Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Immunity ; 57(5): 1019-1036.e9, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38677292

RESUMO

Group 3 innate lymphoid cells (ILC3) are the major subset of gut-resident ILC with essential roles in infections and tissue repair, but how they adapt to the gut environment to maintain tissue residency is unclear. We report that Tox2 is critical for gut ILC3 maintenance and function. Gut ILC3 highly expressed Tox2, and depletion of Tox2 markedly decreased ILC3 in gut but not at central sites, resulting in defective control of Citrobacter rodentium infection. Single-cell transcriptional profiling revealed decreased expression of Hexokinase-2 in Tox2-deficient gut ILC3. Consistent with the requirement for hexokinases in glycolysis, Tox2-/- ILC3 displayed decreased ability to utilize glycolysis for protein translation. Ectopic expression of Hexokinase-2 rescued Tox2-/- gut ILC3 defects. Hypoxia and interleukin (IL)-17A each induced Tox2 expression in ILC3, suggesting a mechanism by which ILC3 adjusts to fluctuating environments by programming glycolytic metabolism. Our results reveal the requirement for Tox2 to support the metabolic adaptation of ILC3 within the gastrointestinal tract.


Assuntos
Citrobacter rodentium , Infecções por Enterobacteriaceae , Glicólise , Imunidade Inata , Linfócitos , Camundongos Knockout , Animais , Camundongos , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Transativadores/metabolismo , Transativadores/genética , Hexoquinase/metabolismo , Hexoquinase/genética , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/metabolismo , Interleucina-17/metabolismo , Adaptação Fisiológica/imunologia
3.
Nat Med ; 30(2): 560-572, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38291301

RESUMO

Nutrition has broad impacts on all physiological processes. However, how nutrition affects human immunity remains largely unknown. Here we explored the impact of a dietary intervention on both immunity and the microbiota by performing a post hoc analysis of a clinical trial in which each of the 20 participants sequentially consumed vegan or ketogenic diets for 2 weeks ( NCT03878108 ). Using a multiomics approach including multidimensional flow cytometry, transcriptomic, proteomic, metabolomic and metagenomic datasets, we assessed the impact of each diet, and dietary switch, on host immunity and the microbiota. Our data revealed that overall, a ketogenic diet was associated with a significant upregulation of pathways and enrichment in cells associated with the adaptive immune system. In contrast, a vegan diet had a significant impact on the innate immune system, including upregulation of pathways associated with antiviral immunity. Both diets significantly and differentially impacted the microbiome and host-associated amino acid metabolism, with a strong downregulation of most microbial pathways following ketogenic diet compared with baseline and vegan diet. Despite the diversity of participants, we also observed a tightly connected network between datasets driven by compounds associated with amino acids, lipids and the immune system. Collectively, this work demonstrates that in diverse participants 2 weeks of controlled dietary intervention is sufficient to significantly and divergently impact host immunity, which could have implications for precision nutritional interventions. ClinicalTrials.gov registration: NCT03878108 .


Assuntos
Dieta Cetogênica , Dieta Vegana , Humanos , Proteômica , Ensaios Clínicos como Assunto
4.
Proc Natl Acad Sci U S A ; 120(49): e2304905120, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38011570

RESUMO

Mild or transient dietary restriction (DR) improves many aspects of health and aging. Emerging evidence from us and others has demonstrated that DR also optimizes the development and quality of immune responses. However, the factors and mechanisms involved remain to be elucidated. Here, we propose that DR-induced optimization of immunological memory requires a complex cascade of events involving memory T cells, the intestinal microbiota, and myeloid cells. Our findings suggest that DR enhances the ability of memory T cells to recruit and activate myeloid cells in the context of a secondary infection. Concomitantly, DR promotes the expansion of commensal Bifidobacteria within the large intestine, which produce the short-chain fatty acid acetate. Acetate conditioning of the myeloid compartment during DR enhances the capacity of these cells to kill pathogens. Enhanced host protection during DR is compromised when Bifidobacteria expansion is prevented, indicating that microbiota configuration and function play an important role in determining immune responsiveness to this dietary intervention. Altogether, our study supports the idea that DR induces both memory T cells and the gut microbiota to produce distinct factors that converge on myeloid cells to promote optimal pathogen control. These findings suggest that nutritional cues can promote adaptation and co-operation between multiple immune cells and the gut microbiota, which synergize to optimize immunity and protect the collective metaorganism.


Assuntos
Microbioma Gastrointestinal , Microbiota , Ácidos Graxos Voláteis , Acetatos
5.
Cell Rep ; 42(1): 112020, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36848230

RESUMO

Variations in the composition of the intestinal bacterial microbiome correlate with acquisition of some sexually transmitted pathogens. To experimentally assess the contribution of intestinal dysbiosis to rectal lentiviral acquisition, we induce dysbiosis in rhesus macaques (RMs) with the antibiotic vancomycin prior to repeated low-dose intrarectal challenge with simian immunodeficiency virus (SIV) SIVmac239X. Vancomycin administration reduces T helper 17 (TH17) and TH22 frequencies, increases expression of host bacterial sensors and antibacterial peptides, and increases numbers of transmitted-founder (T/F) variants detected upon SIV acquisition. We observe that SIV acquisition does not correlate with measures of dysbiosis but rather associates with perturbations in the host antimicrobial program. These findings establish a functional association between the intestinal microbiome and susceptibility to lentiviral acquisition across the rectal epithelial barrier.


Assuntos
Disbiose , Vírus da Imunodeficiência Símia , Animais , Macaca mulatta , Vancomicina , Antibacterianos
6.
Proc Natl Acad Sci U S A ; 120(4): e2214484120, 2023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36652484

RESUMO

The microbiota performs multiple functions vital to host fitness, including defense against pathogens and adaptation to dietary changes. Yet, how environmental challenges shape microbiota resilience to nutrient fluctuation remains largely unexplored. Here, we show that transient gut infection can optimize host metabolism toward the usage of carbohydrates. Following acute infection and clearance of the pathogen, mice gained more weight as a result of white adipose tissue expansion. Concomitantly, previously infected mice exhibited enhanced carbohydrate (glucose) disposal and insulin sensitivity. This metabolic remodeling depended on alterations to the gut microbiota, with infection-elicited Betaproteobacteria being sufficient to enhance host carbohydrate metabolism. Further, infection-induced metabolic alteration protected mice against stunting in the context of limited nutrient availability. Together, these results propose that alterations to the microbiota imposed by acute infection may enhance host fitness and survival in the face of nutrient restriction, a phenomenon that may be adaptive in settings where both infection burden and food precarity are prevalent.


Assuntos
Resistência à Insulina , Microbiota , Animais , Camundongos , Adaptação ao Hospedeiro , Obesidade/metabolismo , Nutrientes
8.
Microbiome ; 10(1): 43, 2022 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-35272717

RESUMO

BACKGROUND: The human intestinal microbiome is a complex community that contributes to host health and disease. In addition to normal microbiota, pathogens like carbapenem-resistant Enterobacteriaceae may be asymptomatically present. When these bacteria are present at very low levels, they are often undetectable in hospital surveillance cultures, known as occult or subclinical colonization. Through the receipt of antibiotics, these subclinical pathogens can increase to sufficiently high levels to become detectable, in a process called outgrowth. However, little is known about the interaction between gut microbiota and Enterobacteriaceae during occult colonization and outgrowth. RESULTS: We developed a clinically relevant mouse model for studying occult colonization. Conventional wild-type mice without antibiotic pre-treatment were exposed to Klebsiella pneumoniae but rapidly tested negative for colonization. This occult colonization was found to perturb the microbiome as detected by both 16S rRNA amplicon and shotgun metagenomic sequencing. Outgrowth of occult K. pneumoniae was induced either by a four-antibiotic cocktail or by individual receipt of ampicillin, vancomycin, or azithromycin, which all reduced overall microbial diversity. Notably, vancomycin was shown to trigger K. pneumoniae outgrowth in only a subset of exposed animals (outgrowth-susceptible). To identify factors that underlie outgrowth susceptibility, we analyzed microbiome-encoded gene functions and were able to classify outgrowth-susceptible microbiomes using pathways associated with mRNA stability. Lastly, an evolutionary approach illuminated the importance of xylose metabolism in K. pneumoniae colonization, supporting xylose abundance as a second susceptibility indicator. We showed that our model is generalizable to other pathogens, including carbapenem-resistant Escherichia coli and Enterobacter cloacae. CONCLUSIONS: Our modeling of occult colonization and outgrowth could help the development of strategies to mitigate the risk of subsequent infection and transmission in medical facilities and the wider community. This study suggests that microbiota mRNA and small-molecule metabolites may be used to predict outgrowth-susceptibility. Video Abstract.


Assuntos
Enterobacteriáceas Resistentes a Carbapenêmicos , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Enterobacteriáceas Resistentes a Carbapenêmicos/genética , Intestinos/microbiologia , Klebsiella pneumoniae/genética , Camundongos , RNA Ribossômico 16S/genética
9.
Science ; 373(6558)2021 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-34446580

RESUMO

The immune system has evolved in the face of microbial exposure. How maternal infection experienced at distinct developmental stages shapes the offspring immune system remains poorly understood. Here, we show that during pregnancy, maternally restricted infection can have permanent and tissue-specific impacts on offspring immunity. Mechanistically, maternal interleukin-6 produced in response to infection can directly impose epigenetic changes on fetal intestinal epithelial stem cells, leading to long-lasting impacts on intestinal immune homeostasis. As a result, offspring of previously infected dams develop enhanced protective immunity to gut infection and increased inflammation in the context of colitis. Thus, maternal infection can be coopted by the fetus to promote long-term, tissue-specific fitness, a phenomenon that may come at the cost of predisposition to inflammatory disorders.


Assuntos
Colite/imunologia , Imunidade , Interleucina-6/imunologia , Intestinos/imunologia , Complicações Infecciosas na Gravidez/imunologia , Células Th17/imunologia , Infecções por Yersinia pseudotuberculosis/imunologia , Animais , Candidíase/imunologia , Cromatina/metabolismo , Epigênese Genética , Epigenoma , Feminino , Desenvolvimento Fetal , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Interleucina-6/sangue , Interleucina-6/farmacologia , Mucosa Intestinal/citologia , Mucosa Intestinal/embriologia , Mucosa Intestinal/imunologia , Intestinos/embriologia , Intestinos/microbiologia , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Salmonelose Animal/imunologia , Células-Tronco/imunologia , Células-Tronco/fisiologia , Subpopulações de Linfócitos T/imunologia
10.
Cell ; 184(3): 615-627.e17, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33453153

RESUMO

The microbiota shields the host against infections in a process known as colonization resistance. How infections themselves shape this fundamental process remains largely unknown. Here, we show that gut microbiota from previously infected hosts display enhanced resistance to infection. This long-term functional remodeling is associated with altered bile acid metabolism leading to the expansion of taxa that utilize the sulfonic acid taurine. Notably, supplying exogenous taurine alone is sufficient to induce this alteration in microbiota function and enhance resistance. Mechanistically, taurine potentiates the microbiota's production of sulfide, an inhibitor of cellular respiration, which is key to host invasion by numerous pathogens. As such, pharmaceutical sequestration of sulfide perturbs the microbiota's composition and promotes pathogen invasion. Together, this work reveals a process by which the host, triggered by infection, can deploy taurine as a nutrient to nourish and train the microbiota, promoting its resistance to subsequent infection.


Assuntos
Microbioma Gastrointestinal , Interações Hospedeiro-Patógeno , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Contagem de Colônia Microbiana , Microbioma Gastrointestinal/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Imunidade , Camundongos Endogâmicos C57BL , Sulfetos/metabolismo , Taurina/farmacologia
11.
Proc Natl Acad Sci U S A ; 116(39): 19685-19694, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31427504

RESUMO

Recent evidence suggests that the genes an organism needs to survive in an environment drastically differ when alone or in a community. However, it is not known if there are universal functions that enable microbes to persist in a community and if there are functions specific to interactions between microbes native to the same (sympatric) or different (allopatric) environments. Here, we ask how the essential functions of the oral pathogen Aggregatibacter actinomycetemcomitans change during pairwise coinfection in a murine abscess with each of 15 microbes commonly found in the oral cavity and 10 microbes that are not. A. actinomycetemcomitans was more abundant when coinfected with allopatric than with sympatric microbes, and this increased fitness correlated with expanded metabolic capacity of the coinfecting microbes. Using transposon sequencing, we discovered that 33% of the A. actinomycetemcomitans genome is required for coinfection fitness. Fifty-nine "core" genes were required across all coinfections and included genes necessary for aerobic respiration. The core genes were also all required in monoinfection, indicating the essentiality of these genes cannot be alleviated by a coinfecting microbe. Furthermore, coinfection with some microbes, predominately sympatric species, induced the requirement for over 100 new community-dependent essential genes. In contrast, in other coinfections, predominately with nonoral species, A. actinomycetemcomitans required 50 fewer genes than in monoinfection, demonstrating that some allopatric microbes can drastically alleviate gene essentialities. These results expand our understanding of how diverse microbes alter growth and gene essentiality within polymicrobial infections.


Assuntos
Aggregatibacter actinomycetemcomitans/genética , Genes Essenciais/genética , Microbiota/genética , Simpatria/genética , Aggregatibacter/genética , Aggregatibacter actinomycetemcomitans/patogenicidade , Animais , Coinfecção , Aptidão Genética/genética , Camundongos , RNA Ribossômico 16S/genética
13.
Chembiochem ; 18(17): 1692-1695, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28628741

RESUMO

It is inherently difficult to quantitate nucleic acid analytes with most isothermal amplification assays. We developed loop-mediated isothermal amplification (LAMP) reactions in which competition between defined numbers of "false" and "true" amplicons leads to order of magnitude quantitation by a single endpoint determination. These thresholded LAMP reactions were successfully used to directly and quantitatively estimate the numbers of nucleic acids in complex biospecimens, including directly from cells and in sewage, with the values obtained closely correlating with qPCR quantitations. Thresholded LAMP reactions are amenable to endpoint readout by cell phone, unlike other methods that require continuous monitoring, and should therefore prove extremely useful in developing one-pot reactions for point-of-care diagnostics without needing sophisticated material or informatics infrastructure.


Assuntos
Técnicas de Amplificação de Ácido Nucleico , Ácidos Nucleicos/metabolismo , Animais , Proteínas de Bactérias/genética , Infecções por Fusobacterium/diagnóstico , Fusobacterium nucleatum/genética , Camundongos , Neuropilina-2/genética , Neuropilina-2/metabolismo , Ácidos Nucleicos/genética , Sistemas Automatizados de Assistência Junto ao Leito , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
Appl Environ Microbiol ; 83(14)2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28476775

RESUMO

Periodontitis is a microbial infection that destroys the structures that support the teeth. Although it is typically a chronic condition, rapidly progressing, aggressive forms are associated with the oral pathogen Aggregatibacter actinomycetemcomitans One of this bacterium's key virulence traits is its ability to attach to surfaces and form robust biofilms that resist killing by the host and antibiotics. Though much has been learned about A. actinomycetemcomitans since its initial discovery, we lack insight into a fundamental aspect of its basic biology, as we do not know the full set of genes that it requires for viability (the essential genome). Furthermore, research on A. actinomycetemcomitans is hampered by the field's lack of a mutant collection. To address these gaps, we used rapid transposon mutant sequencing (Tn-seq) to define the essential genomes of two strains of A. actinomycetemcomitans, revealing a core set of 319 genes. We then generated an arrayed mutant library comprising >1,500 unique insertions and used a sequencing-based approach to define each mutant's position (well and plate) in the library. To demonstrate its utility, we screened the library for mutants with weakened resistance to subinhibitory erythromycin, revealing the multidrug efflux pump AcrAB as a critical resistance factor. During the screen, we discovered that erythromycin induces A. actinomycetemcomitans to form biofilms. We therefore devised a novel Tn-seq-based screen to identify specific factors that mediate this phenotype and in follow-up experiments confirmed 4 mutants. Together, these studies present new insights and resources for investigating the basic biology and disease mechanisms of a human pathogen.IMPORTANCE Millions suffer from gum disease, which often is caused by Aggregatibacter actinomycetemcomitans, a bacterium that forms antibiotic-resistant biofilms. To fully understand any organism, we should be able to answer: what genes does it require for life? Here, we address this question for A. actinomycetemcomitans by determining the genes in its genome that cannot be mutated. As for the genes that can be mutated, we archived these mutants into a library, which we used to find genes that contribute to antibiotic resistance, leading us to discover that antibiotics cause A. actinomycetemcomitans to form biofilms. We then devised an approach to find genes that mediate this process and confirmed 4 genes. These results illuminate new fundamental traits of a human pathogen.


Assuntos
Aggregatibacter actinomycetemcomitans/genética , Genoma Bacteriano , Periodontite/microbiologia , Aggregatibacter actinomycetemcomitans/classificação , Aggregatibacter actinomycetemcomitans/isolamento & purificação , Biblioteca Gênica , Aptidão Genética , Genômica , Humanos , Boca/microbiologia , Mutagênese Insercional
15.
Nat Microbiol ; 2: 17079, 2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28555625

RESUMO

Identifying genes required by pathogens during infection is critical for antimicrobial development. Here, we use a Monte Carlo simulation-based method to analyse high-throughput transposon sequencing data to determine the role of infection site and co-infecting microorganisms on the in vivo 'essential' genome of Staphylococcus aureus. We discovered that co-infection of murine surgical wounds with Pseudomonas aeruginosa results in conversion of ∼25% of the in vivo S. aureus mono-culture essential genes to non-essential. Furthermore, 182 S. aureus genes are uniquely essential during co-infection. These 'community dependent essential' (CoDE) genes illustrate the importance of studying pathogen gene essentiality in polymicrobial communities.


Assuntos
Coinfecção/microbiologia , Genes Bacterianos , Genes Essenciais , Interações Microbianas , Pseudomonas aeruginosa/genética , Staphylococcus aureus/genética , Animais , Camundongos , Mutagênese Insercional , Pseudomonas aeruginosa/crescimento & desenvolvimento , Análise de Sequência de DNA , Staphylococcus aureus/crescimento & desenvolvimento , Infecção da Ferida Cirúrgica/microbiologia
16.
PLoS Pathog ; 12(12): e1006084, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27973608

RESUMO

Iron is an essential nutrient for bacterial pathogenesis, but in the host, iron is tightly sequestered, limiting its availability for bacterial growth. Although this is an important arm of host immunity, most studies examine how bacteria respond to iron restriction in laboratory rather than host settings, where the microbiome can potentially alter pathogen strategies for acquiring iron. One of the most important transcriptional regulators controlling bacterial iron homeostasis is Fur. Here we used a combination of RNA-seq and chromatin immunoprecipitation (ChIP)-seq to characterize the iron-restricted and Fur regulons of the biofilm-forming opportunistic pathogen Aggregatibacter actinomycetemcomitans. We discovered that iron restriction and Fur regulate 4% and 3.5% of the genome, respectively. While most genes in these regulons were related to iron uptake and metabolism, we found that Fur also directly regulates the biofilm-dispersing enzyme Dispersin B, allowing A. actinomycetemcomitans to escape from iron-scarce environments. We then leveraged these datasets to assess the availability of iron to A. actinomycetemcomitans in its primary infection sites, abscesses and the oral cavity. We found that A. actinomycetemcomitans is not restricted for iron in a murine abscess mono-infection, but becomes restricted for iron upon co-infection with the oral commensal Streptococcus gordonii. Furthermore, in the transition from health to disease in human gum infection, A. actinomycetemcomitans also becomes restricted for iron. These results suggest that host iron availability is heterogeneous and dependent on the infecting bacterial community.


Assuntos
Proteínas de Bactérias/metabolismo , Coinfecção/metabolismo , Ferro/metabolismo , Infecções por Pasteurellaceae/metabolismo , Periodontite/microbiologia , Proteínas Repressoras/metabolismo , Animais , Biofilmes/crescimento & desenvolvimento , Coinfecção/microbiologia , Modelos Animais de Doenças , Humanos , Imunoprecipitação , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Pasteurellaceae/microbiologia , Análise de Componente Principal , Infecções Estreptocócicas/microbiologia , Streptococcus gordonii
17.
mBio ; 7(3)2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27353758

RESUMO

UNLABELLED: Bacteria rarely inhabit infection sites alone, instead residing in diverse, multispecies communities. Despite this fact, bacterial pathogenesis studies primarily focus on monoculture infections, overlooking how community interactions influence the course of disease. In this study, we used global mutant fitness profiling (transposon sequencing [Tn-seq]) to determine the genetic requirements for the pathogenic bacterium Aggregatibacter actinomycetemcomitans to cause disease when coinfecting with the commensal bacterium Streptococcus gordonii Our results show that S. gordonii extensively alters A. actinomycetemcomitans requirements for virulence factors and biosynthetic pathways during infection. In addition, we discovered that the presence of S. gordonii enhances the bioavailability of oxygen during infection, allowing A. actinomycetemcomitans to shift from a primarily fermentative to a respiratory metabolism that enhances its growth yields and persistence. Mechanistically, respiratory metabolism enhances the fitness of A. actinomycetemcomitans in vivo by increasing ATP yields via central metabolism and creating a proton motive force. Our results reveal that, similar to cross-feeding, where one species provides another species with a nutrient, commensal bacteria can also provide electron acceptors that promote the respiratory growth and fitness of pathogens in vivo, an interaction that we term cross-respiration. IMPORTANCE: Commensal bacteria can enhance the virulence of pathogens in mixed-species infections. However, knowledge of the mechanisms underlying this clinically relevant phenomenon is lacking. To bridge this gap, we comprehensively determined the genes a pathogen needs to establish coinfection with a commensal. Our findings show that the metabolism of the pathogen is low-energy-yielding in monoinfection, but in coinfection, the commensal improves the fitness of the pathogen by increasing the bioavailability of oxygen, thereby shifting the pathogen toward a high-energy-yielding metabolism. Similar to cross-feeding, this interaction, which we term cross-respiration, illustrates that commensal bacteria can provide electron acceptors that enhance the virulence of pathogens during infection.


Assuntos
Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/patogenicidade , Coinfecção/microbiologia , Elementos de DNA Transponíveis/genética , Oxigênio/metabolismo , Streptococcus gordonii/metabolismo , Simbiose , Trifosfato de Adenosina/metabolismo , Aggregatibacter actinomycetemcomitans/crescimento & desenvolvimento , Aggregatibacter actinomycetemcomitans/metabolismo , Anaerobiose , Animais , Coinfecção/fisiopatologia , Perfilação da Expressão Gênica , Aptidão Genética , Sequenciamento de Nucleotídeos em Larga Escala , Redes e Vias Metabólicas/fisiologia , Camundongos , Infecções por Pasteurellaceae/microbiologia , Infecções por Pasteurellaceae/fisiopatologia , Força Próton-Motriz , Virulência
18.
Nat Rev Microbiol ; 14(2): 93-105, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26714431

RESUMO

Microbial communities are spatially organized in both the environment and the human body. Although patterns exhibited by these communities are described by microbial biogeography, this discipline has previously only considered large-scale, global patterns. By contrast, the fine-scale positioning of a pathogen within an infection site can greatly alter its virulence potential. In this Review, we highlight the importance of considering spatial positioning in the study of polymicrobial infections and discuss targeting biogeography as a therapeutic strategy.


Assuntos
Coinfecção/epidemiologia , Biofilmes , Coinfecção/microbiologia , Epitélio , Humanos , Filogeografia , Simbiose
19.
Proc Natl Acad Sci U S A ; 111(21): 7819-24, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24825893

RESUMO

The oral pathogen Aggregatibacter actinomycetemcomitans (Aa) resides in infection sites with many microbes, including commensal streptococci such as Streptococcus gordonii (Sg). During infection, Sg promotes the virulence of Aa by producing its preferred carbon source, l-lactate, a phenomenon referred to as cross-feeding. However, as with many streptococci, Sg also produces high levels of the antimicrobial hydrogen peroxide (H2O2), leading to the question of how Aa deals with this potent antimicrobial during coinfection. Here, we show that Aa possesses two complementary responses to H2O2: a detoxification or fight response mediated by catalase (KatA) and a dispersion or flight response mediated by Dispersin B (DspB), an enzyme that dissolves Aa biofilms. Using a murine abscess infection model, we show that both of these responses are required for Sg to promote Aa virulence. Although the role of KatA is to detoxify H2O2 during coinfection, 3D spatial analysis of mixed infections revealed that DspB is required for Aa to spatially organize itself at an optimal distance (>4 µm) from Sg, which we propose allows cross-feeding but reduces exposure to inhibitory levels of H2O2. In addition, these behaviors benefit not only Aa but also Sg, suggesting that fight and flight stimulate the fitness of the community. These results reveal that an antimicrobial produced by a human commensal bacterium enhances the virulence of a pathogenic bacterium by modulating its spatial location in the infection site.


Assuntos
Aggregatibacter actinomycetemcomitans/patogenicidade , Proteínas de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Catalase/metabolismo , Coinfecção/fisiopatologia , Glicosídeo Hidrolases/metabolismo , Streptococcus gordonii/metabolismo , Aggregatibacter actinomycetemcomitans/metabolismo , Animais , Coinfecção/microbiologia , Peróxido de Hidrogênio/metabolismo , Ácido Láctico/metabolismo , Camundongos , Análise em Microsséries , Virulência
20.
J Microbiol ; 52(3): 188-99, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24585050

RESUMO

Communities of microbes can live almost anywhere and contain many different species. Interactions between members of these communities often determine the state of the habitat in which they live. When these habitats include sites on the human body, these interactions can affect health and disease. Polymicrobial synergy can occur during infection, in which the combined effect of two or more microbes on disease is worse than seen with any of the individuals alone. Powerful genomic methods are increasingly used to study microbial communities, including metagenomics to reveal the members and genetic content of a community and metatranscriptomics to describe the activities of community members. Recent efforts focused toward a mechanistic understanding of these interactions have led to a better appreciation of the precise bases of polymicrobial synergy in communities containing bacteria, eukaryotic microbes, and/or viruses. These studies have benefited from advances in the development of in vivo models of polymicrobial infection and modern techniques to profile the spatial and chemical bases of intermicrobial communication. This review describes the breadth of mechanisms microbes use to interact in ways that impact pathogenesis and techniques to study polymicrobial communities.


Assuntos
Bactérias/crescimento & desenvolvimento , Infecções Bacterianas/microbiologia , Fenômenos Fisiológicos Bacterianos , Coinfecção/microbiologia , Interações Microbianas , Animais , Interações Hospedeiro-Patógeno , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA