Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Nature ; 613(7945): 639-649, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36697862

RESUMEN

Whether the human fetus and the prenatal intrauterine environment (amniotic fluid and placenta) are stably colonized by microbial communities in a healthy pregnancy remains a subject of debate. Here we evaluate recent studies that characterized microbial populations in human fetuses from the perspectives of reproductive biology, microbial ecology, bioinformatics, immunology, clinical microbiology and gnotobiology, and assess possible mechanisms by which the fetus might interact with microorganisms. Our analysis indicates that the detected microbial signals are likely the result of contamination during the clinical procedures to obtain fetal samples or during DNA extraction and DNA sequencing. Furthermore, the existence of live and replicating microbial populations in healthy fetal tissues is not compatible with fundamental concepts of immunology, clinical microbiology and the derivation of germ-free mammals. These conclusions are important to our understanding of human immune development and illustrate common pitfalls in the microbial analyses of many other low-biomass environments. The pursuit of a fetal microbiome serves as a cautionary example of the challenges of sequence-based microbiome studies when biomass is low or absent, and emphasizes the need for a trans-disciplinary approach that goes beyond contamination controls by also incorporating biological, ecological and mechanistic concepts.


Asunto(s)
Biomasa , Contaminación de ADN , Feto , Microbiota , Animales , Femenino , Humanos , Embarazo , Líquido Amniótico/inmunología , Líquido Amniótico/microbiología , Mamíferos , Microbiota/genética , Placenta/inmunología , Placenta/microbiología , Feto/inmunología , Feto/microbiología , Reproducibilidad de los Resultados
2.
Proc Natl Acad Sci U S A ; 120(52): e2306863120, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38127978

RESUMEN

The gut microbiota is a considerable source of biologically active compounds that can promote intestinal homeostasis and improve immune responses. Here, we used large expression libraries of cloned metagenomic DNA to identify compounds able to sustain an anti-inflammatory reaction on host cells. Starting with a screen for NF-κB activation, we have identified overlapping clones harbouring a heterodimeric ATP-binding cassette (ABC)-transporter from a Firmicutes. Extensive purification of the clone's supernatant demonstrates that the ABC-transporter allows for the efficient extracellular accumulation of three muropeptide precursor, with anti-inflammatory properties. They induce IL-10 secretion from human monocyte-derived dendritic cells and proved effective in reducing AIEC LF82 epithelial damage and IL-8 secretion in human intestinal resections. In addition, treatment with supernatants containing the muropeptide precursor reduces body weight loss and improves histological parameters in Dextran Sulfate Sodium (DSS)-treated mice. Until now, the source of peptidoglycan fragments was shown to come from the natural turnover of the peptidoglycan layer by endogenous peptidoglycan hydrolases. This is a report showing an ABC-transporter as a natural source of secreted muropeptide precursor and as an indirect player in epithelial barrier strengthening. The mechanism described here might represent an important component of the host immune homeostasis.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Humanos , Ratones , Animales , Peptidoglicano/metabolismo , Intestinos/patología , Inflamación/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Antiinflamatorios/metabolismo , Sulfato de Dextran , Colitis/metabolismo , Modelos Animales de Enfermedad , Colon/metabolismo , Ratones Endogámicos C57BL
3.
Int J Mol Sci ; 24(24)2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38139456

RESUMEN

Our understanding of the symbiotic relationship between the microbiota and its host has constantly evolved since our understanding that the "self" was not only defined by our genetic patrimony but also by the genomes of bugs living in us. The first culture-based methods highlighted the important functions of the microbiota. However, these methods had strong limitations and did not allow for a full understanding of the complex relationships that occur at the interface between the microbiota and the host. The recent development of metagenomic approaches has been a groundbreaking step towards this understanding. Its use has provided new insights and perspectives. In the present chapter, we will describe the advances of functional metagenomics to decipher food-microbiota and host-microbiota interactions. This powerful high-throughput approach allows for the assessment of the microbiota as a whole (including non-cultured bacteria) and enabled the discovery of new signaling pathways and functions involved in the crosstalk between food, the gut microbiota and its host. We will present the pipeline and highlight the most important studies that helped to develop the field. To conclude, we will emphasize the most recent developments and hot topics in functional metagenomics.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Humanos , Interacciones Microbiota-Huesped , Metagenómica/métodos , Metagenoma
4.
Int J Mol Sci ; 24(15)2023 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-37569608

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) affects about 20-40% of the adult population in high-income countries and is now a leading indication for liver transplantation and can lead to hepatocellular carcinoma. The link between gut microbiota dysbiosis and NAFLD is now clearly established. Through analyses of the gut microbiota with shotgun metagenomics, we observe that compared to healthy controls, Adlercreutzia equolifaciens is depleted in patients with liver diseases such as NAFLD. Its abundance also decreases as the disease progresses and eventually disappears in the last stages indicating a strong association with disease severity. Moreover, we show that A. equolifaciens possesses anti-inflammatory properties, both in vitro and in vivo in a humanized mouse model of NAFLD. Therefore, our results demonstrate a link between NAFLD and the severity of liver disease and the presence of A. equolifaciens and its anti-inflammatory actions. Counterbalancing dysbiosis with this bacterium may be a promising live biotherapeutic strategy for liver diseases.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias Hepáticas , Enfermedades Metabólicas , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Disbiosis/microbiología , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Neoplasias Hepáticas/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antiinflamatorios/metabolismo
5.
Eur Eat Disord Rev ; 31(1): 76-86, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35751889

RESUMEN

OBJECTIVE: Previous studies of AN showed low-grade inflammation. Are low-grade inflammation and circulating lymphocytes associated with chronic conditions? METHOD: Peripheric blood cytokines were measured using Luminex™ technology in a chronic AN cohort (mean = 67.42 months), compared to Constitutional Thinness (CT), Constitutional Obesity (CO), and Healthy Controls (HC). Secondarily a prospective cohort of chronic AN (mean = 54.11 months) was recruited to compare the functional lymphocyte profile in blood by flow cytometry to CT and HC. RESULTS: In the AN group, most cytokine concentrations were lower than in CT and HC groups. The IL-23 (98.02 pg/ml) was elevated related to HC and CO, and the IL-10 (4.178 pg/ml) was elevated versus CO. In the CT group, IL-9 (0.06216 pg/ml) was elevated compared to AN. The AN group had high Treg (9.259% of CD4+ ) and CD8+ Integrinß7+ (9.552% of CD3+ ) versus HC for lymphocyte populations. In CT group, elevated Treg (9.7% of CD4+ ) elevated percentage of CD4+ CCR9+ (5.867% of CD3+ ) and CD8+ Integrinß7+ (10.21% of CD3+ ) were found versus HC. CONCLUSIONS: The chronic state of AN and CT is surprisingly non-inflammatory with elevated Treg cells. These results suggest that maintaining a dysregulated response to intestinal antigens may contribute to maintaining AN.


Asunto(s)
Linfocitos T , Humanos , Estudios Prospectivos
6.
Gastroenterology ; 161(6): 1969-1981.e12, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34508776

RESUMEN

BACKGROUND & AIMS: Intestinal microbiota-host interactions play a major role in health and disease. This has been documented at the microbiota level ("dysbiosis" in chronic immune-mediated diseases) and through the study of specific bacteria-host interactions but rarely at the level of intestinal ecosystem dynamics. However, understanding the behavior of this ecosystem may be key to the successful treatment of disease. We recently postulated that health and disease represent alternative stable states of the intestinal ecosystem (different configurations that can exist under identical external conditions), which would require adapted strategies in disease treatment. Here, we examine if alternative stable states indeed exist in this ecosystem and if they could affect remission from ulcerative colitis (UC). METHODS: We analyzed data from a study on pediatric UC. The data reflect current treatment practice following the recruitment of treatment-naive patients with new-onset disease. Patients received personalized anti-inflammatory treatments over a period of 1 year. Stool samples at 0, 4, 12, and 52 weeks allowed an estimation of microbiota status (through 16S ribosomal RNA gene sequencing) and host inflammatory status (through the measurement of fecal calprotectin levels). RESULTS: We identify 4 microbiota states and 4 host states. Longitudinal data show that the improvement of inflammatory status is accompanied by an improvement of microbiota status. However, they also provide strong indications that both improvements are retarded or blocked by alternative states barriers. CONCLUSIONS: Our observations strongly suggest that inflammation suppression should be combined with microbiota management where possible to improve the efficacy of UC treatment.


Asunto(s)
Antiinflamatorios/uso terapéutico , Bacterias/efectos de los fármacos , Microambiente Celular , Colitis Ulcerosa/terapia , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Mediadores de Inflamación/antagonistas & inhibidores , Intestinos/efectos de los fármacos , Antiinflamatorios/efectos adversos , Bacterias/genética , Bacterias/inmunología , Estudios de Casos y Controles , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/microbiología , Terapia Combinada , Disbiosis , Trasplante de Microbiota Fecal/efectos adversos , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Intestinos/metabolismo , Intestinos/microbiología , Modelos Biológicos , Inducción de Remisión , Factores de Tiempo , Resultado del Tratamiento
7.
Gastroenterology ; 160(1): 206-218.e13, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32941879

RESUMEN

BACKGROUND AND AIMS: Cirrhosis is associated with changes in gut microbiome composition. Although acute-on-chronic liver failure (ACLF) is the most severe clinical stage of cirrhosis, there is lack of information about gut microbiome alterations in ACLF using quantitative metagenomics. We investigated the gut microbiome in patients with cirrhosis encompassing the whole spectrum of disease (compensated, acutely decompensated without ACLF, and ACLF). A group of healthy subjects was used as control subjects. METHODS: Stool samples were collected prospectively in 182 patients with cirrhosis. DNA library construction and sequencing were performed using the Ion Proton Sequencer (ThermoFisher Scientific, Waltham, MA). Microbial genes were grouped into clusters, denoted as metagenomic species. RESULTS: Cirrhosis was associated with a remarkable reduction in gene and metagenomic species richness compared with healthy subjects. This loss of richness correlated with disease stages and was particularly marked in patients with ACLF and persisted after adjustment for antibiotic therapy. ACLF was associated with a significant increase of Enterococcus and Peptostreptococcus sp and a reduction of some autochthonous bacteria. Gut microbiome alterations correlated with model for end-stage liver disease and Child-Pugh scores and organ failure and was associated with some complications, particularly hepatic encephalopathy and infections. Interestingly, gut microbiome predicted 3-month survival with good stable predictors. Functional analysis showed that patients with cirrhosis had enriched pathways related to ethanol production, γ-aminobutyric acid metabolism, and endotoxin biosynthesis, among others. CONCLUSIONS: Cirrhosis is characterized by marked alterations in gut microbiome that parallel disease stages with maximal changes in ACLF. Altered gut microbiome was associated with complications of cirrhosis and survival. Gut microbiome may contribute to disease progression and poor prognosis. These results should be confirmed in future studies.


Asunto(s)
Insuficiencia Hepática Crónica Agudizada/etiología , Insuficiencia Hepática Crónica Agudizada/patología , Microbioma Gastrointestinal/fisiología , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Insuficiencia Hepática Crónica Agudizada/mortalidad , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Cirrosis Hepática/mortalidad , Masculino , Metagenómica , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia
8.
BMC Microbiol ; 22(1): 54, 2022 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-35151268

RESUMEN

BACKGROUND: Diosmectite, a natural colloidal clay, has been used worldwide for a number of approved indications, including the treatment of chronic functional diarrhea. Here, we used high-resolution whole metagenome shotgun sequencing to assess the impact of a 5 weeks administration of diosmectite (3 g/sachet, 3 sachets/day) on the fecal microbiota of 35 adults with functional chronic diarrhea. RESULTS: Gut microbiota was not impacted by diosmectite administration. In particular, richness remained stable and no microbial species displayed a significant evolution. Segregating patients either by diosmectite response (non responder, early responder, late responder) or by nationality (Great-Britain or Netherlands) yielded the same results. CONCLUSION: We concluded that no microbiota-related physiological alterations are expected upon long-term treatment with diosmectite. TRIAL REGISTRATION: Clinicaltrials.gov NCT03045926.


Asunto(s)
Diarrea/tratamiento farmacológico , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Metagenoma , Silicatos/uso terapéutico , Adolescente , Adulto , Bacterias/clasificación , Bacterias/genética , Enfermedad Crónica/terapia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
9.
Nature ; 535(7612): 376-81, 2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-27409811

RESUMEN

Insulin resistance is a forerunner state of ischaemic cardiovascular disease and type 2 diabetes. Here we show how the human gut microbiome impacts the serum metabolome and associates with insulin resistance in 277 non-diabetic Danish individuals. The serum metabolome of insulin-resistant individuals is characterized by increased levels of branched-chain amino acids (BCAAs), which correlate with a gut microbiome that has an enriched biosynthetic potential for BCAAs and is deprived of genes encoding bacterial inward transporters for these amino acids. Prevotella copri and Bacteroides vulgatus are identified as the main species driving the association between biosynthesis of BCAAs and insulin resistance, and in mice we demonstrate that P. copri can induce insulin resistance, aggravate glucose intolerance and augment circulating levels of BCAAs. Our findings suggest that microbial targets may have the potential to diminish insulin resistance and reduce the incidence of common metabolic and cardiovascular disorders.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Resistencia a la Insulina , Metaboloma , Suero/metabolismo , Aminoácidos de Cadena Ramificada/biosíntesis , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Bacteroides/fisiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/microbiología , Ayuno/sangre , Ayuno/metabolismo , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/microbiología , Humanos , Masculino , Metagenoma , Ratones , Ratones Endogámicos C57BL , Países Bajos , Prevotella/fisiología
10.
J Proteome Res ; 20(3): 1522-1534, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33528260

RESUMEN

The gut microbiota are increasingly considered as a main partner of human health. Metaproteomics enables us to move from the functional potential revealed by metagenomics to the functions actually operating in the microbiome. However, metaproteome deciphering remains challenging. In particular, confident interpretation of a myriad of MS/MS spectra can only be pursued with smart database searches. Here, we compare the interpretation of MS/MS data sets from 48 individual human gut microbiomes using three interrogation strategies of the dedicated Integrated nonredundant Gene Catalog (IGC 9.9 million genes from 1267 individual fecal samples) together with the Homo sapiens database: the classical single-step interrogation strategy and two iterative strategies (in either two or three steps) aimed at preselecting a reduced-sized, more targeted search space for the final peptide spectrum matching. Both iterative searches outperformed the single-step classical search in terms of the number of peptides and protein clusters identified and the depth of taxonomic and functional knowledge, and this was the most convincing with the three-step approach. However, iterative searches do not help in reducing variability of repeated analyses, which is inherent to the traditional data-dependent acquisition mode, but this variability did not affect the hierarchical relationship between replicates and all other samples.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Microbioma Gastrointestinal/genética , Humanos , Metagenómica , Proteómica , Espectrometría de Masas en Tándem
11.
Nature ; 528(7581): 262-266, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26633628

RESUMEN

In recent years, several associations between common chronic human disorders and altered gut microbiome composition and function have been reported. In most of these reports, treatment regimens were not controlled for and conclusions could thus be confounded by the effects of various drugs on the microbiota, which may obscure microbial causes, protective factors or diagnostically relevant signals. Our study addresses disease and drug signatures in the human gut microbiome of type 2 diabetes mellitus (T2D). Two previous quantitative gut metagenomics studies of T2D patients that were unstratified for treatment yielded divergent conclusions regarding its associated gut microbial dysbiosis. Here we show, using 784 available human gut metagenomes, how antidiabetic medication confounds these results, and analyse in detail the effects of the most widely used antidiabetic drug metformin. We provide support for microbial mediation of the therapeutic effects of metformin through short-chain fatty acid production, as well as for potential microbiota-mediated mechanisms behind known intestinal adverse effects in the form of a relative increase in abundance of Escherichia species. Controlling for metformin treatment, we report a unified signature of gut microbiome shifts in T2D with a depletion of butyrate-producing taxa. These in turn cause functional microbiome shifts, in part alleviated by metformin-induced changes. Overall, the present study emphasizes the need to disentangle gut microbiota signatures of specific human diseases from those of medication.


Asunto(s)
Diabetes Mellitus Tipo 2/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Metformina/farmacología , Biodiversidad , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Femenino , Microbioma Gastrointestinal/genética , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Masculino , Metagenoma/efectos de los fármacos , Metagenoma/fisiología , Metformina/uso terapéutico , ARN Ribosómico 16S/genética
12.
Gut ; 68(1): 70-82, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29899081

RESUMEN

OBJECTIVES: Decreased gut microbial gene richness (MGR) and compositional changes are associated with adverse metabolism in overweight or moderate obesity, but lack characterisation in severe obesity. Bariatric surgery (BS) improves metabolism and inflammation in severe obesity and is associated with gut microbiota modifications. Here, we characterised severe obesity-associated dysbiosis (ie, MGR, microbiota composition and functional characteristics) and assessed whether BS would rescue these changes. DESIGN: Sixty-one severely obese subjects, candidates for adjustable gastric banding (AGB, n=20) or Roux-en-Y-gastric bypass (RYGB, n=41), were enrolled. Twenty-four subjects were followed at 1, 3 and 12 months post-BS. Gut microbiota and serum metabolome were analysed using shotgun metagenomics and liquid chromatography mass spectrometry (LC-MS). Confirmation groups were included. RESULTS: Low gene richness (LGC) was present in 75% of patients and correlated with increased trunk-fat mass and comorbidities (type 2 diabetes, hypertension and severity). Seventy-eight metagenomic species were altered with LGC, among which 50% were associated with adverse body composition and metabolic phenotypes. Nine serum metabolites (including glutarate, 3-methoxyphenylacetic acid and L-histidine) and functional modules containing protein families involved in their metabolism were strongly associated with low MGR. BS increased MGR 1 year postsurgery, but most RYGB patients remained with low MGR 1 year post-BS, despite greater metabolic improvement than AGB patients. CONCLUSIONS: We identified major gut microbiota alterations in severe obesity, which include decreased MGR and related functional pathways linked with metabolic deteriorations. The lack of full rescue post-BS calls for additional strategies to improve the gut microbiota ecosystem and microbiome-host interactions in severe obesity. TRIAL REGISTRATION NUMBER: NCT01454232.


Asunto(s)
Cirugía Bariátrica , Disbiosis/etiología , Microbioma Gastrointestinal , Obesidad Mórbida/microbiología , Obesidad Mórbida/cirugía , Adulto , Biomarcadores/sangre , Cromatografía Liquida , Comorbilidad , Femenino , Humanos , Masculino , Espectrometría de Masas , Metagenómica , Fenotipo , Estudios Prospectivos , Factores de Riesgo
13.
Am J Physiol Endocrinol Metab ; 317(3): E446-E459, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31265324

RESUMEN

The gut bacterial species Akkermansia muciniphila is associated with a healthier clinical profile. The purpose of this study was to determine the association between A. muciniphila and glucose homeostasis in patients undergoing bariatric surgery (BS): gastric banding (GB) or Roux-en-Y gastric bypass (RYGB). This nonrandomized prospective study included 65 women with severe obesity. Longitudinal analysis included subjects for whom A. muciniphila data were available at follow-up [1, 3, and 12 mo; GB (n = 10) or RYGB (n = 11)]. Glucose homeostasis markers were measured under fasting conditions (glucose, insulin, and HbA1c) or during an oral glucose tolerance test. Fecal microbiota was analyzed using shotgun metagenomics, and A. muciniphila relative abundance was assessed with 16S rRNA quantitative PCR. A. muciniphila relative abundance was significantly lower in severe obesity [mean body mass index, 45.7 kg/m2 (SD 5.4)] than in moderate obesity [33.2 kg/m2 (SD 3.8)] but not associated with glucose homeostasis markers. A significant increase in A. muciniphila relative abundance after RYGB was not correlated with metabolic improvement. Baseline A. muciniphila abundance was correlated with bacterial gene richness and was highest in the high-richness Ruminococcaceae enterotype. A. muciniphila increased in relative abundance after BS in patients with low baseline A. muciniphila abundance, especially those with a Bacteroides type 2 enterotype classification. Although decreased in severe obesity, relative abundance of A. muciniphila was not associated with glucose homeostasis before or after BS. A certain level of A. muciniphila abundance might be required to observe a beneficial link to health. The severity of obesity and gut dysbiosis may partly explain the discrepancy with previous findings in less obese populations.


Asunto(s)
Cirugía Bariátrica , Microbioma Gastrointestinal , Obesidad Mórbida/microbiología , Obesidad Mórbida/cirugía , Verrucomicrobia , Adulto , Akkermansia , Disbiosis , Heces/microbiología , Femenino , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Estado de Salud , Homeostasis , Humanos , Resistencia a la Insulina , Estudios Longitudinales , Persona de Mediana Edad , Obesidad Mórbida/metabolismo , Estudios Prospectivos , Resultado del Tratamiento , Adulto Joven
14.
Gastroenterology ; 154(4): 1009-1023.e14, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29133078

RESUMEN

BACKGROUND & AIMS: Paneth cell dysfunction causes deficiencies in intestinal C-type lectins and antimicrobial peptides, which leads to dysbiosis of the intestinal microbiota, alters the mucosal barrier, and promotes development of inflammatory bowel diseases. We investigated whether transgenic (TG) expression of the human regenerating family member 3 alpha gene (REG3A) alters the fecal microbiota and affects development of colitis in mice. METHODS: We performed studies with C57BL/6 mice that express human regenerating family member 3 alpha (hREG3A) in hepatocytes, via the albumin gene promoter. In these mice, hREG3A travels via the bile to the intestinal lumen. Some mice were given dextran sodium sulfate (DSS) to induce colitis. Feces were collected from mice and the composition of the microbiota was analyzed by 16S ribosomal RNA sequencing. The fecal microbiome was also analyzed from mice that express only 1 copy of human REG3A transgene but were fed feces from control mice (not expressing hREG3A) as newborns. Mice expressing hREG3A were monitored for DSS-induced colitis after cohousing or feeding feces from control mice. Colitis was induced in another set of control and hREG3A-TG mice by administration of trinitrobenzene sulfonic acid; some mice were given intrarectal injections of the hREG3A protein. Colon tissues were collected from mice and analyzed by histology and immunohistochemistry to detect mucin 2, as well as by 16S ribosomal RNA fluorescence in situ hybridization, transcriptional analyses, and quantitative polymerase chain reaction. We measured levels of reactive oxygen species (ROS) in bacterial cultures and fecal microbiota using 2',7'-dichlorofluorescein diacetate and flow cytometry. RESULTS: The fecal microbiota of mice that express hREG3A had a significant shift in composition, compared with control mice, with enrichment of Clostridiales (Ruminococcaceae, Lachnospiraceae) and depletion of Bacteroidetes (Prevotellaceae); the TG mice developed less-severe colitis following administration of DSS than control mice, associated with preserved gut barrier integrity and reduced bacterial translocation, epithelial inflammation, and oxidative damage. A similar shift in the composition of the fecal microbiota occurred after a few months in TG mice heterozygous for REG3A that harbored a wild-type maternal microbiota at birth; these mice developed less-severe forms of colitis following DSS administration. Cohoused and germ-free mice fed feces from REG3A-TG mice and given DSS developed less-severe forms of colitis and had reduced lipopolysaccharide activation of the toll-like receptor 4 and increased survival times compared with mice not fed feces from REG3A-TG mice. REG3A TG mice developed only mild colonic inflammation after exposure to 2,4,6-trinitrobenzene sulfonic acid, compared with control mice. Control mice given intrarectal hREG3A and exposed to 2,4,6-trinitrobenzene sulfonic acid showed less colon damage and inflammation than mice not given intrarectal hREG3A. Fecal samples from REG3A-TG mice had lower levels of ROS than feces from control mice during DSS administration. Addition of hREG3A to bacterial cultures reduced levels of ROS and increased survival of oxygen-sensitive commensal bacteria (Faecalibacterium prausnitzii and Roseburia intestinalis). CONCLUSIONS: Mice with hepatocytes that express hREG3A, which travels to the intestinal lumen, are less sensitive to colitis than control mice. We found hREG3A to alter the colonic microbiota by decreasing levels of ROS. Fecal microbiota from REG3A-TG mice protect non-TG mice from induction of colitis. These findings indicate a role for reduction of oxidative stress in preserving the gut microbiota and its ability to prevent inflammation.


Asunto(s)
Bacterias/metabolismo , Colitis/prevención & control , Colon/metabolismo , Microbioma Gastrointestinal , Hepatocitos/metabolismo , Proteínas Asociadas a Pancreatitis/metabolismo , Animales , Bacterias/clasificación , Bacterias/crecimiento & desarrollo , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/microbiología , Colon/microbiología , Sulfato de Dextran , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Viabilidad Microbiana , Estrés Oxidativo/efectos de los fármacos , Proteínas Asociadas a Pancreatitis/genética , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Ácido Trinitrobencenosulfónico
15.
Nature ; 500(7464): 585-8, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23985875

RESUMEN

Complex gene-environment interactions are considered important in the development of obesity. The composition of the gut microbiota can determine the efficacy of energy harvest from food and changes in dietary composition have been associated with changes in the composition of gut microbial populations. The capacity to explore microbiota composition was markedly improved by the development of metagenomic approaches, which have already allowed production of the first human gut microbial gene catalogue and stratifying individuals by their gut genomic profile into different enterotypes, but the analyses were carried out mainly in non-intervention settings. To investigate the temporal relationships between food intake, gut microbiota and metabolic and inflammatory phenotypes, we conducted diet-induced weight-loss and weight-stabilization interventions in a study sample of 38 obese and 11 overweight individuals. Here we report that individuals with reduced microbial gene richness (40%) present more pronounced dys-metabolism and low-grade inflammation, as observed concomitantly in the accompanying paper. Dietary intervention improves low gene richness and clinical phenotypes, but seems to be less efficient for inflammation variables in individuals with lower gene richness. Low gene richness may therefore have predictive potential for the efficacy of intervention.


Asunto(s)
Dieta , Tracto Gastrointestinal/microbiología , Metagenoma/genética , Metabolismo Basal , Peso Corporal/efectos de los fármacos , Dieta Baja en Carbohidratos , Fibras de la Dieta/farmacología , Fibras de la Dieta/uso terapéutico , Proteínas en la Dieta/farmacología , Ingestión de Alimentos , Ingestión de Energía , Femenino , Frutas , Tracto Gastrointestinal/efectos de los fármacos , Interacción Gen-Ambiente , Genes Bacterianos/genética , Humanos , Inflamación/microbiología , Masculino , Metagenoma/efectos de los fármacos , Obesidad/dietoterapia , Obesidad/microbiología , Sobrepeso/dietoterapia , Sobrepeso/microbiología , Verduras , Pérdida de Peso/efectos de los fármacos
16.
Nature ; 500(7464): 541-6, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23985870

RESUMEN

We are facing a global metabolic health crisis provoked by an obesity epidemic. Here we report the human gut microbial composition in a population sample of 123 non-obese and 169 obese Danish individuals. We find two groups of individuals that differ by the number of gut microbial genes and thus gut bacterial richness. They contain known and previously unknown bacterial species at different proportions; individuals with a low bacterial richness (23% of the population) are characterized by more marked overall adiposity, insulin resistance and dyslipidaemia and a more pronounced inflammatory phenotype when compared with high bacterial richness individuals. The obese individuals among the lower bacterial richness group also gain more weight over time. Only a few bacterial species are sufficient to distinguish between individuals with high and low bacterial richness, and even between lean and obese participants. Our classifications based on variation in the gut microbiome identify subsets of individuals in the general white adult population who may be at increased risk of progressing to adiposity-associated co-morbidities.


Asunto(s)
Bacterias/aislamiento & purificación , Biomarcadores/metabolismo , Tracto Gastrointestinal/microbiología , Metagenoma , Adiposidad , Adulto , Bacterias/clasificación , Bacterias/genética , Índice de Masa Corporal , Estudios de Casos y Controles , Dieta , Dislipidemias/microbiología , Metabolismo Energético , Europa (Continente)/etnología , Femenino , Genes Bacterianos , Humanos , Inflamación/microbiología , Resistencia a la Insulina , Masculino , Metagenoma/genética , Obesidad/metabolismo , Obesidad/microbiología , Sobrepeso/metabolismo , Sobrepeso/microbiología , Filogenia , Delgadez/microbiología , Aumento de Peso , Pérdida de Peso , Población Blanca
17.
Gastroenterology ; 152(1): 111-123.e8, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27725146

RESUMEN

BACKGROUND & AIMS: We have limited knowledge about the association between the composition of the intestinal microbiota and clinical features of irritable bowel syndrome (IBS). We collected information on the fecal and mucosa-associated microbiota of patients with IBS and evaluated whether these were associated with symptoms. METHODS: We collected fecal and mucosal samples from adult patients who met the Rome III criteria for IBS at a secondary/tertiary care outpatient clinics in Sweden, as well as from healthy subjects. The exploratory set comprised 149 subjects (110 with IBS and 39 healthy subjects); 232 fecal samples and 59 mucosal biopsy samples were collected and analyzed by 16S ribosomal RNA targeted pyrosequencing. The validation set comprised 46 subjects (29 with IBS and 17 healthy subjects); 46 fecal samples, but no mucosal samples, were collected and analyzed. For each subject, we measured exhaled H2 and CH4, oro-anal transit time, and the severity of psychological and gastrointestinal symptoms. Fecal methanogens were measured by quantitative polymerase chain reaction. Numerical ecology analyses and a machine learning procedure were used to analyze the data. RESULTS: Fecal microbiota showed covariation with mucosal adherent microbiota. By using classic approaches, we found no differences in fecal microbiota abundance or composition between patients with IBS vs healthy patients. A machine learning procedure, a computational statistical technique, allowed us to reduce the 16S ribosomal RNA data complexity into a microbial signature for severe IBS, consisting of 90 bacterial operational taxonomic units. We confirmed the robustness of the intestinal microbial signature for severe IBS in the validation set. The signature was able to discriminate between patients with severe symptoms, patients with mild/moderate symptoms, and healthy subjects. By using this intestinal microbiota signature, we found IBS symptom severity to be associated negatively with microbial richness, exhaled CH4, presence of methanogens, and enterotypes enriched with Clostridiales or Prevotella species. This microbiota signature could not be explained by differences in diet or use of medications. CONCLUSIONS: In analyzing fecal and mucosal microbiota from patients with IBS and healthy individuals, we identified an intestinal microbiota profile that is associated with the severity of IBS symptoms. TRIAL REGISTRATION NUMBER: NCT01252550.


Asunto(s)
ADN Bacteriano/análisis , Heces/microbiología , Mucosa Intestinal/microbiología , Síndrome del Colon Irritable/microbiología , Microbiota , ARN Ribosómico 16S/análisis , Adulto , Bacteroides/aislamiento & purificación , Pruebas Respiratorias , Estudios de Casos y Controles , Clostridiales/aislamiento & purificación , Femenino , Microbioma Gastrointestinal , Tránsito Gastrointestinal , Humanos , Hidrógeno/análisis , Síndrome del Colon Irritable/fisiopatología , Aprendizaje Automático , Masculino , Metano/análisis , Methanobacteriales/aislamiento & purificación , Prevotella/aislamiento & purificación , Estudios Prospectivos , Índice de Severidad de la Enfermedad , Adulto Joven
18.
Cell Microbiol ; 19(2)2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27405092

RESUMEN

The intestinal epithelium is an active barrier separating the host from its microbiota. It senses microbial compounds through expression of a wide range of receptors including the Toll-like receptors (TLRs). TLRs have been shown to regulate epithelium permeability or secretion of defensin by Paneth cells. However, the expression and function of TLRs in enteroendocrine L-cells, a specific subtype of intestinal cells secreting PYY and GLP-1, have not yet been assessed. PYY and GLP-1 are implicated in regulation of gut motility, food intake and insulin secretion, and are of great interest regarding obesity and type 2 diabetes. Using a cellular model of human L-cells and a reporter system for NF-κB activation pathway, we reported functional expression of TLRs in these cells. Stimulation with specific TLR-agonists increased expression of Pyy but not Proglucagon in an NF-κB-dependent manner. Moreover, the effect of TLR stimulation was additive to butyrate, a product of bacterial fermentation, on Pyy expression. Additionally, butyrate also increased Tlr expression, including Tlr4, and the NF-κB response to TLR stimulation. Altogether, our results demonstrated a role of TLRs in the modulation of Pyy expression and the importance of butyrate, a product of bacterial fermentation in regulation of microbial TLR-dependent sensing.


Asunto(s)
Butiratos/metabolismo , Células Enteroendocrinas/metabolismo , Regulación de la Expresión Génica , Péptido YY/metabolismo , Receptores Toll-Like/agonistas , Línea Celular , Péptido 1 Similar al Glucagón/biosíntesis , Humanos , FN-kappa B/metabolismo
20.
Environ Microbiol ; 19(9): 3579-3594, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28695648

RESUMEN

Bacterial adhesion is a critical step for colonization of the host. The pioneer colonizer and commensal bacterium of the human gastrointestinal tract, Streptococcus salivarius, has strong adhesive properties but the molecular determinants of this adhesion remain uncharacterized. Serine-rich repeat (SRR) glycoproteins are a family of adhesins that fulfil an important role in adhesion. In general, Gram-positive bacterial genomes have a unique SRR glycoprotein-encoding gene. We demonstrate that S. salivarius expresses three large and glycosylated surface-exposed proteins - SrpA, SrpB and SrpC - that show characteristics of SRR glycoproteins and are secreted through the accessory SecA2/Y2 system. Two glycosyltransferases - GtfE/F - encoded outside of the secA2/Y2 locus, unusually, perform the first step of the sequential glycosylation process, which is crucial for SRR activity. We show that SrpB and SrpC play complementary adhesive roles involved in several steps of the colonization process: auto-aggregation, biofilm formation and adhesion to a variety of host epithelial cells and components. We also show that at least one of the S. salivarius SRR glycoproteins is important for colonization in mice. SrpA, SrpB and SrpC are the main factors underlying the multifaceted adhesion of S. salivarius and, therefore, play a major role in host colonization.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana/fisiología , Proteínas Bacterianas/metabolismo , Mucosa Intestinal/microbiología , Glicoproteínas de Membrana/metabolismo , Streptococcus salivarius/patogenicidad , Animales , Adhesión Bacteriana/genética , Células Epiteliales/microbiología , Tracto Gastrointestinal/microbiología , Glucosiltransferasas/genética , Glicosilación , Humanos , Masculino , Ratones , Modelos Animales , Streptococcus salivarius/genética , Streptococcus salivarius/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda