Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
mSphere ; 9(7): e0036024, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38980072

RESUMEN

Characterizing microbial communities at high resolution and with absolute quantification is crucial to unravel the complexity and diversity of microbial ecosystems. This can be achieved with PCR assays, which enable highly selective detection and absolute quantification of microbial DNA. However, a major challenge that has hindered PCR applications in microbiome research is the design of highly specific primer sets that exclusively amplify intended targets. Here, we introduce Phylogenetically Unique Primers in python (PUPpy), a fully automated pipeline to design microbe- and group-specific primers within a given microbial community. PUPpy can be executed from a user-friendly graphical user interface, or two simple terminal commands, and it only requires coding sequence files of the community members as input. PUPpy-designed primers enable the detection of individual microbes and quantification of absolute microbial abundance in defined communities below the strain level. We experimentally evaluated the performance of PUPpy-designed primers using two bacterial communities as benchmarks. Each community comprises 10 members, exhibiting a range of genetic similarities that spanned from different phyla to substrains. PUPpy-designed primers also enable the detection of groups of bacteria in an undefined community, such as the detection of a gut bacterial family in a complex stool microbiota sample. Taxon-specific primers designed with PUPpy showed 100% specificity to their intended targets, without unintended amplification, in each community tested. Lastly, we show the absolute quantification of microbial abundance using PUPpy-designed primers in droplet digital PCR, benchmarked against 16S rRNA and shotgun sequencing. Our data shows that PUPpy-designed microbe-specific primers can be used to quantify substrain-level absolute counts, providing more resolved and accurate quantification in defined communities than short-read 16S rRNA and shotgun sequencing. IMPORTANCE: Profiling microbial communities at high resolution and with absolute quantification is essential to uncover hidden ecological interactions within microbial ecosystems. Nevertheless, achieving resolved and quantitative investigations has been elusive due to methodological limitations in distinguishing and quantifying highly related microbes. Here, we describe Phylogenetically Unique Primers in python (PUPpy), an automated computational pipeline to design taxon-specific primers within defined microbial communities. Taxon-specific primers can be used to selectively detect and quantify individual microbes and larger taxa within a microbial community. PUPpy achieves substrain-level specificity without the need for computationally intensive databases and prioritizes user-friendliness by enabling both terminal and graphical user interface applications. Altogether, PUPpy enables fast, inexpensive, and highly accurate perspectives into microbial ecosystems, supporting the characterization of bacterial communities in both in vitro and complex microbiota settings.


Asunto(s)
Bacterias , Cartilla de ADN , Microbiota , Reacción en Cadena de la Polimerasa , Bacterias/genética , Bacterias/clasificación , Bacterias/aislamiento & purificación , Cartilla de ADN/genética , Microbiota/genética , Reacción en Cadena de la Polimerasa/métodos , Heces/microbiología , ARN Ribosómico 16S/genética , ADN Bacteriano/genética , Filogenia , Humanos , Microbioma Gastrointestinal/genética , Programas Informáticos , Animales
2.
Proc Natl Acad Sci U S A ; 121(20): e2313971121, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38662573

RESUMEN

There is increasing evidence that interactions between microbes and their hosts not only play a role in determining health and disease but also in emotions, thought, and behavior. Built environments greatly influence microbiome exposures because of their built-in highly specific microbiomes coproduced with myriad metaorganisms including humans, pets, plants, rodents, and insects. Seemingly static built structures host complex ecologies of microorganisms that are only starting to be mapped. These microbial ecologies of built environments are directly and interdependently affected by social, spatial, and technological norms. Advances in technology have made these organisms visible and forced the scientific community and architects to rethink gene-environment and microbe interactions respectively. Thus, built environment design must consider the microbiome, and research involving host-microbiome interaction must consider the built-environment. This paradigm shift becomes increasingly important as evidence grows that contemporary built environments are steadily reducing the microbial diversity essential for human health, well-being, and resilience while accelerating the symptoms of human chronic diseases including environmental allergies, and other more life-altering diseases. New models of design are required to balance maximizing exposure to microbial diversity while minimizing exposure to human-associated diseases. Sustained trans-disciplinary research across time (evolutionary, historical, and generational) and space (cultural and geographical) is needed to develop experimental design protocols that address multigenerational multispecies health and health equity in built environments.


Asunto(s)
Entorno Construido , Microbiota , Animales , Humanos , Microbiota/fisiología
3.
J Vis Exp ; (203)2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38345223

RESUMEN

Bacteriophages (phages) are viruses that infect bacteria with species- and strain-level specificity and are the most abundant biological entities across all known ecosystems. Within bacterial communities, such as those found in the gut microbiota, phages are implicated in regulating microbiota population dynamics and driving bacterial evolution. There has been renewed interest in phage research in the last decade, in part due to the host-specific killing capabilities of lytic phages, which offer a promising tool to counter the increasing threat of antimicrobial resistant bacteria. Furthermore, recent studies demonstrating that phages adhere to intestinal mucus suggest they may have a protective role in preventing bacterial invasion into the underlying epithelium. Importantly, like bacterial microbiomes, disrupted phageomes have been associated with worsened outcomes in diseases such as inflammatory bowel disease. Previous studies have demonstrated that phages can modulate the microbiome of animals and humans through fecal filtrate transplants, benefiting the host's health. With this recent wave of research comes the necessity to establish and standardize protocols for studying phages in the context of the gut microbiome. This protocol provides a set of procedures to study isolated T4 phages and their bacterial host, Escherichia coli, in the context of the murine gastrointestinal tract. The methods described here outline how to start from a phage lysate, administer it to mice and assess effects on bacterial host and phage levels. This protocol can be modified and applied to other phage-bacterial pairs and provides a starting point for studying host-phage dynamics in vivo.


Asunto(s)
Bacteriófagos , Microbiota , Humanos , Ratones , Animales , Bacteriófagos/fisiología , Bacteriófago T4 , Escherichia coli , Tracto Gastrointestinal/microbiología , Intestinos , Bacterias
4.
Nat Rev Microbiol ; 22(2): 105-118, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37740073

RESUMEN

Biogeography is the study of species distribution and diversity within an ecosystem and is at the core of how we understand ecosystem dynamics and interactions at the macroscale. In gut microbial communities, a historical reliance on bulk sequencing to probe community composition and dynamics has overlooked critical processes whereby microscale interactions affect systems-level microbiota function and the relationship with the host. In recent years, higher-resolution sequencing and novel single-cell level data have uncovered an incredible heterogeneity in microbial composition and have enabled a more nuanced spatial understanding of the gut microbiota. In an era when spatial transcriptomics and single-cell imaging and analysis have become key tools in mammalian cell and tissue biology, many of these techniques are now being applied to the microbiota. This fresh approach to intestinal biogeography has given important insights that span temporal and spatial scales, from the discovery of mucus encapsulation of the microbiota to the quantification of bacterial species throughout the gut. In this Review, we highlight emerging knowledge surrounding gut biogeography enabled by the observation and quantification of heterogeneity across multiple scales.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Bacterias/genética , Mamíferos
5.
Curr Biol ; 33(24): R1272-R1274, 2023 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-38113833

RESUMEN

Tropini and Burckhardt introduce inoviruses and discuss how they are unique amongst bacteriophages.


Asunto(s)
Bacteriófagos , Inovirus
6.
mBio ; : e0109123, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37975666

RESUMEN

There is concern that the time taken to publish academic papers in microbiological science has significantly increased in recent years. While the data do not specifically support this, evidence suggests that editors are having to invite more and more reviewers to identify those willing to perform peer review.

7.
mBio ; 14(4): e0075323, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37432034

RESUMEN

Changes to gut environmental factors such as pH and osmolality due to disease or drugs correlate with major shifts in microbiome composition; however, we currently cannot predict which species can tolerate such changes or how the community will be affected. Here, we assessed the growth of 92 representative human gut bacterial strains spanning 28 families across multiple pH values and osmolalities in vitro. The ability to grow in extreme pH or osmolality conditions correlated with the availability of known stress response genes in many cases, but not all, indicating that novel pathways may participate in protecting against acid or osmotic stresses. Machine learning analysis uncovered genes or subsystems that are predictive of differential tolerance in either acid or osmotic stress. For osmotic stress, we corroborated the increased abundance of these genes in vivo during osmotic perturbation. The growth of specific taxa in limiting conditions in isolation in vitro correlated with survival in complex communities in vitro and in an in vivo mouse model of diet-induced intestinal acidification. Our data show that in vitro stress tolerance results are generalizable and that physical parameters may supersede interspecies interactions in determining the relative abundance of community members. This study provides insight into the ability of the microbiota to respond to common perturbations that may be encountered in the gut and provides a list of genes that correlate with increased ability to survive in these conditions. IMPORTANCE To achieve greater predictability in microbiota studies, it is crucial to consider physical environmental factors such as pH and particle concentration, as they play a pivotal role in influencing bacterial function and survival. For example, pH is significantly altered in various diseases, including cancers, inflammatory bowel disease, as well in the case of over-the-counter drug use. Additionally, conditions like malabsorption can affect particle concentration. In our study, we investigate how changes in environmental pH and osmolality can serve as predictive indicators of bacterial growth and abundance. Our research provides a comprehensive resource for anticipating shifts in microbial composition and gene abundance during complex perturbations. Moreover, our findings underscore the significance of the physical environment as a major driver of bacterial composition. Finally, this work emphasizes the necessity of incorporating physical measurements into animal and clinical studies to better understand the factors influencing shifts in microbiota abundance.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Animales , Ratones , Bacterias , Concentración Osmolar , Concentración de Iones de Hidrógeno
8.
Trends Microbiol ; 31(11): 1131-1142, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37100633

RESUMEN

Recent progress in microbial endocrinology has propelled this field from initially providing correlational links to defining the mechanisms by which microbes influence systemic sex hormones. Importantly, the interaction between the gut-resident bacteria and host-secreted hormones has been shown to be critical for host development as well as hormone-mediated disease progression. This review investigates how microbes affect active sex hormone levels, with a focus on gut-associated bacteria hormonal modifications and the resulting host physiological status. Specifically, we focus on the ability of the microbiota to reactivate estrogens and deactivate androgens and thereby influence systemic levels of host hormones in a clinically significant manner.


Asunto(s)
Hormonas Esteroides Gonadales , Microbiota , Hormonas , Microbiota/fisiología , Esteroides
9.
Microbiome ; 11(1): 65, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36991500

RESUMEN

BACKGROUND: Bacteriophages in the family Inoviridae, or inoviruses, are under-characterized phages previously implicated in bacterial pathogenesis by contributing to biofilm formation, immune evasion, and toxin secretion. Unlike most bacteriophages, inoviruses do not lyse their host cells to release new progeny virions; rather, they encode a secretion system that actively pumps them out of the bacterial cell. To date, no inovirus associated with the human gut microbiome has been isolated or characterized. RESULTS: In this study, we utilized in silico, in vitro, and in vivo methods to detect inoviruses in bacterial members of the gut microbiota. By screening a representative genome library of gut commensals, we detected inovirus prophages in Enterocloster spp. (formerly Clostridium spp.). We confirmed the secretion of inovirus particles in in vitro cultures of these organisms using imaging and qPCR. To assess how the gut abiotic environment, bacterial physiology, and inovirus secretion may be linked, we deployed a tripartite in vitro assay that progressively evaluated bacterial growth dynamics, biofilm formation, and inovirus secretion in the presence of changing osmotic environments. Counter to other inovirus-producing bacteria, inovirus production was not correlated with biofilm formation in Enterocloster spp. Instead, the Enterocloster strains had heterogeneous responses to changing osmolality levels relevant to gut physiology. Notably, increasing osmolality induced inovirus secretion in a strain-dependent manner. We confirmed inovirus secretion in a gnotobiotic mouse model inoculated with individual Enterocloster strains in vivo in unperturbed conditions. Furthermore, consistent with our in vitro observations, inovirus secretion was regulated by a changed osmotic environment in the gut due to osmotic laxatives. CONCLUSION: In this study, we report on the detection and characterization of novel inoviruses from gut commensals in the Enterocloster genus. Together, our results demonstrate that human gut-associated bacteria can secrete inoviruses and begin to elucidate the environmental niche filled by inoviruses in commensal bacteria. Video Abstract.


Asunto(s)
Bacteriófagos , Microbioma Gastrointestinal , Inovirus , Ratones , Animales , Humanos , Inovirus/genética , Bacteriófagos/genética , Bacterias , Profagos/genética , Clostridiales
10.
Cell Host Microbe ; 30(7): 905-907, 2022 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-35834961

RESUMEN

Gut microbiota transcripts are notoriously hard to record accurately during perturbations because it is difficult to collect the signals near the source and at the time of variation. A recent study by Schmidt et al. in Science demonstrates a technology that overcomes these barriers.


Asunto(s)
Bacterias , Microbioma Gastrointestinal , Perfilación de la Expresión Génica/métodos , Bacterias/genética
11.
mBio ; 13(2): e0294921, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35266814

RESUMEN

Gut inflammation directly impacts the growth and stability of commensal gut microbes and can lead to long-lasting changes in microbiota composition that can prolong or exacerbate disease states. While mouse models are used extensively to investigate the interplay between microbes and the inflamed state, the paucity of cultured mouse gut microbes has hindered efforts to determine causal relationships. To address this issue, we are assembling the Collection of Inflammation-Associated Mouse Intestinal Bacteria (CIAMIB). The initial release of this collection comprises 41 isolates of 39 unique bacterial species, covering 4 phyla and containing 10 previously uncultivated isolates, including 1 novel family and 7 novel genera. The collection significantly expands the number of available Muribaculaceae, Lachnospiraceae, and Coriobacteriaceae isolates and includes microbes from genera associated with inflammation, such as Prevotella and Klebsiella. We characterized the growth of CIAMIB isolates across a diverse range of nutritional conditions and predicted their metabolic potential and anaerobic fermentation capacity based on the genomes of these isolates. We also provide the first metabolic analysis of species within the genus Adlercreutzia, revealing these representatives to be nitrate-reducing and severely restricted in their ability to grow on carbohydrates. CIAMIB isolates are fully sequenced and available to the scientific community as a powerful tool to study host-microbiota interactions. IMPORTANCE Attempts to explore the role of the microbiota in animal physiology have resulted in large-scale efforts to cultivate the thousands of microbes that are associated with humans. In contrast, relatively few lab mouse-associated bacteria have been isolated, despite the fact that the overwhelming number of studies on the microbiota use laboratory mice that are colonized with microbes that are quite distinct from those in humans. Here, we report the results of a large-scale isolation of bacteria from the intestines of laboratory mice either prone to or suffering from gut inflammation. This collection comprises dozens of novel isolates, many of which represent the only cultured representatives of their genus or species. We report their basic growth characteristics and genomes and are making them widely available to the greater research community.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Bacterias/genética , Microbioma Gastrointestinal/fisiología , Inflamación , Ratones , Simbiosis
12.
mSystems ; 6(4): e0067521, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34427510

RESUMEN

Living systems, from micro- to macro-scales, are strongly impacted by physical factors such as temperature, pH, and the concentration of compounds in their surrounding environment. In the macro-world, it is obvious that small changes in these parameters can have profound, and even devastating, impacts on an ecosystem. For example, in the case of global warming, a change in climate, and specifically a few degrees in temperature, has taken one million species of animals to the brink of extinction. Scale things down 6 orders of magnitude, our gut microbiota also experiences similar changes in temperature due to disease. In this highly competitive environment, physical perturbations inflict long-term consequences on the microbiota ecosystem and, in turn, on the host organism. My laboratory is exploring the feedback between the gut's physical environment, the microbiota, and disease. Our research highlights the importance of measuring physical parameters for the prediction of microbial dynamics and microbiota therapies.

13.
J Vis Exp ; (173)2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34309601

RESUMEN

Measuring the localization of microbes within their in vivo context is an essential step in revealing the functional relationships between the microbiota and the vertebrate gut. The spatial landscape of the gut microbiota is tightly controlled by physical features - intestinal mucus, crypts, and folds - and is affected by host-controlled properties such as pH, oxygen availability, and immune factors. These properties limit the ability of commensal microbes and pathogens alike to colonize the gut stably. At the micron-scale, microbial organization determines the close-range interactions between different microbes as well as the interactions between microbes and their host. These interactions then affect large-scale organ function and host health. This protocol enables the visualization of the gut microbiota spatial organization from distances between cells to organ-wide scales. The method is based on fixing gut tissues while preserving intestinal structure and mucus properties. The fixed samples are then embedded, sectioned, and stained to highlight specific bacterial species through fluorescence in situ hybridization (FISH). Host features, such as mucus and host cell components, are labeled with fluorescently labeled lectins. Finally, the stained sections are imaged using a confocal microscope utilizing tile-scan imaging at high magnification to bridge the micron to centimeter length scales. This type of imaging can be applied to intestinal sections from animal models and biopsies from human tissues to determine the biogeography of the microbiota in the gut in health and disease.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Animales , Humanos , Hibridación Fluorescente in Situ , Lectinas , Moco
14.
Microbes Infect ; 23(6-7): 104815, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33775859

RESUMEN

The human gut hosts a dense and diverse microbial community, spatially organized in multiple scales of structure. Here, we review how microbial organization differs between health and disease. We describe how changes in spatial organization may induce alterations in gut homeostasis, concluding with a future outlook to reveal causality.


Asunto(s)
Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Tracto Gastrointestinal/inmunología , Salud , Humanos
15.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33472859

RESUMEN

The COVID-19 pandemic has the potential to affect the human microbiome in infected and uninfected individuals, having a substantial impact on human health over the long term. This pandemic intersects with a decades-long decline in microbial diversity and ancestral microbes due to hygiene, antibiotics, and urban living (the hygiene hypothesis). High-risk groups succumbing to COVID-19 include those with preexisting conditions, such as diabetes and obesity, which are also associated with microbiome abnormalities. Current pandemic control measures and practices will have broad, uneven, and potentially long-term effects for the human microbiome across the planet, given the implementation of physical separation, extensive hygiene, travel barriers, and other measures that influence overall microbial loss and inability for reinoculation. Although much remains uncertain or unknown about the virus and its consequences, implementing pandemic control practices could significantly affect the microbiome. In this Perspective, we explore many facets of COVID-19-induced societal changes and their possible effects on the microbiome, and discuss current and future challenges regarding the interplay between this pandemic and the microbiome. Recent recognition of the microbiome's influence on human health makes it critical to consider both how the microbiome, shaped by biosocial processes, affects susceptibility to the coronavirus and, conversely, how COVID-19 disease and prevention measures may affect the microbiome. This knowledge may prove key in prevention and treatment, and long-term biological and social outcomes of this pandemic.


Asunto(s)
COVID-19/microbiología , Hipótesis de la Higiene , Microbiota , Anciano , Antiinfecciosos/uso terapéutico , COVID-19/mortalidad , Ingestión de Alimentos , Femenino , Humanos , Lactante , Control de Infecciones/métodos , Masculino , Microbiota/efectos de los fármacos , Distanciamiento Físico , Embarazo
16.
Nature ; 588(7839): 591-592, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33268877
18.
Cell Host Microbe ; 27(4): 659-670.e5, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32101703

RESUMEN

Secondary bile acids (SBAs) are derived from primary bile acids (PBAs) in a process reliant on biosynthetic capabilities possessed by few microbes. To evaluate the role of BAs in intestinal inflammation, we performed metabolomic, microbiome, metagenomic, and transcriptomic profiling of stool from ileal pouches (surgically created resevoirs) in colectomy-treated patients with ulcerative colitis (UC) versus controls (familial adenomatous polyposis [FAP]). We show that relative to FAP, UC pouches have reduced levels of lithocholic acid and deoxycholic acid (normally the most abundant gut SBAs), genes required to convert PBAs to SBAs, and Ruminococcaceae (one of few taxa known to include SBA-producing bacteria). In three murine colitis models, SBA supplementation reduces intestinal inflammation. This anti-inflammatory effect is in part dependent on the TGR5 bile acid receptor. These data suggest that dysbiosis induces SBA deficiency in inflammatory-prone UC patients, which promotes a pro-inflammatory state within the intestine that may be treated by SBA restoration.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Reservorios Cólicos/microbiología , Disbiosis/complicaciones , Heces/microbiología , Receptores Acoplados a Proteínas G/metabolismo , Poliposis Adenomatosa del Colon/microbiología , Animales , Ácidos y Sales Biliares/farmacología , Colitis/etiología , Colitis/microbiología , Modelos Animales de Enfermedad , Humanos , Inflamación/tratamiento farmacológico , Inflamación/etiología , Intestinos/efectos de los fármacos , Intestinos/patología , Metagenoma , Ratones , Microbiota , Receptores Acoplados a Proteínas G/efectos de los fármacos , Ruminococcus/aislamiento & purificación , Transcriptoma
19.
Cell Host Microbe ; 26(5): 650-665.e4, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31726029

RESUMEN

Antibiotics alter microbiota composition and increase infection susceptibility. However, the generalizable effects of antibiotics on and the contribution of environmental variables to gut commensals remain unclear. To address this, we tracked microbiota dynamics with high temporal and taxonomic resolution during antibiotic treatment in a controlled murine system by isolating variables such as diet, treatment history, and housing co-inhabitants. Human microbiotas were remarkably resilient and recovered during antibiotic treatment, with transient dominance of resistant Bacteroides and taxa-asymmetric diversity reduction. In certain cases, in vitro sensitivities were not predictive of in vivo responses, underscoring the significance of host and community context. A fiber-deficient diet exacerbated microbiota collapse and delayed recovery. Species replacement through cross housing after ciprofloxacin treatment established resilience to a second treatment. Single housing drastically disrupted recovery, highlighting the importance of environmental reservoirs. Our findings highlight deterministic microbiota adaptations to perturbations and the translational potential for modulating diet, sanitation, and microbiota composition during antibiotics.


Asunto(s)
Antibacterianos/farmacología , Carga Bacteriana/efectos de los fármacos , Bacteroides/crecimiento & desarrollo , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Animales , Bacteroides/clasificación , Bacteroides/aislamiento & purificación , Biodiversidad , Ciprofloxacina/farmacología , Dieta , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Vida Libre de Gérmenes , Humanos , Masculino , Ratones , Rifaximina/farmacología , Estreptomicina/farmacología
20.
Cell ; 173(7): 1742-1754.e17, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29906449

RESUMEN

Osmotic diarrhea is a prevalent condition in humans caused by food intolerance, malabsorption, and widespread laxative use. Here, we assess the resilience of the gut ecosystem to osmotic perturbation at multiple length and timescales using mice as model hosts. Osmotic stress caused reproducible extinction of highly abundant taxa and expansion of less prevalent members in human and mouse microbiotas. Quantitative imaging revealed decimation of the mucus barrier during osmotic perturbation, followed by recovery. The immune system exhibited temporary changes in cytokine levels and a lasting IgG response against commensal bacteria. Increased osmolality prevented growth of commensal strains in vitro, revealing one mechanism contributing to extinction. Environmental availability of microbiota members mitigated extinction events, demonstrating how species reintroduction can affect community resilience. Our findings (1) demonstrate that even mild osmotic diarrhea can cause lasting changes to the microbiota and host and (2) lay the foundation for interventions that increase system-wide resilience.


Asunto(s)
Diarrea/patología , Microbioma Gastrointestinal/efectos de los fármacos , Polietilenglicoles/farmacología , Animales , Bacteroidetes/efectos de los fármacos , Bacteroidetes/genética , Bacteroidetes/aislamiento & purificación , Ciego/química , Ciego/metabolismo , Ciego/microbiología , Ciego/patología , Colon/química , Colon/microbiología , Colon/patología , Citocinas/metabolismo , Diarrea/inmunología , Diarrea/microbiología , Diarrea/veterinaria , Heces/microbiología , Glicósido Hidrolasas/metabolismo , Humanos , Inmunidad Humoral/efectos de los fármacos , Inmunoglobulina G/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Metagenómica , Ratones , Concentración Osmolar , Polietilenglicoles/metabolismo , Proteoma/análisis , ARN Ribosómico 16S/química , ARN Ribosómico 16S/genética , Verrucomicrobia/efectos de los fármacos , Verrucomicrobia/genética , Verrucomicrobia/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA