Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.636
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 187(19): 5413-5430.e29, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39163861

RESUMO

Bacterial vaginosis (BV), a common syndrome characterized by Lactobacillus-deficient vaginal microbiota, is associated with adverse health outcomes. BV often recurs after standard antibiotic therapy in part because antibiotics promote microbiota dominance by Lactobacillus iners instead of Lactobacillus crispatus, which has more beneficial health associations. Strategies to promote L. crispatus and inhibit L. iners are thus needed. We show that oleic acid (OA) and similar long-chain fatty acids simultaneously inhibit L. iners and enhance L. crispatus growth. These phenotypes require OA-inducible genes conserved in L. crispatus and related lactobacilli, including an oleate hydratase (ohyA) and putative fatty acid efflux pump (farE). FarE mediates OA resistance, while OhyA is robustly active in the vaginal microbiota and enhances bacterial fitness by biochemically sequestering OA in a derivative form only ohyA-harboring organisms can exploit. OA promotes L. crispatus dominance more effectively than antibiotics in an in vitro BV model, suggesting a metabolite-based treatment approach.


Assuntos
Ácidos Graxos , Lactobacillus , Vagina , Vaginose Bacteriana , Vaginose Bacteriana/tratamento farmacológico , Vaginose Bacteriana/microbiologia , Feminino , Humanos , Vagina/microbiologia , Lactobacillus/metabolismo , Ácidos Graxos/metabolismo , Ácido Oleico/metabolismo , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Lactobacillus crispatus/metabolismo , Microbiota/efeitos dos fármacos , Proteínas de Bactérias/metabolismo
2.
Cell ; 187(19): 5453-5467.e15, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39163860

RESUMO

Drug-resistant bacteria are outpacing traditional antibiotic discovery efforts. Here, we computationally screened 444,054 previously reported putative small protein families from 1,773 human metagenomes for antimicrobial properties, identifying 323 candidates encoded in small open reading frames (smORFs). To test our computational predictions, 78 peptides were synthesized and screened for antimicrobial activity in vitro, with 70.5% displaying antimicrobial activity. As these compounds were different compared with previously reported antimicrobial peptides, we termed them smORF-encoded peptides (SEPs). SEPs killed bacteria by targeting their membrane, synergizing with each other, and modulating gut commensals, indicating a potential role in reconfiguring microbiome communities in addition to counteracting pathogens. The lead candidates were anti-infective in both murine skin abscess and deep thigh infection models. Notably, prevotellin-2 from Prevotella copri presented activity comparable to the commonly used antibiotic polymyxin B. Our report supports the existence of hundreds of antimicrobials in the human microbiome amenable to clinical translation.


Assuntos
Antibacterianos , Peptídeos Antimicrobianos , Microbiota , Humanos , Animais , Camundongos , Antibacterianos/farmacologia , Microbiota/efeitos dos fármacos , Peptídeos Antimicrobianos/farmacologia , Peptídeos Antimicrobianos/química , Metagenoma , Feminino , Fases de Leitura Aberta , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/classificação , Prevotella/efeitos dos fármacos
3.
Cell ; 184(21): 5405-5418.e16, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34619078

RESUMO

Lyme disease is on the rise. Caused by a spirochete Borreliella burgdorferi, it affects an estimated 500,000 people in the United States alone. The antibiotics currently used to treat Lyme disease are broad spectrum, damage the microbiome, and select for resistance in non-target bacteria. We therefore sought to identify a compound acting selectively against B. burgdorferi. A screen of soil micro-organisms revealed a compound highly selective against spirochetes, including B. burgdorferi. Unexpectedly, this compound was determined to be hygromycin A, a known antimicrobial produced by Streptomyces hygroscopicus. Hygromycin A targets the ribosomes and is taken up by B. burgdorferi, explaining its selectivity. Hygromycin A cleared the B. burgdorferi infection in mice, including animals that ingested the compound in a bait, and was less disruptive to the fecal microbiome than clinically relevant antibiotics. This selective antibiotic holds the promise of providing a better therapeutic for Lyme disease and eradicating it in the environment.


Assuntos
Antibacterianos/uso terapêutico , Doença de Lyme/tratamento farmacológico , Animais , Borrelia burgdorferi/efeitos dos fármacos , Calibragem , Cinamatos/química , Cinamatos/farmacologia , Cinamatos/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Fezes/microbiologia , Feminino , Células HEK293 , Células Hep G2 , Humanos , Higromicina B/análogos & derivados , Higromicina B/química , Higromicina B/farmacologia , Higromicina B/uso terapêutico , Doença de Lyme/microbiologia , Camundongos , Testes de Sensibilidade Microbiana , Microbiota/efeitos dos fármacos
4.
Cell ; 184(19): 5015-5030.e16, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34407392

RESUMO

Group 3 innate lymphoid cells (ILC3s) regulate immunity and inflammation, yet their role in cancer remains elusive. Here, we identify that colorectal cancer (CRC) manifests with altered ILC3s that are characterized by reduced frequencies, increased plasticity, and an imbalance with T cells. We evaluated the consequences of these changes in mice and determined that a dialog between ILC3s and T cells via major histocompatibility complex class II (MHCII) is necessary to support colonization with microbiota that subsequently induce type-1 immunity in the intestine and tumor microenvironment. As a result, mice lacking ILC3-specific MHCII develop invasive CRC and resistance to anti-PD-1 immunotherapy. Finally, humans with dysregulated intestinal ILC3s harbor microbiota that fail to induce type-1 immunity and immunotherapy responsiveness when transferred to mice. Collectively, these data define a protective role for ILC3s in cancer and indicate that their inherent disruption in CRC drives dysfunctional adaptive immunity, tumor progression, and immunotherapy resistance.


Assuntos
Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Progressão da Doença , Imunidade Inata , Imunoterapia , Linfócitos/imunologia , Animais , Comunicação Celular/efeitos dos fármacos , Plasticidade Celular/efeitos dos fármacos , Neoplasias do Colo/microbiologia , Fezes/microbiologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade Inata/efeitos dos fármacos , Inflamação/imunologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/patologia , Intestinos/patologia , Linfócitos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Invasividade Neoplásica , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Doadores de Tecidos
5.
Cell ; 184(21): 5338-5356.e21, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34624222

RESUMO

The tumor microenvironment (TME) influences cancer progression and therapy response. Therefore, understanding what regulates the TME immune compartment is vital. Here we show that microbiota signals program mononuclear phagocytes in the TME toward immunostimulatory monocytes and dendritic cells (DCs). Single-cell RNA sequencing revealed that absence of microbiota skews the TME toward pro-tumorigenic macrophages. Mechanistically, we show that microbiota-derived stimulator of interferon genes (STING) agonists induce type I interferon (IFN-I) production by intratumoral monocytes to regulate macrophage polarization and natural killer (NK) cell-DC crosstalk. Microbiota modulation with a high-fiber diet triggered the intratumoral IFN-I-NK cell-DC axis and improved the efficacy of immune checkpoint blockade (ICB). We validated our findings in individuals with melanoma treated with ICB and showed that the predicted intratumoral IFN-I and immune compositional differences between responder and non-responder individuals can be transferred by fecal microbiota transplantation. Our study uncovers a mechanistic link between the microbiota and the innate TME that can be harnessed to improve cancer therapies.


Assuntos
Interferon Tipo I/metabolismo , Proteínas de Membrana/metabolismo , Microbiota , Monócitos/metabolismo , Microambiente Tumoral , Akkermansia/efeitos dos fármacos , Akkermansia/fisiologia , Animais , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Fibras na Dieta/farmacologia , Fosfatos de Dinucleosídeos/administração & dosagem , Fosfatos de Dinucleosídeos/farmacologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunomodulação/efeitos dos fármacos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Melanoma/imunologia , Melanoma/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Fagócitos/efeitos dos fármacos , Fagócitos/metabolismo , Transcrição Gênica/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
6.
Cell ; 181(7): 1661-1679.e22, 2020 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-32526207

RESUMO

The human gut microbiome harbors hundreds of bacterial species with diverse biochemical capabilities. Dozens of drugs have been shown to be metabolized by single isolates from the gut microbiome, but the extent of this phenomenon is rarely explored in the context of microbial communities. Here, we develop a quantitative experimental framework for mapping the ability of the human gut microbiome to metabolize small molecule drugs: Microbiome-Derived Metabolism (MDM)-Screen. Included are a batch culturing system for sustained growth of subject-specific gut microbial communities, an ex vivo drug metabolism screen, and targeted and untargeted functional metagenomic screens to identify microbiome-encoded genes responsible for specific metabolic events. Our framework identifies novel drug-microbiome interactions that vary between individuals and demonstrates how the gut microbiome might be used in drug development and personalized medicine.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Microbioma Gastrointestinal/fisiologia , Microbiota/efeitos dos fármacos , Adulto , Animais , Bactérias/classificação , Biomarcadores Farmacológicos/metabolismo , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/genética , Voluntários Saudáveis , Humanos , Masculino , Metagenoma/genética , Metagenômica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/genética , Preparações Farmacêuticas/metabolismo , Medicina de Precisão/métodos , RNA Ribossômico 16S/genética
7.
Cell ; 182(3): 609-624.e21, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32640190

RESUMO

Gastrointestinal enterochromaffin cells regulate bone and gut homeostasis via serotonin (5-hydroxytryptamine [5-HT]) production. A recent report suggested that gut microbes regulate 5-HT levels; however, the precise underlying molecular mechanisms are unexplored. Here, we reveal that the cation channel Piezo1 in the gut acts as a sensor of single-stranded RNA (ssRNA) governing 5-HT production. Intestinal epithelium-specific deletion of mouse Piezo1 profoundly disturbed gut peristalsis, impeded experimental colitis, and suppressed serum 5-HT levels. Because of systemic 5-HT deficiency, conditional knockout of Piezo1 increased bone formation. Notably, fecal ssRNA was identified as a natural Piezo1 ligand, and ssRNA-stimulated 5-HT synthesis from the gut was evoked in a MyD88/TRIF-independent manner. Colonic infusion of RNase A suppressed gut motility and increased bone mass. These findings suggest gut ssRNA as a master determinant of systemic 5-HT levels, indicating the ssRNA-Piezo1 axis as a potential prophylactic target for treatment of bone and gut disorders.


Assuntos
Osso e Ossos/metabolismo , Colo/metabolismo , Motilidade Gastrointestinal/genética , Canais Iônicos/metabolismo , RNA/metabolismo , Serotonina/biossíntese , Serotonina/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Osso e Ossos/citologia , Cálcio/metabolismo , Colite/genética , Colite/metabolismo , Colite/prevenção & controle , Colo/fisiologia , Fezes/química , Feminino , Motilidade Gastrointestinal/fisiologia , Células HEK293 , Humanos , Imuno-Histoquímica , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Canais Iônicos/genética , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/metabolismo , Osteoclastos/metabolismo , Pirazinas/farmacologia , RNA/farmacologia , Ribonuclease Pancreático/administração & dosagem , Serotonina/sangue , Serotonina/deficiência , Tiadiazóis/farmacologia
8.
Cell ; 183(5): 1312-1324.e10, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33212011

RESUMO

Interferon (IFN)-Is are crucial mediators of antiviral immunity and homeostatic immune system regulation. However, the source of IFN-I signaling under homeostatic conditions is unclear. We discovered that commensal microbes regulate the IFN-I response through induction of IFN-ß by colonic DCs. Moreover, the mechanism by which a specific commensal microbe induces IFN-ß was identified. Outer membrane (OM)-associated glycolipids of gut commensal microbes belonging to the Bacteroidetes phylum induce expression of IFN-ß. Using Bacteroides fragilis and its OM-associated polysaccharide A, we determined that IFN-ß expression was induced via TLR4-TRIF signaling. Antiviral activity of this purified microbial molecule against infection with either vesicular stomatitis virus (VSV) or influenza was demonstrated to be dependent on the induction of IFN-ß. In a murine VSV infection model, commensal-induced IFN-ß regulated natural resistance to virus infection. Due to the physiological importance of IFN-Is, discovery of an IFN-ß-inducing microbial molecule represents a potential approach for the treatment of some human diseases.


Assuntos
Imunidade Inata , Microbiota , Viroses/microbiologia , Animais , Bacteroides fragilis/fisiologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Colo/patologia , Colo/virologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glicolipídeos/metabolismo , Imunidade Inata/efeitos dos fármacos , Interferon beta/sangue , Interferon beta/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Polissacarídeos Bacterianos/farmacologia , Receptor 4 Toll-Like/metabolismo , Vesiculovirus/fisiologia , Viroses/genética
9.
Cell ; 178(5): 1041-1056, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31442399

RESUMO

The current understanding of inflammatory bowel disease (IBD) pathogenesis implicates a complex interaction between host genetics, host immunity, microbiome, and environmental exposures. Mechanisms gleaned from genetics and molecular pathogenesis offer clues to the critical triggers of mucosal inflammation and guide the development of therapeutic interventions. A complex network of interactions between host genetic factors, microbes, and microbial metabolites governs intestinal homeostasis, making classification and mechanistic dissection of involved pathways challenging. In this Review, we discuss these challenges, areas of active translation, and opportunities for development of next-generation therapies.


Assuntos
Doenças Inflamatórias Intestinais/patologia , Microbiota , Imunidade Adaptativa , Animais , Bactérias/genética , Bactérias/metabolismo , Produtos Biológicos/farmacologia , Citocinas/genética , Citocinas/metabolismo , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Microbiota/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
10.
Cell ; 158(4): 705-721, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25126780

RESUMO

Acquisition of the intestinal microbiota begins at birth, and a stable microbial community develops from a succession of key organisms. Disruption of the microbiota during maturation by low-dose antibiotic exposure can alter host metabolism and adiposity. We now show that low-dose penicillin (LDP), delivered from birth, induces metabolic alterations and affects ileal expression of genes involved in immunity. LDP that is limited to early life transiently perturbs the microbiota, which is sufficient to induce sustained effects on body composition, indicating that microbiota interactions in infancy may be critical determinants of long-term host metabolic effects. In addition, LDP enhances the effect of high-fat diet induced obesity. The growth promotion phenotype is transferrable to germ-free hosts by LDP-selected microbiota, showing that the altered microbiota, not antibiotics per se, play a causal role. These studies characterize important variables in early-life microbe-host metabolic interaction and identify several taxa consistently linked with metabolic alterations. PAPERCLIP:


Assuntos
Antibacterianos/administração & dosagem , Modelos Animais de Doenças , Intestinos/microbiologia , Microbiota , Obesidade/microbiologia , Penicilinas/administração & dosagem , Animais , Bactérias/classificação , Bactérias/metabolismo , Feminino , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Obesidade/metabolismo
11.
Immunity ; 50(2): 432-445.e7, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30683619

RESUMO

Host microbial cross-talk is essential to maintain intestinal homeostasis. However, maladaptation of this response through microbial dysbiosis or defective host defense toward invasive intestinal bacteria can result in chronic inflammation. We have shown that macrophages differentiated in the presence of the bacterial metabolite butyrate display enhanced antimicrobial activity. Butyrate-induced antimicrobial activity was associated with a shift in macrophage metabolism, a reduction in mTOR kinase activity, increased LC3-associated host defense and anti-microbial peptide production in the absence of an increased inflammatory cytokine response. Butyrate drove this monocyte to macrophage differentiation program through histone deacetylase 3 (HDAC3) inhibition. Administration of butyrate induced antimicrobial activity in intestinal macrophages in vivo and increased resistance to enteropathogens. Our data suggest that (1) increased intestinal butyrate might represent a strategy to bolster host defense without tissue damaging inflammation and (2) that pharmacological HDAC3 inhibition might drive selective macrophage functions toward antimicrobial host defense.


Assuntos
Anti-Infecciosos/farmacologia , Butiratos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Animais , Diferenciação Celular/genética , Células Cultivadas , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/microbiologia , Citocinas/genética , Citocinas/metabolismo , Disbiose/microbiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Microbiota/fisiologia , Monócitos/metabolismo , Monócitos/microbiologia
12.
Mol Cell ; 78(4): 597-613, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32208169

RESUMO

Commensal microbial communities inhabit biological niches in the mammalian host, where they impact the host's physiology through induction of "colonization resistance" against infections by a multitude of molecular mechanisms. These colonization-regulating activities involve microbe-microbe and microbe-host interactions, which induce, through utilization of complex bacterial networks, competition over nutrients, inhibition by antimicrobial peptides, stimulation of the host immune system, and promotion of mucus and intestinal epithelial barrier integrity. Distinct virulent pathogens overcome this colonization resistance and host immunity as part of a hostile takeover of the host niche, leading to clinically overt infection. The following review provides a mechanistic overview of the role of commensal microbes in modulating colonization resistance and pathogenic infections and means by which infectious agents may overcome such inhibition. Last, we outline evidence, unknowns, and challenges in developing strategies to harness this knowledge to treat infections by microbiota transfer, phage therapy, or supplementation by rationally defined bacterial consortia.


Assuntos
Bactérias/imunologia , Resistência Microbiana a Medicamentos/imunologia , Interações Hospedeiro-Patógeno/imunologia , Infecções/imunologia , Infecções/microbiologia , Microbiota , Virulência/imunologia , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Humanos , Infecções/metabolismo , Microbiota/efeitos dos fármacos
13.
Nature ; 599(7883): 120-124, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34646011

RESUMO

Antibiotics are used to fight pathogens but also target commensal bacteria, disturbing the composition of gut microbiota and causing dysbiosis and disease1. Despite this well-known collateral damage, the activity spectrum of different antibiotic classes on gut bacteria remains poorly characterized. Here we characterize further 144 antibiotics from a previous screen of more than 1,000 drugs on 38 representative human gut microbiome species2. Antibiotic classes exhibited distinct inhibition spectra, including generation dependence for quinolones and phylogeny independence for ß-lactams. Macrolides and tetracyclines, both prototypic bacteriostatic protein synthesis inhibitors, inhibited nearly all commensals tested but also killed several species. Killed bacteria were more readily eliminated from in vitro communities than those inhibited. This species-specific killing activity challenges the long-standing distinction between bactericidal and bacteriostatic antibiotic classes and provides a possible explanation for the strong effect of macrolides on animal3-5 and human6,7 gut microbiomes. To mitigate this collateral damage of macrolides and tetracyclines, we screened for drugs that specifically antagonized the antibiotic activity against abundant Bacteroides species but not against relevant pathogens. Such antidotes selectively protected Bacteroides species from erythromycin treatment in human-stool-derived communities and gnotobiotic mice. These findings illluminate the activity spectra of antibiotics in commensal bacteria and suggest strategies to circumvent their adverse effects on the gut microbiota.


Assuntos
Antibacterianos/efeitos adversos , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Animais , Antibacterianos/classificação , Bactérias/classificação , Bactérias Anaeróbias/efeitos dos fármacos , Bacteroides/efeitos dos fármacos , Clostridioides difficile/efeitos dos fármacos , Dicumarol/farmacologia , Eritromicina/farmacologia , Fezes/microbiologia , Feminino , Vida Livre de Germes , Humanos , Macrolídeos/farmacologia , Masculino , Camundongos , Microbiota/efeitos dos fármacos , Simbiose/efeitos dos fármacos , Tetraciclinas/farmacologia
14.
Nature ; 588(7839): 676-681, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33268897

RESUMO

Mapping the complex biogeography of microbial communities in situ with high taxonomic and spatial resolution poses a major challenge because of the high density1 and rich diversity2 of species in environmental microbiomes and the limitations of optical imaging technology3-6. Here we introduce high-phylogenetic-resolution microbiome mapping by fluorescence in situ hybridization (HiPR-FISH), a versatile technology that uses binary encoding, spectral imaging and decoding based on machine learning to create micrometre-scale maps of the locations and identities of hundreds of microbial species in complex communities. We show that 10-bit HiPR-FISH can distinguish between 1,023 isolates of Escherichia coli, each fluorescently labelled with a unique binary barcode. HiPR-FISH, in conjunction with custom algorithms for automated probe design and analysis of single-cell images, reveals the disruption of spatial networks in the mouse gut microbiome in response to treatment with antibiotics, and the longitudinal stability of spatial architectures in the human oral plaque microbiome. Combined with super-resolution imaging, HiPR-FISH shows the diverse strategies of ribosome organization that are exhibited by taxa in the human oral microbiome. HiPR-FISH provides a framework for analysing the spatial ecology of environmental microbial communities at single-cell resolution.


Assuntos
Hibridização in Situ Fluorescente/métodos , Microbiota , Algoritmos , Animais , Antibacterianos/farmacologia , Biofilmes , Escherichia coli/classificação , Escherichia coli/citologia , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Camundongos , Microbiota/efeitos dos fármacos , Boca/efeitos dos fármacos , Boca/microbiologia , Ribossomos/metabolismo , Análise de Célula Única
15.
N Engl J Med ; 386(3): 220-229, 2022 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-35045228

RESUMO

BACKGROUND: Current therapies for recurrent Clostridioides difficile infection do not address the disrupted microbiome, which supports C. difficile spore germination into toxin-producing bacteria. SER-109 is an investigational microbiome therapeutic composed of purified Firmicutes spores for the treatment of recurrent C. difficile infection. METHODS: We conducted a phase 3, double-blind, randomized, placebo-controlled trial in which patients who had had three or more episodes of C. difficile infection (inclusive of the qualifying acute episode) received SER-109 or placebo (four capsules daily for 3 days) after standard-of-care antibiotic treatment. The primary efficacy objective was to show superiority of SER-109 as compared with placebo in reducing the risk of C. difficile infection recurrence up to 8 weeks after treatment. Diagnosis by toxin testing was performed at trial entry, and randomization was stratified according to age and antibiotic agent received. Analyses of safety, microbiome engraftment, and metabolites were also performed. RESULTS: Among the 281 patients screened, 182 were enrolled. The percentage of patients with recurrence of C. difficile infection was 12% in the SER-109 group and 40% in the placebo group (relative risk, 0.32; 95% confidence interval [CI], 0.18 to 0.58; P<0.001 for a relative risk of <1.0; P<0.001 for a relative risk of <0.833). SER-109 led to less frequent recurrence than placebo in analyses stratified according to age stratum (relative risk, 0.24 [95% CI, 0.07 to 0.78] for patients <65 years of age and 0.36 [95% CI, 0.18 to 0.72] for those ≥65 years) and antibiotic received (relative risk, 0.41 [95% CI, 0.22 to 0.79] with vancomycin and 0.09 [95% CI, 0.01 to 0.63] with fidaxomicin). Most adverse events were mild to moderate and were gastrointestinal in nature, with similar numbers in the two groups. SER-109 dose species were detected as early as week 1 and were associated with bile-acid profiles that are known to inhibit C. difficile spore germination. CONCLUSIONS: In patients with symptom resolution of C. difficile infection after treatment with standard-of-care antibiotics, oral administration of SER-109 was superior to placebo in reducing the risk of recurrent infection. The observed safety profile of SER-109 was similar to that of placebo. (Funded by Seres Therapeutics; ECOSPOR III ClinicalTrials.gov number, NCT03183128.).


Assuntos
Clostridioides difficile , Infecções por Clostridium/terapia , Firmicutes , Idoso , Antibacterianos/efeitos adversos , Método Duplo-Cego , Fezes/microbiologia , Feminino , Trato Gastrointestinal/microbiologia , Humanos , Análise de Intenção de Tratamento , Masculino , Microbiota/efeitos dos fármacos , Pessoa de Meia-Idade , Recidiva , Prevenção Secundária , Esporos Bacterianos
16.
FASEB J ; 38(16): e23882, 2024 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-39143727

RESUMO

Phillygenin (PHI) is an active ingredient derived from the leaf of Forsythia suspensa that has been found to alleviate inflammation and peroxidation response. Avian infectious bronchitis (IB) is a major threat to poultry industry viral respiratory tract disease that infected with infectious bronchitis virus (IBV). This study investigated the protection of PHI to CEK cell and broiler's tracheal injury triggered by avian infectious bronchitis virus (IBV). The results showed that IBV infection did not cause serious clinical symptoms and slowing-body weight in PHI-treated broilers. The expression of virus loads, pro-inflammation factors (IL-6, TNF-α, and IL-1ß) in CEK cell, and tracheas were decreased compared to the IBV group, exhibiting its potent anti-inflammation. Mechanistically, the study demonstrated that the inhibition of TLR7/MyD88/NF-κB pathway was mainly involved in the protection effect of PHI to inflammation injury. Interestingly, a higher abundance of Firmicutes and Lactobacillus in respiratory tract was observed in PHI-treated broilers than in the IBV group. Significant differences were observed between the IBV group and PHI-treated group in the Ferroptosis, Tryptophan metabolism, and Glutathione metabolism pathways. PHI exhibited potent protection effect on IBV infection and alleviated inflammation injury, mainly through inhibiting TLR7/MyD88/NF-κB pathway. The study encourages further development of PHI, paving the way to its clinical use as a new candidate drug to relieve IBV-induced respiratory symptoms.


Assuntos
Galinhas , Infecções por Coronavirus , Vírus da Bronquite Infecciosa , Fator 88 de Diferenciação Mieloide , NF-kappa B , Doenças das Aves Domésticas , Receptor 7 Toll-Like , Animais , NF-kappa B/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Doenças das Aves Domésticas/tratamento farmacológico , Doenças das Aves Domésticas/microbiologia , Doenças das Aves Domésticas/metabolismo , Receptor 7 Toll-Like/metabolismo , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/veterinária , Microbiota/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
17.
Nature ; 574(7779): 543-548, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31645720

RESUMO

Multicellular organisms have co-evolved with complex consortia of viruses, bacteria, fungi and parasites, collectively referred to as the microbiota1. In mammals, changes in the composition of the microbiota can influence many physiologic processes (including development, metabolism and immune cell function) and are associated with susceptibility to multiple diseases2. Alterations in the microbiota can also modulate host behaviours-such as social activity, stress, and anxiety-related responses-that are linked to diverse neuropsychiatric disorders3. However, the mechanisms by which the microbiota influence neuronal activity and host behaviour remain poorly defined. Here we show that manipulation of the microbiota in antibiotic-treated or germ-free adult mice results in significant deficits in fear extinction learning. Single-nucleus RNA sequencing of the medial prefrontal cortex of the brain revealed significant alterations in gene expression in excitatory neurons, glia and other cell types. Transcranial two-photon imaging showed that deficits in extinction learning after manipulation of the microbiota in adult mice were associated with defective learning-related remodelling of postsynaptic dendritic spines and reduced activity in cue-encoding neurons in the medial prefrontal cortex. In addition, selective re-establishment of the microbiota revealed a limited neonatal developmental window in which microbiota-derived signals can restore normal extinction learning in adulthood. Finally, unbiased metabolomic analysis identified four metabolites that were significantly downregulated in germ-free mice and have been reported to be related to neuropsychiatric disorders in humans and mouse models, suggesting that microbiota-derived compounds may directly affect brain function and behaviour. Together, these data indicate that fear extinction learning requires microbiota-derived signals both during early postnatal neurodevelopment and in adult mice, with implications for our understanding of how diet, infection, and lifestyle influence brain health and subsequent susceptibility to neuropsychiatric disorders.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Metabolômica , Microbiota/fisiologia , Neurônios/fisiologia , Animais , Antibacterianos/farmacologia , Transtorno Autístico/metabolismo , Sangue/metabolismo , Cálcio/metabolismo , Líquido Cefalorraquidiano/química , Líquido Cefalorraquidiano/metabolismo , Sinais (Psicologia) , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Espinhas Dendríticas/fisiologia , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Fezes/química , Vida Livre de Germes , Indicã/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Microbiota/imunologia , Inibição Neural , Neuroglia/patologia , Neuroglia/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/patologia , Fenilpropionatos/metabolismo , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/imunologia , Córtex Pré-Frontal/fisiologia , Esquizofrenia/metabolismo , Transcriptoma , Nervo Vago/fisiologia
18.
Am J Respir Crit Care Med ; 209(11): 1360-1375, 2024 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-38271553

RESUMO

Rationale: Chronic lung allograft dysfunction (CLAD) is the leading cause of death after lung transplant, and azithromycin has variable efficacy in CLAD. The lung microbiome is a risk factor for developing CLAD, but the relationship between lung dysbiosis, pulmonary inflammation, and allograft dysfunction remains poorly understood. Whether lung microbiota predict outcomes or modify treatment response after CLAD is unknown. Objectives: To determine whether lung microbiota predict post-CLAD outcomes and clinical response to azithromycin. Methods: Retrospective cohort study using acellular BAL fluid prospectively collected from recipients of lung transplant within 90 days of CLAD onset. Lung microbiota were characterized using 16S rRNA gene sequencing and droplet digital PCR. In two additional cohorts, causal relationships of dysbiosis and inflammation were evaluated by comparing lung microbiota with CLAD-associated cytokines and measuring ex vivo P. aeruginosa growth in sterilized BAL fluid. Measurements and Main Results: Patients with higher bacterial burden had shorter post-CLAD survival, independent of CLAD phenotype, azithromycin treatment, and relevant covariates. Azithromycin treatment improved survival in patients with high bacterial burden but had negligible impact on patients with low or moderate burden. Lung bacterial burden was positively associated with CLAD-associated cytokines, and ex vivo growth of P. aeruginosa was augmented in BAL fluid from transplant recipients with CLAD. Conclusions: In recipients of lung transplants with chronic rejection, increased lung bacterial burden is an independent risk factor for mortality and predicts clinical response to azithromycin. Lung bacterial dysbiosis is associated with alveolar inflammation and may be promoted by underlying lung allograft dysfunction.


Assuntos
Azitromicina , Rejeição de Enxerto , Transplante de Pulmão , Microbiota , Humanos , Azitromicina/uso terapêutico , Masculino , Feminino , Pessoa de Meia-Idade , Rejeição de Enxerto/microbiologia , Rejeição de Enxerto/prevenção & controle , Estudos Retrospectivos , Adulto , Microbiota/efeitos dos fármacos , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Pulmão/microbiologia , Doença Crônica , Transplantados/estatística & dados numéricos , Idoso , Disbiose , Estudos de Coortes , Líquido da Lavagem Broncoalveolar/microbiologia
19.
Proc Natl Acad Sci U S A ; 119(15): e2116954119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35394868

RESUMO

Microbial communities often face external perturbations that can induce lasting changes in their composition and functions. Our understanding of how multispecies communities respond to perturbations such as antibiotics is limited, with susceptibility assays performed on individual, isolated species our primary guide in predicting community transitions. Here, we studied how bacterial growth dynamics can overcome differences in antibiotic susceptibility in determining community resilience: the recovery of the original community state following antibiotic exposure. We used an experimental community containing Corynebacterium ammoniagenes and Lactobacillus plantarum that displays two alternative stable states as a result of mutual inhibition. Although C. ammoniagenes was more susceptible to chloramphenicol in monocultures, we found that chloramphenicol exposure nonetheless led to a transition from the L. plantarum-dominated to the C. ammoniagenes-dominated community state. Combining theory and experiments, we demonstrated that growth rate differences between the two species made the L. plantarum-dominated community less resilient to several antibiotics with different mechanisms of action. Taking advantage of an observed cooperativity­a dependence on population abundance­in the growth of C. ammoniagenes, we next analyzed in silico scenarios that could compromise the high resilience of the C. ammoniagenes-dominated state. The model predicted that lowering the dispersal rate, through interacting with the growth at low population densities, could make the C. ammoniagenes state fragile against virtually any kind of antibiotic, a prediction that we confirmed experimentally. Our results highlight that species susceptibility to antibiotics is often uninformative of community resilience, as growth dynamics in the wake of antibiotic exposure can play a dominant role.


Assuntos
Antibacterianos , Corynebacterium , Resistência Microbiana a Medicamentos , Lactobacillus plantarum , Microbiota , Adaptação Fisiológica , Antibacterianos/farmacologia , Corynebacterium/efeitos dos fármacos , Corynebacterium/crescimento & desenvolvimento , Lactobacillus plantarum/efeitos dos fármacos , Lactobacillus plantarum/crescimento & desenvolvimento , Microbiota/efeitos dos fármacos , Microbiota/fisiologia
20.
Proc Natl Acad Sci U S A ; 119(26): e2200348119, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35727974

RESUMO

Immune checkpoint inhibitors (ICIs) are essential components of the cancer therapeutic armamentarium. While ICIs have demonstrated remarkable clinical responses, they can be accompanied by immune-related adverse events (irAEs). These inflammatory side effects are of unclear etiology and impact virtually all organ systems, with the most common being sites colonized by the microbiota such as the skin and gastrointestinal tract. Here, we establish a mouse model of commensal bacteria-driven skin irAEs and demonstrate that immune checkpoint inhibition unleashes commensal-specific inflammatory T cell responses. These aberrant responses were dependent on production of IL-17 by commensal-specific T cells and induced pathology that recapitulated the cutaneous inflammation seen in patients treated with ICIs. Importantly, aberrant T cell responses unleashed by ICIs were sufficient to perpetuate inflammatory memory responses to the microbiota months following the cessation of treatment. Altogether, we have established a mouse model of skin irAEs and reveal that ICIs unleash aberrant immune responses against skin commensals, with long-lasting inflammatory consequences.


Assuntos
Dermatite , Inibidores de Checkpoint Imunológico , Microbiota , Animais , Dermatite/imunologia , Dermatite/microbiologia , Modelos Animais de Doenças , Inibidores de Checkpoint Imunológico/efeitos adversos , Imunidade/efeitos dos fármacos , Interleucina-17/metabolismo , Camundongos , Microbiota/efeitos dos fármacos , Microbiota/imunologia , Staphylococcus epidermidis/efeitos dos fármacos , Staphylococcus epidermidis/imunologia , Simbiose/efeitos dos fármacos , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA