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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(11)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38891933

RESUMEN

The role of the gut microbiota and its interplay with host metabolic health, particularly in the context of type 2 diabetes mellitus (T2DM) management, is garnering increasing attention. Dipeptidyl peptidase 4 (DPP4) inhibitors, commonly known as gliptins, constitute a class of drugs extensively used in T2DM treatment. However, their potential interactions with gut microbiota remain poorly understood. In this study, we employed computational methodologies to investigate the binding affinities of various gliptins to DPP4-like homologs produced by intestinal bacteria. The 3D structures of DPP4 homologs from gut microbiota species, including Segatella copri, Phocaeicola vulgatus, Bacteroides uniformis, Parabacteroides merdae, and Alistipes sp., were predicted using computational modeling techniques. Subsequently, molecular dynamics simulations were conducted for 200 ns to ensure the stability of the predicted structures. Stable structures were then utilized to predict the binding interactions with known gliptins through molecular docking algorithms. Our results revealed binding similarities of gliptins toward bacterial DPP4 homologs compared to human DPP4. Specifically, certain gliptins exhibited similar binding scores to bacterial DPP4 homologs as they did with human DPP4, suggesting a potential interaction of these drugs with gut microbiota. These findings could help in understanding the interplay between gliptins and gut microbiota DPP4 homologs, considering the intricate relationship between the host metabolism and microbial communities in the gut.


Asunto(s)
Diabetes Mellitus Tipo 2 , Dipeptidil Peptidasa 4 , Inhibidores de la Dipeptidil-Peptidasa IV , Microbioma Gastrointestinal , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Dipeptidil Peptidasa 4/metabolismo , Dipeptidil Peptidasa 4/química , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Unión Proteica , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Sitios de Unión
2.
Curr Opin Clin Nutr Metab Care ; 24(6): 536-542, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34622826

RESUMEN

PURPOSE OF REVIEW: Gluten is a complex mixture of highly immunogenic glutamine- and proline-rich proteins found in some cereals. In celiac disease (CeD), gluten triggers an autoimmune response due to its interaction with the human leukocyte antigen heterodimers that confer the genetic risk. The involvement of gluten in other disorders has also been investigated, but its role beyond CeD is still unclear. Here, we review the most recent evidence of the involvement of gluten in diseases and the opportunities of manipulating the gut microbiota to treat or prevent gluten-related conditions. RECENT FINDINGS: Most of the new studies have been conducted in the context of CeD, where important evidence has been gained on associations between the gut microbiota, genotype, and environmental factors such as breastfeeding and antibiotics. The role of the microbiota has been investigated in several prospective, observational and interventional studies with probiotics, which together showed that the gut microbiota could be targeted to ameliorate and aid in the prevention of CeD development. SUMMARY: Several studies have evidenced how genetic and environmental factors influence the gut microbiome with consequences in CeD. These findings could inspire the development of microbiota modulation strategies to support the prevention or treatment of CeD.


Asunto(s)
Enfermedad Celíaca , Microbioma Gastrointestinal , Dieta Sin Gluten , Glútenes/efectos adversos , Humanos , Estudios Prospectivos
3.
Int J Mol Sci ; 22(11)2021 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-34072450

RESUMEN

Obesity currently represents a major societal and health challenge worldwide. Its prevalence has reached epidemic proportions and trends continue to rise, reflecting the need for more effective preventive measures. Hypothalamic circuits that control energy homeostasis in response to food intake are interesting targets for body-weight management, for example, through interventions that reinforce the gut-to-brain nutrient signalling, whose malfunction contributes to obesity. Gut microbiota-diet interactions might interfere in nutrient sensing and signalling from the gut to the brain, where the information is processed to control energy homeostasis. This gut microbiota-brain crosstalk is mediated by metabolites, mainly short chain fatty acids, secondary bile acids or amino acids-derived metabolites and subcellular bacterial components. These activate gut-endocrine and/or neural-mediated pathways or pass to systemic circulation and then reach the brain. Feeding time and dietary composition are the main drivers of the gut microbiota structure and function. Therefore, aberrant feeding patterns or unhealthy diets might alter gut microbiota-diet interactions and modify nutrient availability and/or microbial ligands transmitting information from the gut to the brain in response to food intake, thus impairing energy homeostasis. Herein, we update the scientific evidence supporting that gut microbiota is a source of novel dietary and non-dietary biological products that may beneficially regulate gut-to-brain communication and, thus, improve metabolic health. Additionally, we evaluate how the feeding time and dietary composition modulate the gut microbiota and, thereby, the intraluminal availability of these biological products with potential effects on energy homeostasis. The review also identifies knowledge gaps and the advances required to clinically apply microbiome-based strategies to improve the gut-brain axis function and, thus, combat obesity.


