Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 178(6): 1313-1328.e13, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491384

RESUMEN

Emerging evidence indicates a central role for the microbiome in immunity. However, causal evidence in humans is sparse. Here, we administered broad-spectrum antibiotics to healthy adults prior and subsequent to seasonal influenza vaccination. Despite a 10,000-fold reduction in gut bacterial load and long-lasting diminution in bacterial diversity, antibody responses were not significantly affected. However, in a second trial of subjects with low pre-existing antibody titers, there was significant impairment in H1N1-specific neutralization and binding IgG1 and IgA responses. In addition, in both studies antibiotics treatment resulted in (1) enhanced inflammatory signatures (including AP-1/NR4A expression), observed previously in the elderly, and increased dendritic cell activation; (2) divergent metabolic trajectories, with a 1,000-fold reduction in serum secondary bile acids, which was highly correlated with AP-1/NR4A signaling and inflammasome activation. Multi-omics integration revealed significant associations between bacterial species and metabolic phenotypes, highlighting a key role for the microbiome in modulating human immunity.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Antivirales/inmunología , Microbioma Gastrointestinal/fisiología , Inmunidad/efectos de los fármacos , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Adolescente , Adulto , Formación de Anticuerpos , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Voluntarios Sanos , Humanos , Inmunogenicidad Vacunal/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Masculino , Adulto Joven
2.
Gastroenterology ; 162(3): 743-756, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34774538

RESUMEN

BACKGROUND & AIMS: Epidemiologic and murine studies suggest that dietary emulsifiers promote development of diseases associated with microbiota dysbiosis. Although the detrimental impact of these compounds on the intestinal microbiota and intestinal health have been demonstrated in animal and in vitro models, impact of these food additives in healthy humans remains poorly characterized. METHODS: To examine this notion in humans, we performed a double-blind controlled-feeding study of the ubiquitous synthetic emulsifier carboxymethylcellulose (CMC) in which healthy adults consumed only emulsifier-free diets (n = 9) or an identical diet enriched with 15 g per day of CMC (n = 7) for 11 days. RESULTS: Relative to control subjects, CMC consumption modestly increased postprandial abdominal discomfort and perturbed gut microbiota composition in a way that reduced its diversity. Moreover, CMC-fed subjects exhibited changes in the fecal metabolome, particularly reductions in short-chain fatty acids and free amino acids. Furthermore, we identified 2 subjects consuming CMC who exhibited increased microbiota encroachment into the normally sterile inner mucus layer, a central feature of gut inflammation, as well as stark alterations in microbiota composition. CONCLUSIONS: These results support the notion that the broad use of CMC in processed foods may be contributing to increased prevalence of an array of chronic inflammatory diseases by altering the gut microbiome and metabolome (ClinicalTrials.gov, number NCT03440229).


Asunto(s)
Carboximetilcelulosa de Sodio/efectos adversos , Dieta/efectos adversos , Emulsionantes/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Metaboloma/efectos de los fármacos , Animales , Método Doble Ciego , Disbiosis/etiología , Heces , Femenino , Voluntarios Sanos , Humanos , Masculino , Ratones
3.
Gastroenterology ; 163(6): 1658-1671.e16, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35988658

RESUMEN

BACKGROUND & AIMS: Pathogenesis of hepatocellular carcinoma (HCC), which kills millions annually, is poorly understood. Identification of risk factors and modifiable determinants and mechanistic understanding of how they impact HCC are urgently needed. METHODS: We sought early prognostic indicators of HCC in C57BL/6 mice, which we found were prone to developing this disease when fed a fermentable fiber-enriched diet. Such markers were used to phenotype and interrogate stages of HCC development. Their human relevance was tested using serum collected prospectively from an HCC/case-control cohort. RESULTS: HCC proneness in mice was dictated by the presence of congenitally present portosystemic shunt (PSS), which resulted in markedly elevated serum bile acids (BAs). Approximately 10% of mice from various sources exhibited PSS/cholemia, but lacked an overt phenotype when fed standard chow. However, PSS/cholemic mice fed compositionally defined diets, developed BA- and cyclooxygenase-dependent liver injury, which was exacerbated and uniformly progressed to HCC when diets were enriched with the fermentable fiber inulin. Such progression to cholestatic HCC associated with exacerbated cholemia and an immunosuppressive milieu, both of which were required in that HCC was prevented by impeding BA biosynthesis or neutralizing interleukin-10 or programmed death protein 1. Analysis of human sera revealed that elevated BA was associated with future development of HCC. CONCLUSIONS: PSS is relatively common in C57BL/6 mice and causes silent cholemia, which predisposes to liver injury and HCC, particularly when fed a fermentable fiber-enriched diet. Incidence of silent PSS/cholemia in humans awaits investigation. Regardless, measuring serum BA may aid HCC risk assessment, potentially alerting select individuals to consider dietary or BA interventions.


Asunto(s)
Carcinoma Hepatocelular , Enfermedades del Sistema Digestivo , Neoplasias Hepáticas , Humanos , Ratones , Animales , Neoplasias Hepáticas/etiología , Carcinoma Hepatocelular/etiología , Ratones Endogámicos C57BL , Prótesis e Implantes , Fibras de la Dieta
4.
Proc Natl Acad Sci U S A ; 117(35): 21519-21526, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817517

RESUMEN

The intestinal epithelium is a highly dynamic structure that rejuvenates in response to acute stressors and can undergo alterations in cellular composition as animals age. The microbiota, acting via secreted factors related to indole, appear to regulate the sensitivity of the epithelium to stressors and promote epithelial repair via IL-22 and type I IFN signaling. As animals age, the cellular composition of the intestinal epithelium changes, resulting in a decreased proportion of goblet cells in the colon. We show that colonization of young or geriatric mice with bacteria that secrete indoles and various derivatives or administration of the indole derivative indole-3 aldehyde increases proliferation of epithelial cells and promotes goblet cell differentiation, reversing an effect of aging. To induce goblet cell differentiation, indole acts via the xenobiotic aryl hydrocarbon receptor to increase expression of the cytokine IL-10. However, the effects of indoles on goblet cells do not depend on type I IFN or on IL-22 signaling, pathways responsible for protection against acute stressors. Thus, indoles derived from the commensal microbiota regulate intestinal homeostasis, especially during aging, via mechanisms distinct from those used during responses to acute stressors. Indoles may have utility as an intervention to limit the decline of barrier integrity and the resulting systemic inflammation that occurs with aging.


Asunto(s)
Células Caliciformes/efectos de los fármacos , Células Caliciformes/microbiología , Indoles/farmacología , Interleucina-10/metabolismo , Microbiota/fisiología , Receptores de Hidrocarburo de Aril/metabolismo , Envejecimiento/metabolismo , Animales , Bacterias/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Femenino , Células Caliciformes/citología , Células Caliciformes/metabolismo , Interleucina-10/biosíntesis , Interleucinas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Moco/metabolismo , Transducción de Señal , Interleucina-22
5.
J Autoimmun ; 128: 102814, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35298976

RESUMEN

Inflammatory bowel disease (IBD) is a serious public health problem in Western society with a continuing increase in incidence worldwide. Safe, targeted medicines for IBD are not yet available. Autophagy, a vital process implicated in normal cell homeostasis, provides a potential point of entry for the treatment of IBDs, as several autophagy-related genes are associated with IBD risk. We conducted a series of experiments in three distinct mouse models of colitis to test the effectiveness of therapeutic P140, a phosphopeptide that corrects autophagy dysfunctions in other autoimmune and inflammatory diseases. Colitis was experimentally induced in mice by administering dextran sodium sulfate and 2,4,6 trinitrobenzene sulfonic acid. Transgenic mice lacking both il-10 and iRhom2 - involved in tumor necrosis factor α secretion - were also used. In the three models investigated, P140 treatment attenuated the clinical and histological severity of colitis. Post-treatment, altered expression of several macroautophagy and chaperone-mediated autophagy markers, and of pro-inflammatory mediators was corrected. Our results demonstrate that therapeutic intervention with an autophagy modulator improves colitis in animal models. These findings highlight the potential of therapeutic peptide P140 for use in the treatment of IBD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Animales , Autofagia , Proteínas Portadoras , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Sulfato de Dextran , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/metabolismo , Lisosomas/metabolismo , Ratones
6.
Gut ; 68(10): 1801-1812, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30670576

RESUMEN

OBJECTIVE: Diets rich in fermentable fibres provide an array of health benefits; however, many patients with IBD report poor tolerance to fermentable fibre-rich foods. Intervention studies with dietary fibres in murine models of colonic inflammation have yielded conflicting results on whether fibres ameliorate or exacerbate IBD. Herein, we examined how replacing the insoluble fibre, cellulose, with the fermentable fibres, inulin or pectin, impacted murine colitis resulting from immune dysregulation via inhibition of interleukin (IL)-10 signalling and/or innate immune deficiency (Tlr5KO). DESIGN: Mice were fed with diet containing either cellulose, inulin or pectin and subjected to weekly injections of an IL-10 receptor (αIL-10R) neutralising antibody. Colitis development was examined by serological, biochemical, histological and immunological parameters. RESULTS: Inulin potentiated the severity of αIL10R-induced colitis, while pectin ameliorated the disease. Such exacerbation of colitis following inulin feeding was associated with enrichment of butyrate-producing bacteria and elevated levels of caecal butyrate. Blockade of butyrate production by either metronidazole or hops ß-acids ameliorated colitis severity in inulin-fed mice, whereas augmenting caecal butyrate via tributyrin increased colitis severity in cellulose containing diet-fed mice. Elevated butyrate levels were associated with increased IL-1ß activity, while inhibition of the NOD-like receptor protein 3 by genetic, pharmacologic or dietary means markedly reduced colitis. CONCLUSION: These results not only support the notion that fermentable fibres have the potential to ameliorate colitis but also caution that, in some contexts, prebiotic fibres can lead to gut dysbiosis and surfeit colonic butyrate that might exacerbate IBD.


Asunto(s)
Colitis/metabolismo , Fibras de la Dieta/metabolismo , Microbioma Gastrointestinal/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad Aguda , Animales , Colitis/dietoterapia , Colitis/microbiología , Modelos Animales de Enfermedad , Fermentación , Masculino , Ratones , Ratones Endogámicos NOD
7.
Am J Physiol Endocrinol Metab ; 315(1): E1-E6, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29486139

RESUMEN

The concept that the gut microbiota plays a broadly important role in health and disease in general, and metabolic health in particular, is now well established. However, many of the underlying mechanisms remain poorly understood while approaches to reliably manipulate the microbiota to promote health have not yet been clearly defined. Nonetheless, progress in these areas is steadily accelerating. Herein, we review select areas of progress that have been made in the last year that should hasten the era in which the microbiota can be therapeutically manipulated to promote metabolic health.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Metabolismo/fisiología , Animales , Dieta , Humanos , Enfermedades Metabólicas/microbiología , Enfermedades Metabólicas/fisiopatología , Obesidad/metabolismo
8.
Microbiome ; 12(1): 5, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38178260

RESUMEN

BACKGROUND: The observation that the intestinal microbiota is  central in the development of IBD suggests that dietary fiber, the microbiota's primary source of nourishment, could play a central role in these diseases. Accordingly, enriching diets with specific soluble fibers remodels microbiota and modulates colitis sensitivity. In humans, a recent study suggests that the microbiota of select IBD patients might influence the impacts they would experience upon fiber exposure. We sought here to define the extent to which individual microbiotas varied in their responsiveness to purified soluble fiber inulin and psyllium. Moreover, the extent to which such variance might impact proneness to colitis. RESULTS: We observed a high level of inter-individual variation in microbiota responsiveness to fiber inulin and psyllium: while microbiotas from select donors exhibited stark fiber-induced modulation in composition, pro-inflammatory potential, and metabolomic profile, others were only minimally impacted. Mice transplanted with fiber-sensitive microbiomes exhibited colitis highly modulated by soluble fiber consumption, while mice receiving fiber-resistant microbiotas displayed colitis severity irrespective of fiber exposure. CONCLUSION: The extent to which select soluble fibers alter proneness to colitis is highly influenced by an individual's microbiota composition and further investigation of individual microbiota responsiveness toward specific dietary fiber could pave the way to personalized fiber-based intervention, both in IBD patients and healthy individuals. Video Abstract.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Psyllium , Humanos , Ratones , Animales , Psyllium/efectos adversos , Inulina , Colitis/inducido químicamente , Fibras de la Dieta
9.
Gut Microbes ; 15(1): 2221095, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37305942

RESUMEN

Impacts of dietary fiber on intestinal inflammation are complex, but some specific semi-purified fibers, particularly psyllium, can protect humans and rodents against colitis. Mechanisms underlying such protection are not fully understood but may involve activation of the FXR bile acid receptor. Obesity and its associated consequences, referred to as metabolic syndrome, are associated with, and promoted by, low-grade inflammation in a variety of tissues including the intestine. Hence, we examined whether psyllium might ameliorate the low-grade intestinal inflammation that occurs in diet-induced obesity and, moreover, the extent to which it might ameliorate adiposity and/or dysglycemia in this disease model. We observed that enriching a high-fat diet with psyllium provided strong protection against the low-grade gut inflammation and metabolic consequences that were otherwise induced by the obesogenic diet. Such protection was fully maintained in FXR-deficient mice, indicating that distinct mechanisms mediate psyllium's protection against colitis and metabolic syndrome. Nor did psyllium's protection associate with, or require, fermentation or IL-22 production, both of which are key mediators of beneficial impacts of some other dietary fibers. Psyllium's beneficial impacts were not evident in germfree mice but were observed in Altered Schaedler Flora mice, in which psyllium modestly altered relative and absolute abundance of the small number of taxa present in these gnotobiotic mice. Thus, psyllium protects mice against diet-induced obesity/metabolic syndrome by a mechanism independent of FXR and fermentation but nonetheless requires the presence of at least a minimal microbiota.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Síndrome Metabólico , Psyllium , Humanos , Animales , Ratones , Síndrome Metabólico/prevención & control , Dieta Occidental , Obesidad/prevención & control , Fibras de la Dieta , Inflamación
10.
Cell Mol Gastroenterol Hepatol ; 15(6): 1421-1442, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36828279

RESUMEN

BACKGROUND & AIMS: Fiber-rich foods promote health, but mechanisms by which they do so remain poorly defined. Screening fiber types, in mice, revealed psyllium had unique ability to ameliorate 2 chronic inflammatory states, namely, metabolic syndrome and colitis. We sought to determine the mechanism of action of the latter. METHODS: Mice were fed grain-based chow, which is naturally rich in fiber or compositionally defined diets enriched with semi-purified fibers. Mice were studied basally and in models of chemical-induced and T-cell transfer colitis. RESULTS: Relative to all diets tested, mice consuming psyllium-enriched compositionally defined diets were markedly protected against both dextran sulfate sodium- and T-cell transfer-induced colitis, as revealed by clinical-type, histopathologic, morphologic, and immunologic parameters. Such protection associated with stark basal changes in the gut microbiome but was independent of fermentation and, moreover, maintained in mice harboring a minimal microbiota (ie, Altered Schaedler Flora). Transcriptomic analysis revealed psyllium induced expression of genes mediating bile acids (BA) secretion, suggesting that psyllium's known ability to bind BA might contribute to its ability to prevent colitis. As expected, psyllium resulted in elevated level of fecal BA, reflecting their removal from enterohepatic circulation but, in stark contrast to the BA sequestrant cholestyramine, increased serum BA levels. Moreover, the use of BA mimetics that activate the farnesoid X receptor (FXR), as well as the use of FXR-knockout mice, suggested that activation of FXR plays a central role in psyllium's protection against colitis. CONCLUSIONS: Psyllium protects against colitis via altering BA metabolism resulting in activation of FXR, which suppresses pro-inflammatory signaling.


Asunto(s)
Colitis , Psyllium , Ratones , Animales , Psyllium/efectos adversos , Ácidos y Sales Biliares , Promoción de la Salud , Colitis/inducido químicamente , Colitis/prevención & control , Colitis/metabolismo , Inflamación , Ratones Noqueados
11.
Nutrients ; 13(3)2021 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-33806985

RESUMEN

Diet shapes the gut microbiota which impacts hepatic lipid metabolism. Modifications in liver fat content are associated with metabolic disorders. We investigated the extent of dietary fat and fiber-induced alterations in the composition of gut microbiota and hepatic fatty acids (FAs). Mice were fed a purified low-fat diet (LFD) or high-fat diet (HFD) containing non-soluble fiber cellulose or soluble fiber inulin. HFD induced hepatic decreases in the amounts of C14:0, C16:1n-7, C18:1n-7 and increases in the amounts of C17:0, C20:0, C16:1n-9, C22:5n-3, C20:2n-6, C20:3n-6, and C22:4n-6. When incorporated in a LFD, inulin poorly affected the profile of FAs. However, when incorporated in a HFD, it (i) specifically led to an increase in the amounts of hepatic C18:0, C22:0, total polyunsaturated FAs (PUFAs), total n-6 PUFAs, C18:3n-3, and C18:2n-6, (ii) exacerbated the HFD-induced increase in the amount of C17:0, and (iii) prevented the HFD-induced increases in C16:1n-9 and C20:3n-6. Importantly, the expression/activity of some elongases and desaturases, as well as the gut microbiota composition, were impacted by the dietary fat and fiber content. To conclude, inulin modulated gut microbiota and hepatic fatty acid composition, and further investigations will determine whether a causal relationship exists between these two parameters.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Inulina/administración & dosificación , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Animales , Glucemia/metabolismo , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Dieta con Restricción de Grasas , Grasas de la Dieta/administración & dosificación , Fibras de la Dieta/administración & dosificación , Ácidos Grasos Insaturados/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/aislamiento & purificación , Triglicéridos/sangre
12.
Cell Mol Gastroenterol Hepatol ; 12(3): 983-1000, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33940221

RESUMEN

BACKGROUND & AIMS: Nourishment of gut microbiota via consumption of fermentable fiber promotes gut health and guards against metabolic syndrome. In contrast, how dietary fiber impacts type 1 diabetes is less clear. METHODS: To examine impact of dietary fibers on development of type 1 diabetes in the streptozotocin (STZ)-induced and spontaneous non-obese diabetes (NOD) models, mice were fed grain-based chow (GBC) or compositionally defined diets enriched with a fermentable fiber (inulin) or an insoluble fiber (cellulose). Spontaneous (NOD mice) or STZ-induced (wild-type mice) diabetes was monitored. RESULTS: Relative to GBC, low-fiber diets exacerbated STZ-induced diabetes, whereas diets enriched with inulin, but not cellulose, strongly protected against or treated it. Inulin's restoration of glycemic control prevented loss of adipose depots, while reducing food and water consumption. Inulin normalized pancreatic function and markedly enhanced insulin sensitivity. Such amelioration of diabetes was associated with alterations in gut microbiota composition and was eliminated by antibiotic administration. Pharmacologic blockade of fermentation reduced inulin's beneficial impact on glycemic control, indicating a role for short-chain fatty acids (SCFA). Furthermore, inulin's microbiota-dependent anti-diabetic effect associated with SCFA-independent restoration of interleukin 22, which was necessary and sufficient to ameliorate STZ-induced diabetes. Inulin-enriched diets significantly delayed diabetes in NOD mice. CONCLUSIONS: Fermentable fiber confers microbiota-dependent increases in SCFA and interleukin 22 that, together, may have potential to prevent and/or treat type 1 diabetes.


Asunto(s)
Bacterias/clasificación , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Fibras de la Dieta/administración & dosificación , Ácidos Grasos Volátiles/metabolismo , Interleucinas/metabolismo , Inulina/administración & dosificación , Animales , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/aislamiento & purificación , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/microbiología , Fibras de la Dieta/farmacología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Hemoglobina Glucada/metabolismo , Inulina/farmacología , Masculino , Ratones , Ratones Endogámicos NOD , Tamaño de los Órganos/efectos de los fármacos , Páncreas/efectos de los fármacos , Páncreas/fisiopatología , Estreptozocina/efectos adversos , Resultado del Tratamiento , Interleucina-22
13.
Cell Rep ; 33(1): 108229, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33027647

RESUMEN

Dietary emulsifiers carboxymethylcellulose (CMC) and polysorbate-80 (P80) disturb gut microbiota, promoting chronic inflammation. Mice with minimal microbiota are protected against emulsifiers' effects, leading us to hypothesize that these compounds might provoke select pathobionts to promote inflammation. Gnotobiotic wild-type (WT) and interleukin-10 (IL-10)-/- mice were colonized with Crohn's-disease-associated adherent-invasive E. coli (AIEC) and subsequently administered CMC or P80. AIEC colonization of GF and altered Schaedler flora (ASF) mice results in chronic intestinal inflammation and metabolism dysregulations when consuming the emulsifier. In IL-10-/- mice, AIEC mono-colonization results in severe intestinal inflammation in response to emulsifiers. Exposure of AIEC to emulsifiers in vitro increases its motility and ability to adhere to intestinal epithelial cells. Transcriptomic analysis reveals that emulsifiers directly induce expression of clusters of genes that mediate AIEC virulence and promotion of inflammation. To conclude, emulsifiers promote virulence and encroachment of pathobionts, providing a means by which these compounds may drive inflammation in hosts carrying such bacteria.


Asunto(s)
Emulsionantes/uso terapéutico , Escherichia coli/patogenicidad , Flagelina/metabolismo , Expresión Génica/genética , Intestinos/microbiología , Animales , Enfermedad Crónica , Dieta , Emulsionantes/farmacología , Escherichia coli/crecimiento & desarrollo , Humanos , Inflamación/metabolismo , Ratones
14.
Nat Commun ; 11(1): 2471, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424203

RESUMEN

Gut microbes are linked to host metabolism, but specific mechanisms remain to be uncovered. Ceramides, a type of sphingolipid (SL), have been implicated in the development of a range of metabolic disorders from insulin resistance (IR) to hepatic steatosis. SLs are obtained from the diet and generated by de novo synthesis in mammalian tissues. Another potential, but unexplored, source of mammalian SLs is production by Bacteroidetes, the dominant phylum of the gut microbiome. Genomes of Bacteroides spp. and their relatives encode serine palmitoyltransfease (SPT), allowing them to produce SLs. Here, we explore the contribution of SL-production by gut Bacteroides to host SL homeostasis. In human cell culture, bacterial SLs are processed by host SL-metabolic pathways. In mouse models, Bacteroides-derived lipids transfer to host epithelial tissue and the hepatic portal vein. Administration of B. thetaiotaomicron to mice, but not an SPT-deficient strain, reduces de novo SL production and increases liver ceramides. These results indicate that gut-derived bacterial SLs affect host lipid metabolism.


Asunto(s)
Bacteroides/fisiología , Ceramidas/metabolismo , Microbioma Gastrointestinal , Redes y Vías Metabólicas , Esfingolípidos/metabolismo , Animales , Células CACO-2 , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Vida Libre de Gérmenes , Humanos , Resistencia a la Insulina , Mucosa Intestinal/microbiología , Hígado/metabolismo , Redes y Vías Metabólicas/genética , Ratones , Mutación/genética , Serina C-Palmitoiltransferasa/deficiencia , Serina C-Palmitoiltransferasa/genética , Serina C-Palmitoiltransferasa/metabolismo
15.
Cell Mol Gastroenterol Hepatol ; 9(2): 313-333, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31593782

RESUMEN

BACKGROUND & AIMS: Consumption of a low-fiber, high-fat, Western-style diet (WSD) induces adiposity and adipose inflammation characterized by increases in the M1:M2 macrophage ratio and proinflammatory cytokine expression, both of which contribute to WSD-induced metabolic syndrome. WSD-induced adipose inflammation might result from endoplasmic reticulum stress in lipid-overloaded adipocytes and/or dissemination of gut bacterial products, resulting in activation of innate immune signaling. Hence, we aimed to investigate the role of the gut microbiota, and its detection by innate immune signaling pathways, in WSD-induced adipose inflammation. METHODS: Mice were fed grain-based chow or a WSD for 8 weeks, assessed metabolically, and intestinal and adipose tissue were analyzed by flow cytometry and quantitative reverse transcription polymerase chain reaction. Microbiota was ablated via antibiotics and use of gnotobiotic mice that completely lacked microbiota (germ-free mice) or had a low-complexity microbiota (altered Schaedler flora). Innate immune signaling was ablated by genetic deletion of Toll-like receptor signaling adaptor myeloid differentiation primary response 88. RESULTS: Ablation of microbiota via antibiotic, germ-free, or altered Schaedler flora approaches did not significantly impact WSD-induced adiposity, yet dramatically reduced WSD-induced adipose inflammation as assessed by macrophage populations and cytokine expression. Microbiota ablation also prevented colonic neutrophil and CD103- dendritic cell infiltration. Such reduced indices of inflammation correlated with protection against WSD-induced dysglycemia, hypercholesterolemia, and liver dysfunction. Genetic deletion of myeloid differentiation primary response 88 also prevented WSD-induced adipose inflammation. CONCLUSIONS: These results indicate that adipose inflammation, and some aspects of metabolic syndrome, are not purely a consequence of diet-induced adiposity per se but, rather, may require disturbance of intestine-microbiota interactions and subsequent activation of innate immunity.


Asunto(s)
Tejido Adiposo/inmunología , Adiposidad/inmunología , Dieta Occidental/efectos adversos , Microbioma Gastrointestinal/inmunología , Síndrome Metabólico/inmunología , Adipocitos/inmunología , Adipocitos/metabolismo , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/inmunología , Trasplante de Microbiota Fecal , Heces/microbiología , Humanos , Inflamación/inmunología , Inflamación/microbiología , Macrófagos/inmunología , Masculino , Síndrome Metabólico/microbiología , Ratones , Transducción de Señal
16.
Sci Rep ; 8(1): 12301, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-30120269

RESUMEN

A high prevalence of adherent-invasive E. coli (AIEC) in the intestinal mucosa of Crohn's disease patients has been shown. AIEC colonize the intestine and induce inflammation in genetically predisposed mouse models including CEABAC10 transgenic (Tg) mice expressing human CEACAM6-receptor for AIEC and eif2ak4-/- mice exhibiting autophagy defect in response to AIEC infection. Here, we aimed at investigating whether gut microbiota modification contributes to AIEC-induced intestinal inflammation in these mouse models. For this, eif2ak4+/+ and eif2ak4-/- mice or CEABAC10 Tg mice invalidated for Eif2ak4 gene (Tg/eif2ak4-/-) or not (Tg/eif2ak4+/+) were infected with the AIEC reference strain LF82 or the non-pathogenic E. coli K12 MG1655 strain. In all mouse groups, LF82 colonized the gut better and longer than MG1655. No difference in fecal microbiota composition was observed in eif2ak4+/+ and eif2ak4-/- mice before infection and at day 1 and 4 post-infection. LF82-infected eif2ak4-/- mice exhibited altered fecal microbiota composition at day 14 and 21 post-infection and increased fecal lipocalin-2 level at day 21 post-infection compared to other groups, indicating that intestinal inflammation developed after microbiota modification. Similar results were obtained for LF82-infected Tg/eif2ak4-/- mice. These results suggest that in genetically predisposed hosts, AIEC colonization might induce chronic intestinal inflammation by altering the gut microbiota composition.


Asunto(s)
Escherichia coli K12/aislamiento & purificación , Microbioma Gastrointestinal/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Animales , Escherichia coli/genética , Escherichia coli/fisiología , Escherichia coli K12/genética , Femenino , Microbioma Gastrointestinal/genética , Intestinos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/genética
17.
Inflamm Bowel Dis ; 22(3): 516-28, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26595556

RESUMEN

BACKGROUND: Crohn's disease is a chronic inflammatory bowel disease, of which the etiology involves environmental, genetic, and microbial factors. A high prevalence of adherent-invasive Escherichia coli, named AIEC, has been reported in the intestinal mucosa of patients with Crohn's disease. Exosomes are extracellular vesicles that function in intercellular communication and have been implicated in host responses to intracellular pathogens. We investigated the potential involvement of exosomes in host response to AIEC infection. METHODS: Human intestinal epithelial T84 cells, THP-1 macrophages, and CEABAC10 transgenic mice were infected with the AIEC reference strain LF82 or the nonpathogenic E. coli K-12 MG1655 strain. Exosomes were purified using the ExoQuick reagent. RESULTS: LF82 infection induced the release of exosomes by T84 and THP-1 cells. Compared with exosomes released from the uninfected or MG1655-infected T84 cells, those released from LF82-infected cells activated nuclear factor-kappa B, mitogen-activated protein kinases p38, and c-Jun N-terminal kinase and increased the secretion of proinflammatory cytokines in naive THP-1 macrophages. LF82 infection of THP-1 macrophages also induced the release of exosomes that triggered a proinflammatory response in recipient THP-1 cells. Importantly, stimulation of T84 or THP-1 cells with exosomes released from LF82-infected cells increased LF82 intracellular replication compared with stimulation with exosomes secreted by uninfected cells. Exosomes purified from intestinal lumen of CEABAC10 transgenic mice infected with LF82 increased proinflammatory responses in murine RAW 264.7 macrophages compared with those from uninfected or MG1655-infected mice. CONCLUSIONS: Exosomes are new mediators of host-AIEC interaction with their capacity to activate innate immune responses and subvert the control of AIEC replication.


Asunto(s)
Adhesión Bacteriana/inmunología , Enfermedad de Crohn/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli/patogenicidad , Exosomas/microbiología , Inmunidad Innata/inmunología , Mucosa Intestinal/microbiología , Animales , Western Blotting , Enfermedad de Crohn/inmunología , Escherichia coli/crecimiento & desarrollo , Infecciones por Escherichia coli/inmunología , Exosomas/inmunología , Femenino , Humanos , Mucosa Intestinal/inmunología , Masculino , Ratones , Ratones Transgénicos
18.
Autophagy ; 12(5): 770-83, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-26986695

RESUMEN

The intestinal mucosa of Crohn disease (CD) patients is abnormally colonized by adherent-invasive E. coli (AIEC). Upon AIEC infection, autophagy is induced in host cells to restrain bacterial intracellular replication. The underlying mechanism, however, remains unknown. Here, we investigated the role of the EIF2AK4-EIF2A/eIF2α-ATF4 pathway in the autophagic response to AIEC infection. We showed that infection of human intestinal epithelial T84 cells with the AIEC reference strain LF82 activated the EIF2AK4-EIF2A-ATF4 pathway, as evidenced by increased phospho-EIF2AK4, phospho-EIF2A and ATF4 levels. EIF2AK4 depletion inhibited autophagy activation in response to LF82 infection, leading to increased LF82 intracellular replication and elevated pro-inflammatory cytokine production. Mechanistically, EIF2AK4 depletion suppressed the LF82-induced ATF4 binding to promoters of several autophagy genes including MAP1LC3B, BECN1, SQSTM1, ATG3 and ATG7, and this subsequently inhibited transcription of these genes. LF82 infection of wild-type (WT), but not eif2ak4(-/-), mice activated the EIF2AK4-EIF2A-ATF4 pathway, inducing autophagy gene transcription and autophagy response in enterocytes. Consequently, eif2ak4(-/-) mice exhibited increased intestinal colonization by LF82 bacteria and aggravated inflammation compared to WT mice. Activation of the EIF2AK4-EIF2A-ATF4 pathway was observed in ileal biopsies from patients with noninflamed CD, and this was suppressed in inflamed CD, suggesting that a defect in the activation of this pathway could be one of the mechanisms contributing to active disease. In conclusion, we show that activation of the EIF2AK4-EIF2A-ATF4 pathway upon AIEC infection serves as a host defense mechanism to induce functional autophagy to control AIEC intracellular replication.


Asunto(s)
Autofagia/fisiología , Enfermedad de Crohn/microbiología , Infecciones por Escherichia coli/metabolismo , Mucosa Intestinal/microbiología , Transducción de Señal , Factor de Transcripción Activador 4/metabolismo , Adolescente , Adulto , Enfermedad de Crohn/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Humanos , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Intestinos/patología , Masculino , Proteínas Serina-Treonina Quinasas/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA