Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
2.
Gut Microbes ; 15(2): 2266627, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37853762

RESUMEN

Immigration to a highly industrialized nation has been associated with metabolic disease and simultaneous shifts in microbiota composition, but the underlying mechanisms are challenging to test in human studies. Here, we conducted a pilot study to assess the differential effects of human gut microbiota collected from the United States (US) and rural Thailand on the murine gut mucosa and immune system. Colonization of germ-free mice with microbiota from US individuals resulted in an increased accumulation of innate-like CD8 T cells in the small intestine lamina propria and intra-epithelial compartments when compared to colonization with microbiota from Thai individuals. Both TCRγδ and CD8αα T cells showed a marked increase in mice receiving Western microbiota and, interestingly, this phenotype was also associated with an increase in intestinal mucus thickness. Serendipitously, an accidentally infected group of mice corroborated this association between elevated inflammatory response and increased mucus thickness. These results suggest that Western-associated human gut microbes contribute to a pro-inflammatory immune response.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Ratones , Animales , Inflamación , Proyectos Piloto , Mucosa Intestinal/metabolismo , Moco , Linfocitos T CD8-positivos
3.
Sci Rep ; 13(1): 7668, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37169816

RESUMEN

The microbiome is known to play a role in many human diseases, but identifying key microbes and their functions generally requires large studies due to the vast number of species and genes, and the high levels of intra-individual and inter-individual variation. 16S amplicon sequencing of the rRNA gene is commonly used for large studies due to its comparatively low sequencing cost, but it has poor taxonomic and functional resolution. Deep shotgun sequencing is a more accurate and comprehensive alternative for small studies, but can be cost-prohibitive for biomarker discovery in large populations. Shallow or moderate-depth shotgun metagenomics may serve as a viable alternative to 16S sequencing for large-scale and/or dense longitudinal studies, but only if resolution and reproducibility are comparable. Here we applied both 16S and shallow shotgun stool microbiome sequencing to a cohort of 5 subjects sampled twice daily and weekly, with technical replication at the DNA extraction and the library preparation/sequencing steps, for a total of 80 16S samples and 80 shallow shotgun sequencing samples. We found that shallow shotgun sequencing produced lower technical variation and higher taxonomic resolution than 16S sequencing, at a much lower cost than deep shotgun sequencing. These findings suggest that shallow shotgun sequencing provides a more specific and more reproducible alternative to 16S sequencing for large-scale microbiome studies where costs prohibit deep shotgun sequencing and where bacterial species are expected to have good coverage in whole-genome reference databases.


Asunto(s)
Microbiota , Humanos , Reproducibilidad de los Resultados , ARN Ribosómico 16S/genética , Microbiota/genética , Bacterias/genética , Análisis de Secuencia de ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Metagenómica
4.
Gut ; 72(4): 759-771, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36343978

RESUMEN

OBJECTIVE: First decompensation development is a critical milestone that needs to be predicted. Transkingdom gut microbial interactions, including archaeal methanogens, may be important targets and predictors but a longitudinal approach is needed. DESIGN: Cirrhosis outpatients who provided stool twice were included. Group 1: compensated, group 2: 1 decompensation (decomp), group 3: >1 decompensationwere followed and divided into those who remained stable or decompensated. Bacteria, viral and archaeal presence, α/ß diversity and taxa changes over time adjusted for clinical variables were analysed. Correlation networks between kingdoms were analysed. RESULTS: 157 outpatients (72 group 1, 33 group 2 and 52 group 3) were followed and 28%-47% developed outcomes. Baseline between those who remained stable/developed outcome: While no α/ß diversity differences were seen, commensals were lower and pathobionts were higher in those who decompensated. After decompensation: those experiencing their first decompensation showed greater decrease in α/ß-diversity, bacterial change (↑Lactobacillus spp, Streptococcus parasanguinis and ↓ beneficial Lachnospiraceae and Eubacterium hallii) and viral change (↑Siphoviridae, ↓ Myoviridae) versus those with further decompensation. Archaea: 19% had Methanobacter brevii, which was similar between/within groups. Correlation networks: Baseline archaeal-viral-bacterial networks were denser and more homogeneous in those who decompensated versus the rest. Archaea-bacterial correlations collapsed post first decompensation. Lactobacillus phage Lc Nu and C2-like viruses were negatively linked with beneficial bacteria. CONCLUSION: In this longitudinal study of cirrhosis outpatients, the greatest transkingdom gut microbial changes were seen in those reaching the first decompensation, compared with subsequent decompensating events. A transkingdom approach may refine prediction and provide therapeutic targets to prevent cirrhosis progression.


Asunto(s)
Bacteriófagos , Microbioma Gastrointestinal , Humanos , Estudios Longitudinales , Pacientes Ambulatorios , Cirrosis Hepática , Lactobacillus
5.
Nat Microbiol ; 7(6): 780-795, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35577971

RESUMEN

While gut microbiome and host gene regulation independently contribute to gastrointestinal disorders, it is unclear how the two may interact to influence host pathophysiology. Here we developed a machine learning-based framework to jointly analyse paired host transcriptomic (n = 208) and gut microbiome (n = 208) profiles from colonic mucosal samples of patients with colorectal cancer, inflammatory bowel disease and irritable bowel syndrome. We identified associations between gut microbes and host genes that depict shared as well as disease-specific patterns. We found that a common set of host genes and pathways implicated in gastrointestinal inflammation, gut barrier protection and energy metabolism are associated with disease-specific gut microbes. Additionally, we also found that mucosal gut microbes that have been implicated in all three diseases, such as Streptococcus, are associated with different host pathways in each disease, suggesting that similar microbes can affect host pathophysiology in a disease-specific manner through regulation of different host genes. Our framework can be applied to other diseases for the identification of host gene-microbiome associations that may influence disease outcomes.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Microbiota , Colon/metabolismo , Microbioma Gastrointestinal/genética , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , Microbiota/genética
6.
Gastroenterology ; 161(4): 1194-1207.e8, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34245762

RESUMEN

BACKGROUND & AIMS: The gut virome includes eukaryotic viruses and bacteriophages that can shape the gut bacterial community and elicit host responses. The virome can be implicated in diseases, such as irritable bowel syndrome (IBS), where gut bacteria play an important role in pathogenesis. We provide a comprehensive and longitudinal characterization of the virome, including DNA and RNA viruses and paired multi-omics data in a cohort of healthy subjects and patients with IBS. METHODS: We selected 2 consecutive stool samples per subject from a longitudinal study cohort and performed metagenomic sequencing on DNA and RNA viruses after enriching for viral-like particles. Viral sequence abundance was evaluated over time, as well as in the context of diet, bacterial composition and function, metabolite levels, colonic gene expression, host genetics, and IBS subsets. RESULTS: We found that the gut virome was temporally stable and correlated with the colonic transcriptome. We identified IBS-subset-specific changes in phage populations; Microviridae, Myoviridae, and Podoviridae species were elevated in diarrhea-predominant IBS, and other Microviridae and Myoviridae species were elevated in constipation-predominant IBS compared to healthy controls. We identified correlations between subsets of the virome and bacterial composition (unclassifiable "dark matter" and phages) and diet (eukaryotic viruses). CONCLUSIONS: We found that the gut virome is stable over time but varies among subsets of patients with IBS. It can be affected by diet and potentially influences host function via interactions with gut bacteria and/or altering host gene expression.


Asunto(s)
Dieta , Intestinos/virología , Síndrome del Colon Irritable/virología , Transcriptoma , Viroma , Virus/crecimiento & desarrollo , Adulto , Bacteriófagos/genética , Bacteriófagos/crecimiento & desarrollo , Estudios de Casos y Controles , Dieta/efectos adversos , Femenino , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Intestinos/microbiología , Síndrome del Colon Irritable/diagnóstico , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/microbiología , Estudios Longitudinales , Masculino , Metagenoma , Metagenómica , Persona de Mediana Edad , Virología , Virus/genética
7.
Gut ; 70(6): 1162-1173, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32998876

RESUMEN

OBJECTIVE: Altered bacterial composition is associated with disease progression in cirrhosis but the role of virome, especially phages, is unclear. DESIGN: Cross-sectional and pre/post rifaximin cohorts were enrolled. Cross-sectional: controls and cirrhotic outpatients (compensated, on lactulose (Cirr-L), on rifaximin (Cirr-LR)) were included and followed for 90-day hospitalisations. Pre/post: compensated cirrhotics underwent stool collection pre/post 8 weeks of rifaximin. Stool metagenomics for bacteria and phages and their correlation networks were analysed in controls versus cirrhosis, within cirrhotics, hospitalised/not and pre/post rifaximin. RESULTS: Cross-sectional: 40 controls and 163 cirrhotics (63 compensated, 43 Cirr-L, 57 Cirr-LR) were enrolled. Cirr-L/LR groups were similar on model for end-stage liver disease (MELD) score but Cirr-L developed greater hospitalisations versus Cirr-LR (56% vs 30%, p=0.008). Bacterial alpha/beta diversity worsened from controls through Cirr-LR. While phage alpha diversity was similar, beta diversity was different between groups. Autochthonous bacteria linked negatively, pathobionts linked positively with MELD but only modest phage-MELD correlations were seen. Phage-bacterial correlation network complexity was highest in controls, lowest in Cirr-L and increased in Cirr-LR. Microviridae and Faecalibacterium phages were linked with autochthonous bacteria in Cirr-LR, but not Cirr-L hospitalised patients had greater pathobionts, lower commensal bacteria and phages focused on Streptococcus, Lactococcus and Myoviridae. Pre/post: No changes in alpha/beta diversity of phages or bacteria were seen postrifaximin. Phage-bacterial linkages centred around urease-producing Streptococcus species collapsed postrifaximin. CONCLUSION: Unlike bacteria, faecal phages are sparsely linked with cirrhosis characteristics and 90-day outcomes. Phage and bacterial linkages centred on urease-producing, ammonia-generating Streptococcus species were affected by disease progression and rifaximin therapy and were altered in patients who experienced 90-day hospitalisations.


Asunto(s)
Antibacterianos/uso terapéutico , Enfermedad Hepática en Estado Terminal/microbiología , Firmicutes/virología , Encefalopatía Hepática/microbiología , Cirrosis Hepática/microbiología , Rifaximina/uso terapéutico , Anciano , Antibacterianos/farmacología , Estudios Transversales , Progresión de la Enfermedad , Enfermedad Hepática en Estado Terminal/etiología , Faecalibacterium/genética , Faecalibacterium/virología , Heces/microbiología , Femenino , Firmicutes/genética , Fármacos Gastrointestinales/uso terapéutico , Hospitalización , Humanos , Lactococcus/genética , Lactococcus/virología , Lactulosa/uso terapéutico , Cirrosis Hepática/complicaciones , Cirrosis Hepática/tratamiento farmacológico , Masculino , Metagenoma/efectos de los fármacos , Metagenómica , Interacciones Microbianas , Microviridae/genética , Persona de Mediana Edad , Myoviridae/genética , Gravedad del Paciente , Rifaximina/farmacología , Streptococcus/genética , Streptococcus/virología , Viroma/efectos de los fármacos
9.
Genes (Basel) ; 11(11)2020 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-33233349

RESUMEN

Sequencing of the 16S rRNA gene (16S) has long been a go-to method for microbiome characterization due to its accessibility and lower cost compared to shotgun metagenomic sequencing (SMS). However, 16S sequencing rarely provides species-level resolution and cannot provide direct assessment of other taxa (e.g., viruses and fungi) or functional gene content. Shallow shotgun metagenomic sequencing (SSMS) has emerged as an approach to bridge the gap between 16S sequencing and deep metagenomic sequencing. SSMS is cost-competitive with 16S sequencing, while also providing species-level resolution and functional gene content insights. In the present study, we evaluated the effects of sequencing depth on marker gene-mapping- and alignment-based annotation of bacteria in healthy human stool samples. The number of identified taxa decreased with lower sequencing depths, particularly with the marker gene-mapping-based approach. Other annotations, including viruses and pathways, also showed a depth-dependent effect on feature recovery. These results refine the understanding of the suitability and shortcomings of SSMS, as well as annotation tools for metagenomic analyses in human stool samples. Results may also translate to other sample types and may open the opportunity to explore the effect of sequencing depth and annotation method.


Asunto(s)
Heces/microbiología , Microbioma Gastrointestinal/genética , Metagenómica/métodos , Virus/genética , Bacterias/genética , Marcadores Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Metagenoma , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN/métodos
10.
Cell ; 182(6): 1460-1473.e17, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32916129

RESUMEN

The gut microbiome has been implicated in multiple human chronic gastrointestinal (GI) disorders. Determining its mechanistic role in disease has been difficult due to apparent disconnects between animal and human studies and lack of an integrated multi-omics view of disease-specific physiological changes. We integrated longitudinal multi-omics data from the gut microbiome, metabolome, host epigenome, and transcriptome in the context of irritable bowel syndrome (IBS) host physiology. We identified IBS subtype-specific and symptom-related variation in microbial composition and function. A subset of identified changes in microbial metabolites correspond to host physiological mechanisms that are relevant to IBS. By integrating multiple data layers, we identified purine metabolism as a novel host-microbial metabolic pathway in IBS with translational potential. Our study highlights the importance of longitudinal sampling and integrating complementary multi-omics data to identify functional mechanisms that can serve as therapeutic targets in a comprehensive treatment strategy for chronic GI diseases. VIDEO ABSTRACT.


Asunto(s)
Microbioma Gastrointestinal/genética , Regulación de la Expresión Génica/genética , Síndrome del Colon Irritable/metabolismo , Metaboloma , Purinas/metabolismo , Transcriptoma/genética , Animales , Ácidos y Sales Biliares/metabolismo , Biopsia , Butiratos/metabolismo , Cromatografía Liquida , Estudios Transversales , Epigenómica , Heces/microbiología , Femenino , Microbioma Gastrointestinal/fisiología , Regulación de la Expresión Génica/fisiología , Interacciones Microbiota-Huesped/genética , Humanos , Hipoxantina/metabolismo , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/microbiología , Estudios Longitudinales , Masculino , Metaboloma/fisiología , Ratones , Estudios Observacionales como Asunto , Estudios Prospectivos , Programas Informáticos , Espectrometría de Masas en Tándem , Transcriptoma/fisiología
11.
Anim Microbiome ; 2(1): 16, 2020 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-33499991

RESUMEN

BACKGROUND: The gut microbiome harbors trillions of bacteria that play a major role in dietary nutrient extraction and host metabolism. Metabolic diseases such as obesity and diabetes are associated with shifts in microbiome composition and have been on the rise in Westernized or highly industrialized countries. At the same time, Westernized diets low in dietary fiber have been shown to cause loss of gut microbial diversity. However, the link between microbiome composition, loss of dietary fiber, and obesity has not been well defined. RESULTS: To study the interactions between gut microbiota, dietary fiber, and weight gain, we transplanted captive and wild douc gut microbiota into germ-free mice and then exposed them to either a high- or low-fiber diet. The group receiving captive douc microbiota gained significantly more weight, regardless of diet, while mice receiving a high-fiber diet and wild douc microbiota remained lean. In the presence of a low-fiber diet, the wild douc microbiota partially prevented weight gain. Using 16S rRNA gene amplicon sequencing we identified key bacterial taxa in each group, specifically a high relative abundance of Bacteroides and Akkermansia in captive douc FMT mice and a higher relative abundance of Lactobacillus and Clostridium in the wild douc FMT mice. CONCLUSIONS: In the context of our germ-free mouse experiment, wild douc microbiota could serve as a reservoir for microbes for cross-species transplants. Our results suggest that wild douc microbiota are tailored to diverse fiber diets and can prevent weight gain when exposed to a native diet.

12.
mBio ; 10(5)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31615957

RESUMEN

The microbiome is important to all animals, including poultry, playing a critical role in health and performance. Low-dose antibiotics have historically been used to modulate food production animals and their microbiome. Identifying alternatives to antibiotics conferring similar modulatory properties has been elusive. The purpose of this study was to determine if a host-tailored probiotic could recapitulate effects of a low-dose antibiotic on host response and the developing microbiome. Over 13 days of life, turkey poults were supplemented continuously with a low-dose antibiotic or oral supplementation of a prebiotic with or without two different probiotics (8 cage units, n = 80 per group). Gastrointestinal bacterial and fungal communities of poults were characterized by 16S rRNA gene and ITS2 amplicon sequencing. Localized and systemic host gene expression was assessed using transcriptome sequencing (RNA-Seq), kinase activity was assessed by avian-specific kinome peptide arrays, and performance parameters were assessed. We found that development of the early-life microbiome of turkey poults was tightly ordered in a tissue- and time-specific manner. Low-dose antibiotic and turkey-tailored probiotic supplementation, but not nontailored probiotic supplementation, elicited similar shifts in overall microbiome composition during development compared to controls. Treatment-induced bacterial changes were accompanied by parallel shifts in the fungal community and host gene expression and enhanced performance metrics. These results were validated in pen trials that identified further additive effects of the turkey-tailored probiotic combined with different prebiotics. Alternative approaches to low-dose antibiotic use in poultry are feasible and can be optimized utilizing the indigenous poultry microbiome. Similar approaches may also be beneficial for humans.IMPORTANCE Alternative approaches are greatly needed to reduce the need for antibiotic use in food animal production. This study utilized a pipeline for the development of a host-tailored probiotic to enhance performance in commercial turkeys and modulate their microbiota, similar to the effects of low-dose antibiotic administration. We determined that a host-tailored probiotic, developed in the context of the commercial turkey gut microbiome, was more effective at modulating these parameters than a nontailored probiotic cocktail. Furthermore, the host-tailored probiotic mimicked many of the effects of a low-dose antibiotic growth promoter. Surprisingly, the effects of the antibiotic growth promoter and host-tailored probiotic were observed across kingdoms, illustrating the coordinated interkingdom effects of these approaches. This work suggests that tailored approaches to probiotic development hold promise for modulating the avian host and its microbiota.


Asunto(s)
Antibacterianos/farmacología , Probióticos , Animales , Microbiota/efectos de los fármacos , Micobioma/efectos de los fármacos , ARN Ribosómico 16S/genética , Pavos
13.
Cell Host Microbe ; 25(6): 789-802.e5, 2019 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-31194939

RESUMEN

Diet is a key determinant of human gut microbiome variation. However, the fine-scale relationships between daily food choices and human gut microbiome composition remain unexplored. Here, we used multivariate methods to integrate 24-h food records and fecal shotgun metagenomes from 34 healthy human subjects collected daily over 17 days. Microbiome composition depended on multiple days of dietary history and was more strongly associated with food choices than with conventional nutrient profiles, and daily microbial responses to diet were highly personalized. Data from two subjects consuming only meal replacement beverages suggest that a monotonous diet does not induce microbiome stability in humans, and instead, overall dietary diversity associates with microbiome stability. Our work provides key methodological insights for future diet-microbiome studies and suggests that food-based interventions seeking to modulate the gut microbiota may need to be tailored to the individual microbiome. Trial Registration: ClinicalTrials.gov: NCT03610477.


Asunto(s)
Dieta , Microbioma Gastrointestinal , Microbiota , Adulto , Heces/microbiología , Femenino , Humanos , Estudios Longitudinales , Masculino , Metagenómica , Persona de Mediana Edad , Adulto Joven
14.
Cell ; 175(4): 962-972.e10, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388453

RESUMEN

Many US immigrant populations develop metabolic diseases post immigration, but the causes are not well understood. Although the microbiome plays a role in metabolic disease, there have been no studies measuring the effects of US immigration on the gut microbiome. We collected stool, dietary recalls, and anthropometrics from 514 Hmong and Karen individuals living in Thailand and the United States, including first- and second-generation immigrants and 19 Karen individuals sampled before and after immigration, as well as from 36 US-born European American individuals. Using 16S and deep shotgun metagenomic DNA sequencing, we found that migration from a non-Western country to the United States is associated with immediate loss of gut microbiome diversity and function in which US-associated strains and functions displace native strains and functions. These effects increase with duration of US residence and are compounded by obesity and across generations.


Asunto(s)
Pueblo Asiatico , Emigración e Inmigración , Microbioma Gastrointestinal , Adulto , Bacteroides/aislamiento & purificación , Fibras de la Dieta/metabolismo , Emigrantes e Inmigrantes , Humanos , Metagenoma , Obesidad/epidemiología , Obesidad/microbiología , Prevotella/aislamiento & purificación , Estados Unidos
15.
Sci Rep ; 8(1): 11159, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30042392

RESUMEN

Red-shanked doucs (Pygathrix nemaeus) are endangered, foregut-fermenting colobine primates which are difficult to maintain in captivity. There are critical gaps in our understanding of their natural lifestyle, including dietary habits such as consumption of leaves, unripe fruit, flowers, seeds, and other plant parts. There is also a lack of understanding of enteric adaptations, including their unique microflora. To address these knowledge gaps, we used the douc as a model to study relationships between gastrointestinal microbial community structure and lifestyle. We analyzed published fecal samples as well as detailed dietary history from doucs with four distinct lifestyles (wild, semi-wild, semi-captive, and captive) and determined gastrointestinal bacterial microbiome composition using 16S rRNA sequencing. A clear gradient of microbiome composition was revealed along an axis of natural lifestyle disruption, including significant associations with diet, biodiversity, and microbial function. We also identified potential microbial biomarkers of douc dysbiosis, including Bacteroides and Prevotella, which may be related to health. Our results suggest a gradient-like shift in captivity causes an attendant shift to severe gut dysbiosis, thereby resulting in gastrointestinal issues.


Asunto(s)
Cercopithecidae/fisiología , Microbioma Gastrointestinal/genética , Estado de Salud , Estado Nutricional/fisiología , Animales , Bacteroidetes/clasificación , Bacteroidetes/genética , Biodiversidad , Cloroplastos/genética , Dieta Vegana , Disbiosis , Especies en Peligro de Extinción , Heces/microbiología , Firmicutes/clasificación , Firmicutes/genética , Tracto Gastrointestinal/microbiología , Estilo de Vida , Metagenoma , Modelos Animales , ARN Ribosómico 16S/genética , Análisis de Secuencia de ARN , Estadísticas no Paramétricas
16.
Sci Rep ; 8(1): 6219, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29670191

RESUMEN

Fecal microbiota transplantation (FMT) is now widely used to treat recurrent Clostridium difficile infection, but has been less studied as a means to restore microbiome diversity and composition following antibiotic or chemotherapy treatments. The purpose of our study was to assess the efficacy of FMT to reverse antibiotic- and chemotherapy-induced gut dysbiosis in a mouse model. C57BL/6J mice were treated with ampicillin for 1 week and/or received a single intraperitoneal injection of 5-Fluorouracil. Fresh stool was collected and analyzed using shotgun metagenomics and the Illumina sequencing platform. Ampicillin caused a significant and immediate decrease in bacterial species richness and diversity that persisted for one week. In mice that received FMT, disruption of the intestinal microbiota was reversed immediately. Antibiotic and chemotherapy administration caused significant alteration in species distribution, including a decrease in the relative proportions of Clostridium scindens and Faecalibacterium prausnitzii, and an increase in known pathogenic species. In mice receiving FMT, we observed a significant increase in species known to exhibit anti-inflammatory properties. Moreover, chemotherapy led to a critical decrease in key 'health-promoting' species and to an altered functional profile, especially when chemotherapy was administered in tandem with antibiotics, and that FMT can ameliorate these effects.


Asunto(s)
Antibacterianos/efectos adversos , Antineoplásicos/efectos adversos , Disbiosis/etiología , Disbiosis/microbiología , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Animales , Antibacterianos/farmacología , Antineoplásicos/farmacología , Biodiversidad , Modelos Animales de Enfermedad , Disbiosis/terapia , Heces/microbiología , Metagenoma , Metagenómica/métodos , Ratones
17.
mSystems ; 3(3)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29546248

RESUMEN

With the advent of next-generation sequencing and microbial community characterization, we are beginning to understand the key factors that shape early-life microbial colonization and associated health outcomes. Studies characterizing infant microbial colonization have focused mostly on bacteria in the microbiome and have largely neglected fungi (the mycobiome), despite their relevance to mucosal infections in healthy infants. In this pilot study, we characterized the skin, oral, and anal mycobiomes of infants over the first month of life (n = 17) and the anal and vaginal mycobiomes of mothers (n = 16) by internal transcribed spacer 2 (ITS2) amplicon sequencing. We found that infant mycobiomes differed by body site, with the infant mycobiomes at the anal sites being different from those at the skin and oral sites. The relative abundances of body site-specific taxa differed by birth mode, with significantly more Candida albicans fungi present on the skin of vaginally born infants on day 30 and significantly more Candida orthopsilosis fungi present in the oral cavity of caesarean section-born infants throughout the first month of life. We found the mycobiomes within individual infants to be variable over the first month of life, and vaginal birth did not result in infant mycobiomes that were more similar to the mother's vaginal mycobiome. Therefore, although vertical transmission of specific fungal isolates from mother to infant has been reported, it is likely that other sources (environment, other caregivers) also contribute to early-life mycobiome establishment. Thus, future longitudinal studies of mycobiome and bacterial microbiome codevelopment, with dense sampling from birth to beyond the first month of life, are warranted. IMPORTANCE Humans are colonized by diverse fungi (mycobiome), which have received much less study to date than colonizing bacteria. We know very little about the succession of fungal colonization in early life and whether it may relate to long-term health. To better understand fungal colonization and its sources, we studied the skin, oral, and anal mycobiomes of healthy term infants and the vaginal and anal mycobiomes of their mothers. Generally, infants were colonized by few fungal taxa, and fungal alpha diversity did not increase over the first month of life. There was no clear community maturation over the first month of life, regardless of body site. Key body-site-specific taxa, but not overall fungal community structures, were impacted by birth mode. Thus, additional studies to characterize mycobiome acquisition and succession throughout early life are needed to form a foundation for research into the relationship between mycobiome development and human disease.

18.
Sci Rep ; 8(1): 2857, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29434295

RESUMEN

Salmonella enterica serovar Typhimurium continues to be a major cause of foodborne illness worldwide and pork can serve as a source of infection. Co-infection of S. enterica with Lawsonia intracellularis, a common intestinal pathogen of swine, has been found as risk factor for increased S. enterica shedding. The objective of this study was to investigate if vaccination against L. intracellularis could lead to decreased S. Typhimurium shedding. To test this hypothesis, pigs were challenged with either S. Typhimurium or S. Typhimurium and L. intracellularis, with and without L. intracellularis vaccination (n = 9 per group). A non-challenged group served as a negative control. Vaccination decreased the shedding of S. Typhimurium in co-infected animals by 2.12 log10 organisms per gram of feces at 7 days post infection. Analysis of the microbiome showed that vaccination led to changes in the abundance of Clostridium species, including Clostridium butyricum, in addition to other compositional changes that may explain the protection mediated against S. Typhimurium. These results indicate that vaccination against L. intracellularis in co-infected herds may provide a new tool to increase food safety by helping to prevent S. enterica without the need for antibiotics.


Asunto(s)
Derrame de Bacterias/efectos de los fármacos , Vacunas Bacterianas/administración & dosificación , Infecciones por Desulfovibrionaceae/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Salmonelosis Animal/prevención & control , Salmonella typhimurium/fisiología , Enfermedades de los Porcinos/microbiología , Animales , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/aislamiento & purificación , Vacunas Bacterianas/farmacología , Coinfección/prevención & control , Infecciones por Desulfovibrionaceae/inmunología , Infecciones por Desulfovibrionaceae/veterinaria , Heces/microbiología , Microbiología de Alimentos , Inocuidad de los Alimentos , Lawsonia (Bacteria)/efectos de los fármacos , Lawsonia (Bacteria)/inmunología , Filogenia , Salmonelosis Animal/inmunología , Salmonella typhimurium/efectos de los fármacos , Porcinos , Enfermedades de los Porcinos/prevención & control , Vacunación/veterinaria
19.
Nat Microbiol ; 3(2): 234-242, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29180726

RESUMEN

Antibiotic exposure in children has been associated with the risk of inflammatory bowel disease (IBD). Antibiotic use in children or in their pregnant mother can affect how the intestinal microbiome develops, so we asked whether the transfer of an antibiotic-perturbed microbiota from mothers to their children could affect their risk of developing IBD. Here we demonstrate that germ-free adult pregnant mice inoculated with a gut microbial community shaped by antibiotic exposure transmitted their perturbed microbiota to their offspring with high fidelity. Without any direct or continued exposure to antibiotics, this dysbiotic microbiota in the offspring remained distinct from controls for at least 21 weeks. By using both IL-10-deficient and wild-type mothers, we showed that both inoculum and genotype shape microbiota populations in the offspring. Because IL10-/- mice are genetically susceptible to colitis, we could assess the risk due to maternal transmission of an antibiotic-perturbed microbiota. We found that the IL10-/- offspring that had received the perturbed gut microbiota developed markedly increased colitis. Taken together, our findings indicate that antibiotic exposure shaping the maternal gut microbiota has effects that extend to the offspring, with both ecological and long-term disease consequences.


Asunto(s)
Antibacterianos/administración & dosificación , Colitis/microbiología , Susceptibilidad a Enfermedades/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/microbiología , Animales , Colitis/inducido químicamente , Colon/inmunología , Colon/microbiología , Colon/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Disbiosis/microbiología , Heces/microbiología , Femenino , Enfermedades Inflamatorias del Intestino/inducido químicamente , Interleucina-10 , Metagenoma/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Fenotipo , Embarazo
20.
PeerJ ; 5: e3075, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28289569

RESUMEN

It has previously been shown that domestic dogs and their household owners share bacterial populations, and that sharing of bacteria between humans is facilitated through the presence of dogs in the household. However, less is known regarding the bacterial communities of dogs, how these communities vary by location and over time, and how cohabitation of dogs themselves influences their bacterial community. Furthermore, the effects of factors such as breed, hair coat length, sex, shedding, and age on the canine skin microbiome is unknown. This study sampled the skin bacterial communities of 40 dogs belonging to 20 households longitudinally across three seasons (spring, summer, and winter). Significant differences in bacterial community structure between samples were identified when stratified by season, but not by dog sex, age, breed, hair type, or skin site. Cohabitating dogs were more likely to share bacteria of the skin than non-cohabitating dogs. Similar to human bacterial microbiomes, dogs' microbiomes were more similar to their own microbiomes over time than to microbiomes of other individuals. Dogs sampled during the same season were also more similar to each other than to dogs from different seasons, irrespective of household. However, there were very few core operational taxonomic units (OTUs) identified across all dogs sampled. Taxonomic classification revealed Propionibacterium acnes and Haemophilus sp. as key members of the dog skin bacterial community, along with Corynebacterium sp. and Staphylococcus epidermidis. This study shows that the skin bacterial community structure of dogs is highly individualized, but can be shared among dogs through cohabitation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...