Asunto(s)
Encéfalo/fisiología , Metabolismo Energético , Microbioma Gastrointestinal/fisiología , Tracto Gastrointestinal/fisiología , Homeostasis , Microbiota/fisiología , Ritmo Circadiano , Dieta , Susceptibilidad a Enfermedades , Ingestión de Alimentos , Conducta Alimentaria , Humanos , Micronutrientes , Nutrientes , Obesidad/etiología , Obesidad/metabolismo
4.
Rev Endocr Metab Disord ; 21(3): 369-380, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32691288

RESUMEN

Numerous studies in humans and animal models describe disturbances of the gut microbial ecosystem associated with adiposity and hallmarks of the metabolic syndrome, including hepatic and cardiovascular diseases. The manipulation of the microbiome, which is largely influenced by the diet, appears as an innovative therapeutic tool to prevent or control obesity and related diseases. This review describes the impact of nutrients on the gut microbiota composition and/or function and when available, the consequences on host physiology. A special emphasis is made on the contribution of bacterial-derived metabolites in the regulation of key gut functions that may explain their systemic effect.


Asunto(s)
Dieta , Microbioma Gastrointestinal/fisiología , Obesidad/dietoterapia , Obesidad/microbiología , Animales , Factores de Riesgo Cardiometabólico , Enfermedades Cardiovasculares/dietoterapia , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestinos/efectos de los fármacos , Intestinos/microbiología , Síndrome Metabólico/dietoterapia , Síndrome Metabólico/etiología , Síndrome Metabólico/microbiología , Nutrientes/farmacología , Obesidad/complicaciones
5.
FASEB J ; : fj201800544, 2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29906245

RESUMEN

The gut microbiota regulates key hepatic functions, notably through the production of bacterial metabolites that are transported via the portal circulation. We evaluated the effects of metabolites produced by the gut microbiota from aromatic amino acids (phenylacetate, benzoate, p-cresol, and indole) on liver inflammation induced by bacterial endotoxin. Precision-cut liver slices prepared from control mice, Kupffer cell (KC)-depleted mice, and obese mice ( ob/ ob) were treated with or without LPS and bacterial metabolites. We observed beneficial effects of indole that dose-dependently reduced the LPS-induced up-regulation of proinflammatory mediators at both mRNA and protein levels in precision-cut liver slices prepared from control or ob/ ob mice. KC depletion partly prevented the antiinflammatory effects of indole, notably through a reduction of nucleotide-binding domain and leucine-rich repeat containing (NLR) family pyrin domain-containing 3 (NLRP3) pathway activation. In vivo, the oral administration of indole before an LPS injection reduced the expression of key proteins of the NF-κB pathway and downstream proinflammatory gene up-regulation. Indole also prevented LPS-induced alterations of cholesterol metabolism through a transcriptional regulation associated with increased 4ß-hydroxycholesterol hepatic levels. In summary, indole appears as a bacterial metabolite produced from tryptophan that is able to counteract the detrimental effects of LPS in the liver. Indole could be a new target to develop innovative strategies to decrease hepatic inflammation.-Beaumont, M., Neyrinck, A. M., Olivares, M., Rodriguez, J., de Rocca Serra, A., Roumain, M., Bindels, L. B., Cani, P. D., Evenepoel, P., Muccioli, G. G., Demoulin, J.-B., Delzenne, N. M. The gut microbiota metabolite indole alleviates liver inflammation in mice.

6.
Nutr Neurosci ; 22(12): 877-893, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29697017

RESUMEN

Objectives: The biological mechanisms linking diet-related obesity and depression remain unclear. Therefore, we examined the impact of high-fat diet (HFD) on murine behaviour, intestinal microbiome, brain metabolome, neuropeptide Y (NPY) expression, and dipeptidyl peptidase-4 (DPP-4) activity.Methods: Male C57Bl/6J mice were fed an HFD (60 kJ% from fat) or control diet (12 kJ% from fat) for 8 weeks, followed by behavioural phenotyping. Caecal microbiome was analysed by 16S rDNA sequencing, brain metabolome by 1H nuclear magnetic resonance, NPY expression by PCR and immunoassay, and dipeptidyl peptidase-4 (DPP-4) activity by enzymatic assay. The effect of a 4-week treatment with imipramine (7 mg/kg/day) and the DPP-4 inhibitor sitagliptin (50 mg/kg/day) on HFD-induced behavioural changes was also tested.Results: HFD led to a depression-like phenotype as revealed by reduced sociability and sucrose preference. In the caecum, HFD diminished the relative abundance of Bacteroidetes and increased the relative abundance of Firmicutes and Cyanobacteria. In the brain, HFD modified the metabolome of prefrontal cortex and striatum, changing the relative concentrations of molecules involved in energy metabolism (e.g. lactate) and neuronal signalling (e.g. γ-aminobutyric acid). The expression of NPY in hypothalamus and hippocampus was decreased by HFD, whereas plasma NPY and DPP-4-like activity were increased. The HFD-induced anhedonia remained unaltered by imipramine and sitagliptin.Discussion: The depression-like behaviour induced by prolonged HFD in mice is associated with distinct alterations of intestinal microbiome, brain metabolome, NPY system, and DPP-4-like activity. Importantly, the HFD-evoked behavioural disturbance remains unaltered by DPP-4 inhibition and antidepressant treatment with imipramine.


Asunto(s)
Encéfalo/metabolismo , Depresión/etiología , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/fisiología , Metaboloma/fisiología , Neuropéptido Y/metabolismo , Animales , Conducta Animal/fisiología , Cuerpo Estriado/metabolismo , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptido Y/sangre , Neuropéptido Y/genética , Corteza Prefrontal/metabolismo , Aumento de Peso
7.
Diabetologia ; 61(8): 1838-1848, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29797022

RESUMEN

AIMS/HYPOTHESIS: Dipeptidyl peptidase 4 (DPP-4) inhibitors are agents designed to increase the half-life of incretins. Although they are administered orally, little is known about their effects on the gut microbiota and functions, despite the fact that some bacteria present in the gut microbiota exhibit DPP-4-like activity. Our objective was to study the impact of the DPP-4 inhibitor vildagliptin on gut functions and the intestinal ecosystem in a murine model of obesity induced by a Western diet (WD). METHODS: Twenty seven male C57BL/6J mice were randomised to receive a control diet, a WD (45% kJ from fat and 17% kJ from sucrose) or a WD + vildagliptin (0.6 mg/ml in drinking water) for 8 weeks. RESULTS: Vildagliptin significantly reduced DPP-4 activity in the caecal content and faeces. Vildagliptin impacted on the composition of the gut microbiota and its metabolic activity. It mainly decreased Oscillibacter spp. (a direct effect independent of DPP-4 activity was shown on cultured O. valericigenes), increased Lactobacillus spp. and propionate, and reduced the ligands of Toll-like receptors 2 and 4. Vildagliptin protected against the reductions in crypt depth and ileal expression of antimicrobial peptides induced by the WD. In the liver, the expression of immune cell populations (Cd3g and Cd11c [also known as Itgax]) and cytokines was decreased in the WD + vildagliptin-fed mice compared with the WD-fed group. Ex vivo exposure of precision-cut liver slices to vildagliptin showed that this response was not related to a direct effect of the drug on the liver tissue. CONCLUSIONS/INTERPRETATION: Our study is the first to consider the DPP-4-like activity of the gut microbiota as a target of DPP-4 inhibition. We propose that vildagliptin exerts beneficial effects at the intestinal level in association with modulation of gut microbiota, with consequences for hepatic immunity. If relevant in humans, this could open new therapeutic uses of DPP-4 inhibition to tackle gut dysfunctions in different pathophysiological contexts. DATA AVAILABILITY: The sequences used for analysis can be found in the MG-RAST database under the project name MYNEWGUT3.


Asunto(s)
Dieta Occidental/efectos adversos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Microbioma Gastrointestinal , Intestinos/efectos de los fármacos , Vildagliptina/farmacología , Animales , Citocinas/metabolismo , Ecosistema , Ácidos Grasos Volátiles/metabolismo , Perfilación de la Expresión Génica , Homeostasis/efectos de los fármacos , Hipoglucemiantes/farmacología , Inflamación/metabolismo , Intestinos/microbiología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo
9.
Stroke ; 48(5): 1147-1153, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28411264

RESUMEN

BACKGROUND AND PURPOSE: Vascular recurrence occurs in 11% of patients during the first year after ischemic stroke (IS) or transient ischemic attack. Clinical scores do not predict the whole vascular recurrence risk; therefore, we aimed to find genetic variants associated with recurrence that might improve the clinical predictive models in IS. METHODS: We analyzed 256 polymorphisms from 115 candidate genes in 3 patient cohorts comprising 4482 IS or transient ischemic attack patients. The discovery cohort was prospectively recruited and included 1494 patients, 6.2% of them developed a new IS during the first year of follow-up. Replication analysis was performed in 2988 patients using SNPlex or HumanOmni1-Quad technology. We generated a predictive model using Cox regression (GRECOS score [Genotyping Reurrence Risk of Stroke]) and generated risk groups using a classification tree method. RESULTS: The analyses revealed that rs1800801 in the MGP gene (hazard ratio, 1.33; P=9×10-03), a gene related to artery calcification, was associated with new IS during the first year of follow-up. This polymorphism was replicated in a Spanish cohort (n=1.305); however, it was not significantly associated in a North American cohort (n=1.683). The GRECOS score predicted new IS (P=3.2×10-09) and could classify patients, from low risk of stroke recurrence (1.9%) to high risk (12.6%). Moreover, the addition of genetic risk factors to the GRECOS score improves the prediction compared with previous Stroke Prognosis Instrument-II score (P=0.03). CONCLUSIONS: The use of genetics could be useful to estimate vascular recurrence risk after IS. Genetic variability in the MGP gene was associated with vascular recurrence in the Spanish population.


Asunto(s)
Isquemia Encefálica/genética , Enfermedades Cardiovasculares/genética , Accidente Cerebrovascular/genética , Anciano , Isquemia Encefálica/diagnóstico , Enfermedades Cardiovasculares/diagnóstico , Estudios de Cohortes , Femenino , Genotipo , Humanos , Ataque Isquémico Transitorio/diagnóstico , Ataque Isquémico Transitorio/genética , Masculino , América del Norte , Polimorfismo de Nucleótido Simple , Pronóstico , Recurrencia , Riesgo , Escocia , España , Accidente Cerebrovascular/diagnóstico
10.
Pediatr Res ; 77(1-2): 236-44, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25314581

RESUMEN

The gut microbiota structure, dynamics, and function result from interactions with environmental and host factors, which jointly influence the communication between the gut and peripheral tissues, thereby contributing to health programming and disease risk. Incidence of both type-1 and type-2 diabetes has increased during the past decades, suggesting that there have been changes in the interactions between predisposing genetic and environmental factors. Animal studies show that gut microbiota and its genome (microbiome) influence alterations in energy balance (increased energy harvest) and immunity (inflammation and autoimmunity), leading to metabolic dysfunction (e.g., insulin resistance and deficiency). Thus, although they have different origins, both disorders are linked by the association of the gut microbiota with the immune-metabolic axis. Human studies have also revealed shifts in microbiome signatures in diseased subjects as compared with controls, and a few of them precede the development of these disorders. These studies contribute to pinpointing specific microbiome components and functions (e.g., butyrate-producing bacteria) that can protect against both disorders. These could exert protective roles by strengthening gut barrier function and regulating inflammation, as alterations in these are a pathophysiological feature of both disorders, constituting common targets for future preventive approaches.


Asunto(s)
Diabetes Mellitus/epidemiología , Diabetes Mellitus/microbiología , Tracto Gastrointestinal/microbiología , Microbiota , Modelos Biológicos , Obesidad/epidemiología , Obesidad/microbiología , Humanos , Factores de Riesgo
11.
Eur J Nutr ; 54(1): 119-28, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24700375

RESUMEN

PURPOSE: To investigate whether breast-milk composition and microbiota differ in healthy mothers and mothers with celiac disease (CD) to ultimately contribute to identify additional factors determining CD risk. METHODS: Breast-milk samples from healthy mothers (n = 12) and mothers with CD (n = 12) were collected. Cytokines and secretory immunoglobulin A (sIgA) were analyzed by bead-arrays and flow cytometry and human milk oligosaccharides (HMOs) were assessed by capillary electrophoresis with laser-induced fluorescence (CE-LIF) detection. Breast-milk microbiota composition was analyzed by conventional and quantitative real-time PCR. RESULT: Breast milk from CD mothers showed significantly lower levels of interleukin (IL) 12p70 (P < 0.042), transforming growth factor (TGF)-ß1 (P < 0.018) and sIgA (P < 0.003) and almost significantly lower levels of interferon (IFN)-γ (P < 0.058). Six mothers in each group belonged to the secretor Le(a-b+) type, one to the secretor Le(a-b-) type and five to the non-secretor Le(a+b-) type. CD mothers of non-secretor Le(a+b-) type showed increased Lacto-N-tetraose content (P < 0.042) compared with healthy mothers. CD mothers' milk showed reduced gene copy numbers of Bifidobacterium spp. (P < 0.026) and B. fragilis group (P < 0.044). CONCLUSION: CD mothers' breast milk is characterized by a reduced abundance of immunoprotective compounds (TGF-ß1 and sIgA) and bifidobacteria. The reduction in these components could theoretically diminish the protective effects of breast-feeding on the child's future risk of developing CD.


Asunto(s)
Bacteroides fragilis/aislamiento & purificación , Bifidobacterium/aislamiento & purificación , Enfermedad Celíaca/metabolismo , Citocinas/análisis , Inmunoglobulina A Secretora/análisis , Leche Humana/química , Oligosacáridos/análisis , Adulto , Bacteroides fragilis/clasificación , Bacteroides fragilis/genética , Bacteroides fragilis/crecimiento & desarrollo , Bifidobacterium/clasificación , Bifidobacterium/genética , Bifidobacterium/crecimiento & desarrollo , Estudios de Casos y Controles , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Citocinas/metabolismo , Dieta Sin Gluten , Salud de la Familia , Femenino , Dosificación de Gen , Genes Bacterianos , Humanos , Inmunoglobulina A Secretora/metabolismo , Interferón gamma/análisis , Interferón gamma/metabolismo , Interleucina-12/análisis , Interleucina-12/metabolismo , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Leche Humana/microbiología , Tipificación Molecular , Oligosacáridos/metabolismo , Factor de Crecimiento Transformador beta1/análisis , Factor de Crecimiento Transformador beta1/metabolismo
12.
Br J Nutr ; 112(1): 30-40, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24774670

RESUMEN

Interactions between the immune system and the intestinal microbiota may play a role in coeliac disease (CD). In the present study, the potential effects of Bifidobacterium longum CECT 7347 in children with newly diagnosed CD were evaluated. A double-blind, randomised, placebo-controlled trial was conducted in thirty-three children who received a capsule containing either B. longum CECT 7347 (109 colony-forming units) or placebo (excipients) daily for 3 months together with a gluten-free diet (GFD). Outcome measures (baseline and post-intervention) included immune phenotype of peripheral blood cells, serum cytokine concentration, faecal secretory IgA (sIgA) content, anthropometric parameters and intestinal microbiota composition. Comparisons between the groups revealed greater height percentile increases (P= 0·048) in the B. longum CECT 7347 group than in the placebo group, as well as decreased peripheral CD3⁺ T lymphocytes (P= 0·004) and slightly reduced TNF-α concentration (P= 0·067). Within-group comparisons of baseline and final values did not reveal any differences in T lymphocytes and cytokines in the placebo group, while decreased CD3⁺ (P =0·013) and human leucocyte antigen (HLA)-DR⁺ T lymphocytes (P =0·029) and slightly reduced TNF-α concentration (P= 0·085) were detected in the B. longum CECT 7347 group. Comparison between the groups showed that the administration of B. longum CECT 7347 reduced the numbers of the Bacteroides fragilis group (P= 0·020) and the content of sIgA in stools (P= 0·011) compared with the administration of placebo. Although this is a first exploratory intervention with limitations, the findings suggest that B. longum CECT 7347 could help improve the health status of CD patients who tend to show alterations in gut microbiota composition and a biased immune response even on a GFD.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Bacteroides fragilis/inmunología , Bifidobacterium , Enfermedad Celíaca/dietoterapia , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Probióticos/uso terapéutico , Bacteroides fragilis/crecimiento & desarrollo , Bacteroides fragilis/aislamiento & purificación , Bifidobacterium/inmunología , Enfermedad Celíaca/sangre , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Niño , Desarrollo Infantil , Preescolar , Terapia Combinada , Citocinas/sangre , Dieta Sin Gluten , Método Doble Ciego , Heces/química , Heces/microbiología , Femenino , Bacterias Gramnegativas/crecimiento & desarrollo , Bacterias Gramnegativas/inmunología , Bacterias Gramnegativas/aislamiento & purificación , Bacterias Grampositivas/crecimiento & desarrollo , Bacterias Grampositivas/inmunología , Bacterias Grampositivas/aislamiento & purificación , Humanos , Inmunoglobulina A Secretora/análisis , Inmunoglobulina A Secretora/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Viabilidad Microbiana
13.
Genome Biol ; 25(1): 174, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38961511

RESUMEN

BACKGROUND: The gut microbiota controls broad aspects of human metabolism and feeding behavior, but the basis for this control remains largely unclear. Given the key role of human dipeptidyl peptidase 4 (DPP4) in host metabolism, we investigate whether microbiota DPP4-like counterparts perform the same function. RESULTS: We identify novel functional homologs of human DPP4 in several bacterial species inhabiting the human gut, and specific associations between Parabacteroides and Porphyromonas DPP4-like genes and type 2 diabetes (T2D). We also find that the DPP4-like enzyme from the gut symbiont Parabacteroides merdae mimics the proteolytic activity of the human enzyme on peptide YY, neuropeptide Y, gastric inhibitory polypeptide (GIP), and glucagon-like peptide 1 (GLP-1) hormones in vitro. Importantly, administration of E. coli overexpressing the P. merdae DPP4-like enzyme to lipopolysaccharide-treated mice with impaired gut barrier function reduces active GIP and GLP-1 levels, which is attributed to increased DPP4 activity in the portal circulation and the cecal content. Finally, we observe that linagliptin, saxagliptin, sitagliptin, and vildagliptin, antidiabetic drugs with DPP4 inhibitory activity, differentially inhibit the activity of the DPP4-like enzyme from P. merdae. CONCLUSIONS: Our findings confirm that proteolytic enzymes produced by the gut microbiota are likely to contribute to the glucose metabolic dysfunction that underlies T2D by inactivating incretins, which might inspire the development of improved antidiabetic therapies.


Asunto(s)
Diabetes Mellitus Tipo 2 , Dipeptidil Peptidasa 4 , Microbioma Gastrointestinal , Incretinas , Diabetes Mellitus Tipo 2/microbiología , Diabetes Mellitus Tipo 2/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Animales , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Incretinas/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Masculino
14.
Br J Nutr ; 109 Suppl 2: S76-80, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23360883

RESUMEN

Intestinal microbiota may influence human physiology and disease risk due to the role it plays in mediating appropriate immune responses to harmful and innocuous antigens. Colonisation of the intestine in early life seems particularly important as it is the main environmental stimulus for immune system maturation. This is a dynamic process, which depends on both environmental and genetic factors. The pathogenesis of inflammatory bowel disease, such as Crohn's disease, involves genetic polymorphisms (e.g. CARD15/nucleotide-binding oligomerisation domain 2) related to an excessive inflammatory response to commensal microbiota and to its unbalanced composition. Atopic diseases have also been linked to imbalances in microbiota and to related genetic factors (e.g. TLR4 and CD14 genes), although these associations are still controversial. Research into the relationship between the genetic risk of developing celiac disease (human leukocyte antigen (HLA)-DQ2/DQ8) and the early colonisation process in infants at family risk of the disease has also reported that the HLA-DQ genotype could influence staphylococcal colonisation. Future observational studies considering both host genetics and microbiota could be critical in defining the complex host-microbe interactions and the respective role each plays in inflammatory disorders.


Asunto(s)
Enterocolitis/genética , Enterocolitis/microbiología , Predisposición Genética a la Enfermedad , Interacciones Huésped-Parásitos , Enfermedades Intestinales/genética , Enfermedades Intestinales/microbiología , Mucosa Intestinal/microbiología , Animales , Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/microbiología , Enterocolitis/inmunología , Enterocolitis/metabolismo , Genotipo , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/metabolismo , Interacciones Huésped-Patógeno , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Intestinales/inmunología , Enfermedades Intestinales/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo
15.
Br J Nutr ; 110(10): 1828-36, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23657133

RESUMEN

Coeliac disease is an autoimmune disorder triggered by gluten intake, causing intestinal inflammation and mucosal damage commonly associated with the malabsorption of nutrients and ferropenic anaemia. The present study evaluates the effects of the oral administration of Bifidobacterium longum CECT 7347 on gliadin-mediated alterations in hepatic Fe deposition and Hb concentration, liver transferrin receptor (TfR)-2, IL-6, TNF-α and hepcidin (Hamp) expression (mRNA), and active hepcidin peptide production by liquid chromatography­MS/MS. Weanling rats, sensitised or not with interferon (IFN)-γ, were fed with gliadins and/or the bifidobacterial strain. Gliadin feeding increased hepatic Fe deposition; however, only gliadin-fed sensitised animals showed lower Hb concentrations than the controls. TfR2 expression decreased after gliadins were fed to both sensitised and non-sensitised animals,and restored by the administration of B. longum. These observations were accompanied by increases in IL-6 expression levels in all the treatment groups; however, TNF-α expression only increased significantly in animals fed gliadins alone or together with B. longum if they had previously been sensitised with IFN-γ. Liver expression levels of Hamp diminished in all cases to the lowest values in animals sensitised with IFN-γ after being fed with gliadins and/or bifidobacteria. In these animals, plasma Hamp active peptide concentrations significantly increased when compared with the controls. Significant correlations were calculated between Hamp expression and liver Fe contents (liver Fe = 1/0·0032 + 0·032 x Hamp(exp)), and Hb concentrations (Hb = 11·49 + 10·13 x (Hamp(exp))1/2). These data indicate that oral administration of B. longum ameliorates gliadin-mediated perturbations in liver Fe deposition and mobilisation.


Asunto(s)
Anemia/tratamiento farmacológico , Bifidobacterium , Enfermedad Celíaca/tratamiento farmacológico , Gliadina/efectos adversos , Hierro/metabolismo , Hígado/metabolismo , Probióticos/uso terapéutico , Administración Oral , Anemia/etiología , Anemia/metabolismo , Animales , Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/metabolismo , Femenino , Hemoglobinas/metabolismo , Hepcidinas/metabolismo , Interferón gamma/farmacología , Interleucina-6/metabolismo , Síndromes de Malabsorción/tratamiento farmacológico , Síndromes de Malabsorción/etiología , Síndromes de Malabsorción/metabolismo , Ratas , Ratas Wistar , Receptores de Transferrina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
16.
BMC Neurol ; 13: 203, 2013 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-24341857

RESUMEN

BACKGROUND: The detection of early neuropsychological abnormalities as precursors of cognitive decline of vascular origin in patients with lacunar stroke is a subject of increasing interest. The objective of this study was to assess whether there were differences in the performance of a battery of neuropsychological tests in first-ever lacunar stroke patients with and without associated silent multiple lacunar infarctions found incidentally on the brain magnetic resonance imaging (MRI) scan. METHODS: A total of 72 consecutive patients with first-ever lacunar infarction were studied 1 month after stroke. All patients underwent a comprehensive neuropsychological evaluation, which included the California Verbal Learning Test (CVLT), Phonetic Verbal Fluency Test (PMR), Semantic Verbal Fluency Test (category "animals"), Digit Span Forward and Backward from the Wechsler Adult Intelligence Scale (WAIS-III), and Mini-Mental State Examination (MMSE). RESULTS: A total of 38 patients (52.7%) had silent multiple lacunar infarcts, with corona radiata as the most frequent topography (P < 0.023). White matter hyperintensities (leukoaraiosis) were observed in 81.1% of patients with silent multiple lacunar infarcts and in 50% with a single lacunar infarction (P < 0.007). Patients in both groups showed similar scores in the MMSE, but those with associated silent lacunar infarctions showed a poorer performance in the semantic fluency test (P < 0.008) and in short delayed verbal memory (P < 0.001). In both cases, however, leukoaraiosis was not statistically significant in multivariate linear regression models adjusted by confounding covariates. In these models, multiple silent lacunar infarctions and education were independent predictors of poor performance in the semantic fluency test and in short delayed verbal memory. CONCLUSIONS: The presence of silent multiple lacunar infarctions documented on brain MRI scans in patients with first-ever lacunar stroke was associated with mild neuropsychological abnormalities, particularly in the performance of executive functions (semantic fluency) and short delayed verbal memory. According to these findings, in the initial stages of small vessel disease, mild neuropsychological abnormalities appear to be related to lacunes rather than to leukoaraiosis or perivascular hyperintensities of vascular cause.


Asunto(s)
Trastornos del Conocimiento/etiología , Accidente Vascular Cerebral Lacunar/complicaciones , Anciano , Anciano de 80 o más Años , Trastornos del Conocimiento/diagnóstico , Femenino , Humanos , Modelos Lineales , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Estudios Retrospectivos , Accidente Vascular Cerebral Lacunar/patología
17.
Gut Microbes ; 15(1): 2181928, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36823075

RESUMEN

Hypercaloric diets overactivate the intestinal immune system and disrupt the microbiome and epithelial cell functions, impairing glucose metabolism. The origins of this inflammatory cascade are poorly characterized. We investigated the involvement of intestinal proinflammatory group 1 innate lymphoid cells (ILC1s) in obesity progression and metabolic disruption. In obese mice, we studied longitudinally the ILC1s response to the diet and ILC1s depletion to address its role in obesity. ILC1s are required for the expansion of pro-inflammatory macrophages and ILC2s. ILC1s depletion induced the ILC3-IL-22 pathway, increasing mucin production, antimicrobial peptides, and neuroendocrine cells. These changes were translated into higher gut hormones and reduced insulinemia and adiposity. ILC1s depletion was also associated with a bloom in Akkermansia muciniphila and decreases in Bilophila spp. Intestinal-ILC1s are upstream activators of inflammatory signals, connecting immunity with the microbiome, the enteroendocrine system, and the intestinal barrier in the control of glucose metabolism and adiposity.


Asunto(s)
Microbioma Gastrointestinal , Inmunidad Innata , Ratones , Animales , Linfocitos/metabolismo , Obesidad/metabolismo , Inflamación , Macrófagos/metabolismo , Glucosa
18.
Microbiol Mol Biol Rev ; 87(3): e0021222, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37367231

RESUMEN

Microbiomes have highly important roles for ecosystem functioning and carry out key functions that support planetary health, including nutrient cycling, climate regulation, and water filtration. Microbiomes are also intimately associated with complex multicellular organisms such as humans, other animals, plants, and insects and perform crucial roles for the health of their hosts. Although we are starting to understand that microbiomes in different systems are interconnected, there is still a poor understanding of microbiome transfer and connectivity. In this review we show how microbiomes are connected within and transferred between different habitats and discuss the functional consequences of these connections. Microbiome transfer occurs between and within abiotic (e.g., air, soil, and water) and biotic environments, and can either be mediated through different vectors (e.g., insects or food) or direct interactions. Such transfer processes may also include the transmission of pathogens or antibiotic resistance genes. However, here, we highlight the fact that microbiome transmission can have positive effects on planetary and human health, where transmitted microorganisms potentially providing novel functions may be important for the adaptation of ecosystems.


Asunto(s)
Microbiota , Planetas , Animales , Humanos , Microbiología del Suelo , Microbiota/fisiología , Suelo , Agua
19.
Clin Dev Immunol ; 2012: 654143, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23008734

RESUMEN

Celiac disease (CD) is an immune-mediated enteropathy, triggered by dietary wheat gluten and similar proteins of barley and rye in genetically susceptible individuals. The etiology of this disorder is complex, involving both environmental and genetic factors. The major genetic risk factor for CD is represented by HLA-DQ genes, which account for approximately 40% of the genetic risk; however, only a small percentage of carriers develop the disease. Gluten is the main environmental factor responsible for the signs and symptoms of the disease, but exposure to gluten does not fully explain the manifestation of CD. Epidemiological and clinical data suggest that environmental factors other than gluten might play a role in disease development, including early feeding practices (e.g., breast milk versus formula and duration of breastfeeding), infections, and alterations in the intestinal microbiota composition. Herein, we review what is known about the influence of dietary factors, exposure to infectious agents, and intestinal microbiota composition, particularly in early life, on the risk of developing CD, as well as the possible dietary strategies to induce or increase gluten tolerance.


Asunto(s)
Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Dieta , Intestinos/inmunología , Intestinos/microbiología , Metagenoma/inmunología , Animales , Conducta Alimentaria , Humanos , Factores de Riesgo
20.
Nutrients ; 14(3)2022 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-35276798

RESUMEN

Allium species and their organosulfur-derived compounds could prevent obesity and metabolic dysfunction, as they exhibit immunomodulatory and antimicrobial properties. Here, we report the anti-obesogenic potential and dose-dependent effects (0.1 or 1 mg/kg/day) of propyl propane thiosulfinate (PTS) in a murine model of diet-induced obesity. The obesogenic diet increased body weight gain and adipocyte size, and boosted inflammatory marker (Cd11c) expression in the adipose tissue. Conversely, PTS prevented these effects in a dose-dependent manner. Moreover, the higher dose of PTS improved glucose and hepatic homeostasis, modulated lipid metabolism, and raised markers of the thermogenic capacity of brown adipose tissue. In the colon, the obesogenic diet reduced IL-22 levels and increased gut barrier function markers (Cldn3, Muc2, Reg3g, DefaA); however, the highest PTS dose normalized all of these markers to the levels of mice fed a standard diet. Gut microbiota analyses revealed no differences in diversity indexes and only minor taxonomic changes, such as an increase in butyrate producers, Intestimonas and Alistipes, and a decrease in Bifidobacterium in mice receiving the highest PTS dose. In summary, our study provides preclinical evidence for the protective effects of PTS against obesity, which if confirmed in humans, might provide a novel plant-based dietary product to counteract this condition.


Asunto(s)
Allium , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/metabolismo , Propano/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA