNT protein family. Site-directed mutagenesis of residues near the predicted NAD+-binding site revealed that Phe727, Arg757, and Arg780 are essential for NAD+ hydrolysis by TNT. These results identify the NAD+-binding site of TNT. Our findings also show that TNT is an NAD+ glycohydrolase with properties distinct from those of other bacterial glycohydrolases. Because many of these residues are conserved within the TNT family, our findings provide insights into understanding the function of the >300 TNT homologs.


Assuntos
Toxinas Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , NAD+ Nucleosidase/metabolismo , Sequência de Aminoácidos , Toxinas Bacterianas/química , Hidrólise , Espaço Intracelular/microbiologia , Modelos Moleculares , Mycobacterium tuberculosis/fisiologia , NAD/metabolismo , NADP/metabolismo , Conformação Proteica , Domínios Proteicos
4.
Proc Natl Acad Sci U S A ; 113(11): 3042-7, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929342

RESUMO

The bacterial type 6 secretion system (T6SS) is a dynamic apparatus that translocates proteins between cells by a mechanism analogous to phage tail contraction. T6SS sheaths are cytoplasmic tubular structures composed of stable VipA-VipB (named for ClpV-interacting protein A and B) heterodimers. Here, the structure of the VipA/B sheath was exploited to generate immunogenic multivalent particles for vaccine delivery. Sheaths composed of VipB and VipA fused to an antigen of interest were purified from Vibrio cholerae or Escherichia coli and used for immunization. Sheaths displaying heterologous antigens generated better immune responses against the antigen and different IgG subclasses compared with soluble antigen alone. Moreover, antigen-specific antibodies raised against sheaths presenting Neisseria meningitidis factor H binding protein (fHbp) antigen were functional in a serum bactericidal assay. Our results demonstrate that multivalent nanoparticles based on the T6SS sheath represent a versatile scaffold for vaccine applications.


Assuntos
Antígenos/administração & dosagem , Nanopartículas/química , Sistemas de Secreção Tipo VI/ultraestrutura , Vacinas/administração & dosagem , Acinetobacter/química , Acinetobacter/genética , Aminoaciltransferases/metabolismo , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Antibacterianos/imunologia , Antígenos/imunologia , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/química , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Dimerização , Sistemas de Liberação de Medicamentos , Escherichia coli/química , Feminino , Genes Reporter , Imunoglobulina G/biossíntese , Imunoglobulina G/genética , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Sistemas de Secreção Tipo VI/química , Vacinação , Vacinas/imunologia , Vibrio cholerae/química
5.
Mol Biol Evol ; 33(4): 898-914, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26613786

RESUMO

In the context of the great concern about the impact of human activities on the environment, we studied 403 commensal Escherichia coli/Escherichia clade strains isolated from several animal and human populations that have variable contacts to one another. Multilocus sequence typing (MLST) showed a decrease of diversity 1) in strains isolated from animals that had an increasing contact with humans and 2) in all strains that had increased antimicrobial resistance. A specific B1 phylogroup clonal complex (CC87, Institut Pasteur schema nomenclature) of animal origin was identified and characterized as being responsible for the increased antimicrobial resistance prevalence observed in strains from the environments with a high human-mediated antimicrobial pressure. CC87 strains have a high capacity of acquiring and disseminating resistance genes with specific metabolic and genetic determinants as demonstrated by high-throughput sequencing and phenotyping. They are good mouse gut colonizers but are not virulent. Our data confirm the predominant role of human activities in the emergence of antimicrobial resistance in the environmental bacterial strains and unveil a particular E. coli clonal complex of animal origin capable of spreading antimicrobial resistance to other members of microbial communities.


Assuntos
Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli/genética , Variação Genética , Animais , Anti-Infecciosos/efeitos adversos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/genética , Genótipo , Humanos , Camundongos , Tipagem de Sequências Multilocus , Filogenia
6.
Proc Natl Acad Sci U S A ; 111(18): 6750-5, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753609

RESUMO

The ability to control the timing and mode of host cell death plays a pivotal role in microbial infections. Many bacteria use toxins to kill host cells and evade immune responses. Such toxins are unknown in Mycobacterium tuberculosis. Virulent M. tuberculosis strains induce necrotic cell death in macrophages by an obscure molecular mechanism. Here we show that the M. tuberculosis protein Rv3903c (channel protein with necrosis-inducing toxin, CpnT) consists of an N-terminal channel domain that is used for uptake of nutrients across the outer membrane and a secreted toxic C-terminal domain. Infection experiments revealed that CpnT is required for survival and cytotoxicity of M. tuberculosis in macrophages. Furthermore, we demonstrate that the C-terminal domain of CpnT causes necrotic cell death in eukaryotic cells. Thus, CpnT has a dual function in uptake of nutrients and induction of host cell death by M. tuberculosis.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Toxinas Bacterianas/metabolismo , Exotoxinas/metabolismo , Mycobacterium tuberculosis/metabolismo , Sequência de Aminoácidos , Animais , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Linhagem Celular , Exotoxinas/química , Exotoxinas/genética , Genes Bacterianos , Glicerol/metabolismo , Células HEK293 , Humanos , Células Jurkat , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Mycobacterium bovis/genética , Mycobacterium bovis/crescimento & desenvolvimento , Mycobacterium bovis/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Filogenia , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/toxicidade , Homologia de Sequência de Aminoácidos , Virulência/genética , Virulência/fisiologia
7.
Mol Microbiol ; 97(5): 881-97, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26036301

RESUMO

Sphingomyelinases secreted by pathogenic bacteria play important roles in host-pathogen interactions ranging from interfering with phagocytosis and oxidative burst to iron acquisition. This study shows that the Mtb protein Rv0888 possesses potent sphingomyelinase activity cleaving sphingomyelin, a major lipid in eukaryotic cells, into ceramide and phosphocholine, which are then utilized by Mtb as carbon, nitrogen and phosphorus sources, respectively. An Mtb rv0888 deletion mutant did not grow on sphingomyelin as a sole carbon source anymore and replicated poorly in macrophages indicating that Mtb utilizes sphingomyelin during infection. Rv0888 is an unusual membrane protein with a surface-exposed C-terminal sphingomyelinase domain and a putative N-terminal channel domain that mediated glucose and phosphocholine uptake across the outer membrane in an M. smegmatis porin mutant. Hence, we propose to name Rv0888 as SpmT (sphingomyelinase of Mycobacterium tuberculosis). Erythrocyte membranes contain up to 27% sphingomyelin. The finding that Rv0888 accounts for half of Mtb's hemolytic activity is consistent with its sphingomyelinase activity and the observation that Rv0888 levels are increased in the presence of erythrocytes and sphingomyelin by 5- and 100-fold, respectively. Thus, Rv0888 is a novel outer membrane protein that enables Mtb to utilize sphingomyelin as a source of several essential nutrients during intracellular growth.


Assuntos
Proteínas da Membrana Bacteriana Externa/genética , Macrófagos/microbiologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Esfingomielinas/metabolismo , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Proteínas da Membrana Bacteriana Externa/metabolismo , Células Cultivadas , Ceramidas/metabolismo , Membrana Eritrocítica/química , Hemólise , Interações Hospedeiro-Patógeno , Humanos , Hidrólise , Macrófagos/metabolismo , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidade , Fagocitose , Fosforilcolina/metabolismo , Porinas/genética , Deleção de Sequência
8.
Proc Natl Acad Sci U S A ; 110(51): 20747-52, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24248354

RESUMO

An important question regarding the biologic implications of antibiotic-resistant microbes is how resistance impacts the organism's overall fitness and virulence. Currently it is generally thought that antibiotic resistance carries a fitness cost and reduces virulence. For the human pathogen Pseudomonas aeruginosa, treatment with carbapenem antibiotics is a mainstay of therapy that can lead to the emergence of resistance, often through the loss of the carbapenem entry channel OprD. Transposon insertion-site sequencing was used to analyze the fitness of 300,000 mutants of P. aeruginosa strain PA14 in a mouse model for gut colonization and systemic dissemination after induction of neutropenia. Transposon insertions in the oprD gene led not only to carbapenem resistance but also to a dramatic increase in mucosal colonization and dissemination to the spleen. These findings were confirmed in vivo with different oprD mutants of PA14 as well as with related pairs of carbapenem-susceptible and -resistant clinical isolates. Compared with OprD(+) strains, those lacking OprD were more resistant to killing by acidic pH or normal human serum and had increased cytotoxicity against murine macrophages. RNA-sequencing analysis revealed that an oprD mutant showed dramatic changes in the transcription of genes that may contribute to the various phenotypic changes observed. The association between carbapenem resistance and enhanced survival of P. aeruginosa in infected murine hosts suggests that either drug resistance or host colonization can cause the emergence of more pathogenic, drug-resistant P. aeruginosa clones in a single genetic event.


Assuntos
Carbapenêmicos/farmacologia , Farmacorresistência Bacteriana/genética , Mutação , Porinas , Pseudomonas aeruginosa , Animais , Modelos Animais de Doenças , Farmacorresistência Bacteriana/efeitos dos fármacos , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Concentração de Íons de Hidrogênio , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Masculino , Camundongos , Porinas/biossíntese , Porinas/genética , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade
9.
Antimicrob Agents Chemother ; 59(4): 2328-36, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25645841

RESUMO

Mycobacterium tuberculosis, the causative agent of tuberculosis, is protected from toxic solutes by an effective outer membrane permeability barrier. Recently, we showed that the outer membrane channel protein CpnT is required for efficient nutrient uptake by M. tuberculosis and Mycobacterium bovis BCG. In this study, we found that the cpnT mutant of M. bovis BCG is more resistant than the wild type to a large number of drugs and antibiotics, including rifampin, ethambutol, clarithromycin, tetracycline, and ampicillin, by 8- to 32-fold. Furthermore, the cpnT mutant of M. bovis BCG was 100-fold more resistant to nitric oxide, a major bactericidal agent required to control M. tuberculosis infections in mice. Thus, CpnT constitutes the first outer membrane susceptibility factor in slow-growing mycobacteria. The dual functions of CpnT in uptake of nutrients and mediating susceptibility to toxic molecules are reflected in macrophage infection experiments: while loss of CpnT was detrimental for M. bovis BCG in macrophages that enable bacterial replication, presumably due to inadequate nutrient uptake, it conferred a survival advantage in macrophages that mount a strong bactericidal response. Importantly, the cpnT gene showed a significantly higher density of nonsynonymous mutations in drug-resistant clinical M. tuberculosis strains, indicating that CpnT is under selective pressure in human tuberculosis and/or during chemotherapy. Our results indicate that the CpnT channel constitutes an outer membrane gateway controlling the influx of nutrients and toxic molecules into slow-growing mycobacteria. This study revealed that reducing protein-mediated outer membrane permeability might constitute a new drug resistance mechanism in slow-growing mycobacteria.


Assuntos
Proteínas da Membrana Bacteriana Externa/efeitos dos fármacos , Proteínas da Membrana Bacteriana Externa/genética , Farmacorresistência Bacteriana/genética , Mycobacterium tuberculosis/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Toxinas Bacterianas , Linhagem Celular , Humanos , Interleucina-1beta/biossíntese , Macrófagos/microbiologia , Camundongos , Testes de Sensibilidade Microbiana , Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo II/biossíntese
10.
PLoS Pathog ; 9(1): e1003120, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23431276

RESUMO

Iron is an essential nutrient for most bacterial pathogens, but is restricted by the host immune system. Mycobacterium tuberculosis (Mtb) utilizes two classes of small molecules, mycobactins and carboxymycobactins, to capture iron from the human host. Here, we show that an Mtb mutant lacking the mmpS4 and mmpS5 genes did not grow under low iron conditions. A cytoplasmic iron reporter indicated that the double mutant experienced iron starvation even under high-iron conditions. Loss of mmpS4 and mmpS5 did not change uptake of carboxymycobactin by Mtb. Thin layer chromatography showed that the ΔmmpS4/S5 mutant was strongly impaired in biosynthesis and secretion of siderophores. Pull-down experiments with purified proteins demonstrated that MmpS4 binds to a periplasmic loop of the associated transporter protein MmpL4. This interaction was corroborated by genetic experiments. While MmpS5 interacted only with MmpL5, MmpS4 interacted with both MmpL4 and MmpL5. These results identified MmpS4/MmpL4 and MmpS5/MmpL5 as siderophore export systems in Mtb and revealed that the MmpL proteins transport small molecules other than lipids. MmpS4 and MmpS5 resemble periplasmic adapter proteins of tripartite efflux pumps of Gram-negative bacteria, however, they are not only required for export but also for efficient siderophore synthesis. Membrane association of MbtG suggests a link between siderophore synthesis and transport. The structure of the soluble domain of MmpS4 (residues 52-140) was solved by NMR and indicates that mycobacterial MmpS proteins constitute a novel class of transport accessory proteins. The bacterial burden of the mmpS4/S5 deletion mutant in mouse lungs was lower by 10,000-fold and none of the infected mice died within 180 days compared to wild-type Mtb. This is the strongest attenuation observed so far for Mtb mutants lacking genes involved in iron utilization. In conclusion, this study identified the first components of novel siderophore export systems which are essential for virulence of Mtb.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Mycobacterium tuberculosis/patogenicidade , Oxazóis/metabolismo , Receptores de Superfície Celular/metabolismo , Sideróforos/metabolismo , Tuberculose/microbiologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Cromatografia em Camada Fina , Feminino , Interações Hospedeiro-Patógeno , Ferro/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Estrutura Terciária de Proteína , Deleção de Sequência , Baço/microbiologia , Baço/patologia , Taxa de Sobrevida , Tuberculose/mortalidade , Tuberculose/patologia , Virulência/genética
11.
EBioMedicine ; 88: 104439, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36709579

RESUMO

BACKGROUND: Worldwide, Escherichia coli is the leading cause of neonatal Gram-negative bacterial meningitis, but full understanding of the pathogenesis of this disease is not yet achieved. Moreover, to date, no vaccine is available against bacterial neonatal meningitis. METHODS: Here, we used Transposon Sequencing of saturated banks of mutants (TnSeq) to evaluate E. coli K1 genetic fitness in murine neonatal meningitis. We identified E. coli K1 genes encoding for factors important for systemic dissemination and brain infection, and focused on products with a likely outer-membrane or extra-cellular localization, as these are potential vaccine candidates. We used in vitro and in vivo models to study the efficacy of active and passive immunization. RESULTS: We selected for further study the conserved surface polysaccharide Poly-ß-(1-6)-N-Acetyl Glucosamine (PNAG), as a strong candidate for vaccine development. We found that PNAG was a virulence factor in our animal model. We showed that both passive and active immunization successfully prevented and/or treated meningitis caused by E. coli K1 in neonatal mice. We found an excellent opsonophagocytic killing activity of the antibodies to PNAG and in vitro these antibodies were also able to decrease binding, invasion and crossing of E. coli K1 through two blood brain barrier cell lines. Finally, to reinforce the potential of PNAG as a vaccine candidate in bacterial neonatal meningitis, we demonstrated that Group B Streptococcus, the main cause of neonatal meningitis in developed countries, also produced PNAG and that antibodies to PNAG could protect in vitro and in vivo against this major neonatal pathogen. INTERPRETATION: Altogether, these results indicate the utility of a high-throughput DNA sequencing method to identify potential immunotherapy targets for a pathogen, including in this study a potential broad-spectrum target for prevention of neonatal bacterial infections. FUNDINGS: ANR Seq-N-Vaq, Charles Hood Foundation, Hearst Foundation, and Groupe Pasteur Mutualité.


Assuntos
Escherichia coli , Meningites Bacterianas , Animais , Camundongos , Escherichia coli/genética , Anticorpos Antibacterianos , Bactérias/genética , Imunoterapia , Sequenciamento de Nucleotídeos em Larga Escala
12.
Nat Commun ; 12(1): 6592, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34782620

RESUMO

The tuberculosis necrotizing toxin (TNT) is the major cytotoxicity factor of Mycobacterium tuberculosis (Mtb) in macrophages. TNT is the C-terminal domain of the outer membrane protein CpnT and gains access to the cytosol to kill macrophages infected with Mtb. However, molecular mechanisms of TNT secretion and trafficking are largely unknown. A comprehensive analysis of the five type VII secretion systems of Mtb revealed that the ESX-4 system is required for export of CpnT and surface accessibility of TNT. Furthermore, the ESX-2 and ESX-4 systems are required for permeabilization of the phagosomal membrane in addition to the ESX-1 system. Thus, these three ESX systems need to act in concert to enable trafficking of TNT into the cytosol of Mtb-infected macrophages. These discoveries establish new molecular roles for the two previously uncharacterized type VII secretion systems ESX-2 and ESX-4 and reveal an intricate link between toxin secretion and phagosomal permeabilization by Mtb.


Assuntos
Toxinas Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , Toxinas Biológicas/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa , Proteínas de Bactérias , Morte Celular , Macrófagos/metabolismo , Fagossomos/metabolismo , Sistemas de Secreção Tipo VII
13.
Vaccines (Basel) ; 9(11)2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34835271

RESUMO

Emerging evidence demonstrates a connection between microbiome composition and suboptimal response to vaccines (vaccine hyporesponse). Harnessing the interaction between microbes and the immune system could provide novel therapeutic strategies for improving vaccine response. Currently we do not fully understand the mechanisms and dynamics by which the microbiome influences vaccine response. Using both mouse and non-human primate models, we report that short-term oral treatment with a single antibiotic (vancomycin) results in the disruption of the gut microbiome and this correlates with a decrease in systemic levels of antigen-specific IgG upon subsequent parenteral vaccination. We further show that recovery of microbial diversity before vaccination prevents antibiotic-induced vaccine hyporesponse, and that the antigen specific IgG response correlates with the recovery of microbiome diversity. RNA sequencing analysis of small intestine, spleen, whole blood, and secondary lymphoid organs from antibiotic treated mice revealed a dramatic impact on the immune system, and a muted inflammatory signature is correlated with loss of bacteria from Lachnospiraceae, Ruminococcaceae, and Clostridiaceae. These results suggest that microbially modulated immune pathways may be leveraged to promote vaccine response and will inform future vaccine design and development strategies.

14.
Antimicrob Agents Chemother ; 52(9): 3127-34, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18559650

RESUMO

The outer membrane of mycobacteria presents an effective permeability barrier for many antibiotics. Transport pathways across this membrane are unknown for most drugs. Here, we examined which antibiotics utilize the porin pathway across the outer membrane of the model organism Mycobacterium smegmatis. Deletion of the porins MspA and MspC drastically increased the resistance of M. smegmatis ML10 to beta-lactam antibiotics, while its beta-lactamase activity remained unchanged. These results are consistent with the ninefold-reduced outer membrane permeability of the M. smegmatis porin mutants for cephaloridine and strongly indicate that beta-lactam antibiotics rely on the porin pathway. The porin mutant ML10 accumulated less chloramphenicol and norfloxacin and was less susceptible to these antibiotics than wild-type M. smegmatis. These results demonstrated that small and hydrophilic antibiotics use the Msp porins for entering the cell. In contrast to norfloxacin, the hydrophobic moxifloxacin was 32-fold more effective in inhibiting the growth of M. smegmatis, presumably because it was able to diffuse through the lipid membrane. Structural models indicated that erythromycin, kanamycin, and vancomycin are too large to move through the MspA channel. This study presents the first experimental evidence that hydrophilic fluoroquinolones and chloramphenicol diffuse through porins in mycobacteria. Thus, mutations resulting in less efficient porins or lower porin expression levels are likely to represent a mechanism for the opportunistic pathogens M. avium, M. chelonae, and M. fortuitum, which have Msp-like porins, to acquire resistance to fluoroquinolones.


Assuntos
Antibacterianos/metabolismo , Cloranfenicol/metabolismo , Fluoroquinolonas/metabolismo , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium smegmatis/metabolismo , Porinas/metabolismo , Antibacterianos/farmacologia , Transporte Biológico , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cloranfenicol/farmacologia , Farmacorresistência Bacteriana , Fluoroquinolonas/farmacologia , Testes de Sensibilidade Microbiana/métodos , Mutação , Mycobacterium smegmatis/enzimologia , Porinas/genética , beta-Lactamas/farmacologia
15.
Antimicrob Agents Chemother ; 52(7): 2503-11, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18458127

RESUMO

The impermeability of the outer membrane in combination with drug efflux are major determinants of the natural drug resistance of mycobacteria. beta-Lactams are the most widely used antibiotics for treatment of bacterial infections. However, it is unknown how beta-lactams enter Mycobacterium tuberculosis and whether efflux pumps exist that can export these drugs out of the cell. To identify the molecular mechanisms of M. tuberculosis resistance to beta-lactams, a library of 7,500 transposon mutants was generated in the model organism Mycobacterium bovis BCG. Thirty-three unique insertion sites were determined that conferred medium or high-level (> or =2,000 microg/ml) resistance to ampicillin. Three mutants in sulfolipid synthesis or transport were highly resistant to ampicillin, indicating an indirect effect of the lipid composition on the outer membrane permeability of M. bovis BCG to ampicillin. Mutants with insertions in genes encoding surface molecules such as PPE proteins or lipoarabinomannan were also completely resistant to ampicillin, thus suggesting a lack of transport across the outer membrane. Insertion of the transposon in front of bcg0231 increased transcription of the gene and concomitantly the resistance of M. bovis BCG to ampicillin, streptomycin, and chloramphenicol by 32- to 64-fold. Resistance to vancomycin and tetracycline was increased four- to eightfold. Bcg0231 and Rv0194 are almost identical ATP-binding cassette transporters. Expression of rv0194 significantly reduced accumulation of ethidium bromide and conferred multidrug resistance to Mycobacterium smegmatis. Both effects were abrogated in the presence of the efflux pump inhibitor reserpine. These results demonstrate that Rv0194 is a novel multidrug efflux pump of M. tuberculosis.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Mycobacterium tuberculosis/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Resistência a Ampicilina/genética , Proteínas de Bactérias/genética , Sequência de Bases , DNA Bacteriano/genética , Farmacorresistência Bacteriana Múltipla/genética , Etídio/metabolismo , Genes Bacterianos , Humanos , Proteínas de Membrana Transportadoras/genética , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese Insercional , Mycobacterium bovis/efeitos dos fármacos , Mycobacterium bovis/genética , Mycobacterium bovis/metabolismo , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/metabolismo , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , Transcrição Gênica , Tuberculose Resistente a Múltiplos Medicamentos/tratamento farmacológico , Tuberculose Resistente a Múltiplos Medicamentos/microbiologia , Resistência beta-Lactâmica/genética , beta-Lactamases/genética , beta-Lactamases/metabolismo , beta-Lactamas/farmacologia
16.
Sci Transl Med ; 7(297): 297ra114, 2015 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-26203082

RESUMO

Advances in high-throughput DNA sequencing allow for a comprehensive analysis of bacterial genes that contribute to virulence in a specific infectious setting. Such information can yield new insights that affect decisions on how to best manage major public health issues such as the threat posed by increasing antimicrobial drug resistance. Much of the focus has been on the consequences of the selective advantage conferred on drug-resistant strains during antibiotic therapy. It is thought that the genetic and phenotypic changes that confer resistance also result in concomitant reductions in in vivo fitness, virulence, and transmission. However, experimental validation of this accepted paradigm is modest. Using a saturated transposon library of Pseudomonas aeruginosa, we identified genes across many functional categories and operons that contributed to maximal in vivo fitness during lung infections in animal models. Genes that bestowed both intrinsic and acquired antibiotic resistance provided a positive in vivo fitness advantage to P. aeruginosa during infection. We confirmed these findings in the pathogenic bacteria Acinetobacter baumannii and Vibrio cholerae using murine and rabbit infection models, respectively. Our results show that efforts to confront the worldwide increase in antibiotic resistance might be exacerbated by fitness advantages that enhance virulence in drug-resistant microbes.


Assuntos
Antibacterianos/uso terapêutico , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/microbiologia , Efeitos Psicossociais da Doença , Acinetobacter baumannii/efeitos dos fármacos , Acinetobacter baumannii/genética , Acinetobacter baumannii/fisiologia , Animais , Antibacterianos/farmacologia , Contagem de Colônia Microbiana , Elementos de DNA Transponíveis/genética , Modelos Animais de Doenças , Resistência Microbiana a Medicamentos/genética , Trato Gastrointestinal/patologia , Genes Bacterianos , Pulmão/microbiologia , Camundongos , Testes de Sensibilidade Microbiana , Mutagênese Insercional/genética , Mutação/genética , Óperon/genética , Pneumonia/tratamento farmacológico , Pneumonia/microbiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/fisiologia , Coelhos , Análise de Sequência de DNA , Vibrio cholerae/efeitos dos fármacos , Vibrio cholerae/genética , Vibrio cholerae/fisiologia
17.
Cell Host Microbe ; 16(5): 581-91, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25450857

RESUMO

The cellular surveillance-activated detoxification and defenses (cSADD) theory postulates the presence of host surveillance mechanisms that monitor the integrity of common cellular processes and components targeted by pathogen effectors. Being organelles essential for multiple cellular processes, including innate immune responses, mitochondria represent an attractive target for pathogens. We describe a Vibrio cholerae Type 3 secretion system effector VopE that localizes to mitochondria during infection and acts as a specific GTPase-activating protein to interfere with the function of mitochondrial Rho GTPases Miro1 and Miro2. Miro GTPases modulate mitochondrial dynamics and interfering with this functionality effectively blocks innate immune responses that presumably require mitochondria as signaling platforms. Our data indicate that interference with mitochondrial dynamics may be an unappreciated strategy that pathogens use to block host innate immune responses that would otherwise control these bacterial infections. VopE might represent a bacterial effector that targets the cSADD surveillance response.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos , Imunidade Inata , Dinâmica Mitocondrial/fisiologia , Vibrio cholerae/imunologia , Proteínas rho de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cricetulus , Meios de Cultura , Proteínas Ativadoras de GTPase/metabolismo , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais
18.
Trends Microbiol ; 18(3): 109-16, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20060722

RESUMO

The cell wall is a major virulence factor of Mycobacterium tuberculosis and contributes to its intrinsic drug resistance. Recently, cryo-electron microscopy showed that mycobacterial cell wall lipids form an unusual outer membrane. Identification of the components of the uptake and secretion machinery across this membrane will be crucial for understanding the physiology and pathogenicity of M. tuberculosis and for the development of better anti-tuberculosis drugs. Although the genome of M. tuberculosis appears to encode over 100 putative outer membrane proteins, only a few have been identified and characterized. Here, we summarize the current knowledge on the structure of the mycobacterial outer membrane and its known proteins. Through comparison to transport processes in Gram-negative bacteria, we highlight several hypothetical outer membrane proteins of M. tuberculosis that await discovery.


Assuntos
Proteínas da Membrana Bacteriana Externa/análise , Parede Celular/química , Proteínas de Membrana Transportadoras/análise , Mycobacterium tuberculosis/química , Proteínas da Membrana Bacteriana Externa/genética , Parede Celular/genética , Parede Celular/ultraestrutura , Humanos , Proteínas de Membrana Transportadoras/genética , Modelos Biológicos , Modelos Químicos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/ultraestrutura
19.
Adv Microb Physiol ; 55: 81-182, 318-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19573696

RESUMO

Mycobacterium tuberculosis is a prototrophic, metabolically flexible bacterium that has achieved a spread in the human population that is unmatched by any other bacterial pathogen. The success of M. tuberculosis as a pathogen can be attributed to its extraordinary stealth and capacity to adapt to environmental changes throughout the course of infection. These changes include: nutrient deprivation, hypoxia, various exogenous stress conditions and, in the case of the pathogenic species, the intraphagosomal environment. Knowledge of the physiology of M. tuberculosis during this process has been limited by the slow growth of the bacterium in the laboratory and other technical problems such as cell aggregation. Advances in genomics and molecular methods to analyze the M. tuberculosis genome have revealed that adaptive changes are mediated by complex regulatory networks and signals, resulting in temporal gene expression coupled to metabolic and energetic changes. An important goal for bacterial physiologists will be to elucidate the physiology of M. tuberculosis during the transition between the diverse conditions encountered by M. tuberculosis. This review covers the growth of the mycobacterial cell and how environmental stimuli are sensed by this bacterium. Adaptation to different environments is described from the viewpoint of nutrient acquisition, energy generation, and regulation. To gain quantitative understanding of mycobacterial physiology will require a systems biology approach and recent efforts in this area are discussed.


Assuntos
Fenômenos Fisiológicos Bacterianos , Mycobacterium tuberculosis/fisiologia , Estresse Fisiológico , Metabolismo Energético , Regulação Bacteriana da Expressão Gênica , Humanos , Transdução de Sinais
20.
J Biol Chem ; 283(26): 17827-37, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18434314

RESUMO

Mycobacteria contain an outer membrane composed of mycolic acids and a large variety of other lipids. Its protective function is an essential virulence factor of Mycobacterium tuberculosis. Only OmpA, which has numerous homologs in Gram-negative bacteria, is known to form channels in the outer membrane of M. tuberculosis so far. Rv1698 was predicted to be an outer membrane protein of unknown function. Expression of rv1698 restored the sensitivity to ampicillin and chloramphenicol of a Mycobacterium smegmatis mutant lacking the main porin MspA. Uptake experiments showed that Rv1698 partially complemented the permeability defect of the M. smegmatis porin mutant for glucose. These results indicated that Rv1698 provides an unspecific pore that can partially substitute for MspA. Lipid bilayer experiments demonstrated that purified Rv1698 is an integral membrane protein that indeed produces channels. The main single channel conductance is 4.5 +/- 0.3 nanosiemens in 1 M KCl. Zero current potential measurements revealed a weak preference for cations. Whole cell digestion of recombinant M. smegmatis with proteinase K showed that Rv1698 is surface-accessible. Taken together, these experiments demonstrated that Rv1698 is a channel protein that is likely involved in transport processes across the outer membrane of M. tuberculosis. Rv1698 has single homologs of unknown functions in Corynebacterineae and thus represents the first member of a new class of channel proteins specific for mycolic acid-containing outer membranes.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Membrana Celular/metabolismo , Mycobacterium tuberculosis/metabolismo , Antibacterianos/farmacologia , Proteínas da Membrana Bacteriana Externa/classificação , Endopeptidase K/química , Escherichia coli/metabolismo , Glucose/química , Glucose/metabolismo , Bicamadas Lipídicas/química , Modelos Biológicos , Mutação , Mycobacterium bovis/metabolismo , Mycobacterium smegmatis/metabolismo , Mutação Puntual , Porinas/química , Estrutura Secundária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA