Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Brain Behav Immun ; 119: 750-766, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38710336

RESUMEN

Chronic pain is a heavily debilitating condition and a huge socio-economic burden, with no efficient treatment. Over the past decade, the gut microbiota has emerged as an important regulator of nervous system's health and disease states. Yet, its contribution to the pathogenesis of chronic somatic pain remains poorly documented. Here, we report that male but not female mice lacking Myosin1a (KO) raised under single genotype housing conditions (KO-SGH) are predisposed to develop chronic pain in response to a peripheral tissue injury. We further underscore the potential of MYO1A loss-of-function to alter the composition of the gut microbiota and uncover a functional connection between the vulnerability to chronic pain and the dysbiotic gut microbiota of KO-SGH males. As such, parental antibiotic treatment modifies gut microbiota composition and completely rescues the injury-induced pain chronicity in male KO-SGH offspring. Furthermore, in KO-SGH males, this dysbiosis is accompanied by a transcriptomic activation signature in the dorsal root ganglia (DRG) macrophage compartment, in response to tissue injury. We identify CD206+CD163- and CD206+CD163+ as the main subsets of DRG resident macrophages and show that both are long-lived and self-maintained and exhibit the capacity to monitor the vasculature. Consistently, in vivo depletion of DRG macrophages rescues KO-SGH males from injury-induced chronic pain underscoring a deleterious role for DRG macrophages in a Myo1a-loss-of function context. Together, our findings reveal gene-sex-microbiota interactions in determining the predisposition to injury-induced chronic pain and point-out DRG macrophages as potential effector cells.


Asunto(s)
Dolor Crónico , Disbiosis , Ganglios Espinales , Microbioma Gastrointestinal , Ratones Noqueados , Miosina Tipo I , Animales , Femenino , Masculino , Ratones , Dolor Crónico/metabolismo , Dolor Crónico/microbiología , Disbiosis/metabolismo , Ganglios Espinales/metabolismo , Microbioma Gastrointestinal/fisiología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Miosina Tipo I/metabolismo
2.
Gut ; 72(5): 939-950, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36241390

RESUMEN

OBJECTIVES: Clinical studies revealed that early-life adverse events contribute to the development of IBS in adulthood. The aim of our study was to investigate the relationship between prenatal stress (PS), gut microbiota and visceral hypersensitivity with a focus on bacterial lipopeptides containing γ-aminobutyric acid (GABA). DESIGN: We developed a model of PS in mice and evaluated, in adult offspring, visceral hypersensitivity to colorectal distension (CRD), colon inflammation, barrier function and gut microbiota taxonomy. We quantified the production of lipopeptides containing GABA by mass spectrometry in a specific strain of bacteria decreased in PS, in PS mouse colons, and in faeces of patients with IBS and healthy volunteers (HVs). Finally, we assessed their effect on PS-induced visceral hypersensitivity. RESULTS: Prenatally stressed mice of both sexes presented visceral hypersensitivity, no overt colon inflammation or barrier dysfunction but a gut microbiota dysbiosis. The dysbiosis was distinguished by a decreased abundance of Ligilactobacillus murinus, in both sexes, inversely correlated with visceral hypersensitivity to CRD in mice. An isolate from this bacterial species produced several lipopeptides containing GABA including C14AsnGABA. Interestingly, intracolonic treatment with C14AsnGABA decreased the visceral sensitivity of PS mice to CRD. The concentration of C16LeuGABA, a lipopeptide which inhibited sensory neurons activation, was decreased in faeces of patients with IBS compared with HVs. CONCLUSION: PS impacts the gut microbiota composition and metabolic function in adulthood. The reduced capacity of the gut microbiota to produce GABA lipopeptides could be one of the mechanisms linking PS and visceral hypersensitivity in adulthood.


Asunto(s)
Microbioma Gastrointestinal , Síndrome del Colon Irritable , Masculino , Femenino , Ratones , Animales , Síndrome del Colon Irritable/microbiología , Disbiosis , Heces/microbiología , Inflamación
3.
J Transl Med ; 20(1): 111, 2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35255932

RESUMEN

The human gastrointestinal tract is inhabited by the largest microbial community within the human body consisting of trillions of microbes called gut microbiota. The normal flora is the site of many physiological functions such as enhancing the host immunity, participating in the nutrient absorption and protecting the body against pathogenic microorganisms. Numerous investigations showed a bidirectional interplay between gut microbiota and many organs within the human body such as the intestines, the lungs, the brain, and the skin. Large body of evidence demonstrated, more than a decade ago, that the gut microbial alteration is a key factor in the pathogenesis of many local and systemic disorders. In this regard, a deep understanding of the mechanisms involved in the gut microbial symbiosis/dysbiosis is crucial for the clinical and health field. We review the most recent studies on the involvement of gut microbiota in the pathogenesis of many diseases. We also elaborate the different strategies used to manipulate the gut microbiota in the prevention and treatment of disorders. The future of medicine is strongly related to the quality of our microbiota. Targeting microbiota dysbiosis will be a huge challenge.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Probióticos , Disbiosis/terapia , Tracto Gastrointestinal , Humanos , Prebióticos , Probióticos/uso terapéutico
4.
Rev Endocr Metab Disord ; 20(4): 449-459, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31741266

RESUMEN

It has recently become evident that the periodontium (gingiva, desmodontal ligament, cementum and alveolar bone) and the associated microbiota play a pivotal role in regulating human health and diseases. The oral cavity is the second largest microbiota in the body with around 500 different bacterial species identified today. When disruption of oral cavity and dysbiosis occur, the proportion of strict anaerobic Gram-negative bacteria is then increased. Patients with periodontitis present 27 to 53% more risk to develop diabetes than the control population suggesting that periodontitis is an aggravating factor in the incidence of diabetes. Moreover, dysbiosis of oral microbiota is involved in both periodontal and metabolic disorders (cardiovascular diseases, dyslipidaemia …). The oral diabetic dysbiosis is characterized by a specific bacteria Porphyromonas, which is highly expressed in periodontal diseases and could exacerbate insulin resistance. In this review, we will address the nature of the oral microbiota and how it affects systemic pathologies with a bidirectional interaction. We also propose that using prebiotics like Akkermansia muciniphila may influence oral microbiota as novel therapeutic strategies. The discovery of the implication of oral microbiota for the control of metabolic diseases could be a new way for personalized medicine.


Asunto(s)
Enfermedades Metabólicas/microbiología , Boca/microbiología , Periodontitis/microbiología , Animales , Humanos , Enfermedades Metabólicas/metabolismo , Boca/metabolismo , Periodontitis/metabolismo , Factores de Riesgo
5.
Mol Syst Biol ; 13(3): 921, 2017 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-28302863

RESUMEN

Gut microbiota dysbiosis has been implicated in a variety of systemic disorders, notably metabolic diseases including obesity and impaired liver function, but the underlying mechanisms are uncertain. To investigate this question, we transferred caecal microbiota from either obese or lean mice to antibiotic-free, conventional wild-type mice. We found that transferring obese-mouse gut microbiota to mice on normal chow (NC) acutely reduces markers of hepatic gluconeogenesis with decreased hepatic PEPCK activity, compared to non-inoculated mice, a phenotypic trait blunted in conventional NOD2 KO mice. Furthermore, transferring of obese-mouse microbiota changes both the gut microbiota and the microbiome of recipient mice. We also found that transferring obese gut microbiota to NC-fed mice then fed with a high-fat diet (HFD) acutely impacts hepatic metabolism and prevents HFD-increased hepatic gluconeogenesis compared to non-inoculated mice. Moreover, the recipient mice exhibit reduced hepatic PEPCK and G6Pase activity, fed glycaemia and adiposity. Conversely, transfer of lean-mouse microbiota does not affect markers of hepatic gluconeogenesis. Our findings provide a new perspective on gut microbiota dysbiosis, potentially useful to better understand the aetiology of metabolic diseases.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/fisiología , Hígado/metabolismo , Obesidad/microbiología , Animales , Disbiosis , Gluconeogénesis , Glucosa-6-Fosfatasa/genética , Ratones , Ratones Endogámicos C57BL , Obesidad/inducido químicamente , Obesidad/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética
6.
Gut ; 66(5): 872-885, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-26838600

RESUMEN

OBJECTIVE: To identify a causal mechanism responsible for the enhancement of insulin resistance and hyperglycaemia following periodontitis in mice fed a fat-enriched diet. DESIGN: We set-up a unique animal model of periodontitis in C57Bl/6 female mice by infecting the periodontal tissue with specific and alive pathogens like Porphyromonas gingivalis (Pg), Fusobacterium nucleatum and Prevotella intermedia. The mice were then fed with a diabetogenic/non-obesogenic fat-enriched diet for up to 3 months. Alveolar bone loss, periodontal microbiota dysbiosis and features of glucose metabolism were quantified. Eventually, adoptive transfer of cervical (regional) and systemic immune cells was performed to demonstrate the causal role of the cervical immune system. RESULTS: Periodontitis induced a periodontal microbiota dysbiosis without mainly affecting gut microbiota. The disease concomitantly impacted on the regional and systemic immune response impairing glucose metabolism. The transfer of cervical lymph-node cells from infected mice to naive recipients guarded against periodontitis-aggravated metabolic disease. A treatment with inactivated Pg prior to the periodontal infection induced specific antibodies against Pg and protected the mouse from periodontitis-induced dysmetabolism. Finally, a 1-month subcutaneous chronic infusion of low rates of lipopolysaccharides from Pg mimicked the impact of periodontitis on immune and metabolic parameters. CONCLUSIONS: We identified that insulin resistance in the high-fat fed mouse is enhanced by pathogen-induced periodontitis. This is caused by an adaptive immune response specifically directed against pathogens and associated with a periodontal dysbiosis.


Asunto(s)
Inmunidad Adaptativa , Infecciones por Bacteroidaceae/complicaciones , Disbiosis/inmunología , Resistencia a la Insulina/inmunología , Periodontitis/inmunología , Periodontitis/prevención & control , Porphyromonas gingivalis , Animales , Trasplante de Células , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Disbiosis/microbiología , Disbiosis/prevención & control , Femenino , Encía/microbiología , Hiperglucemia/inmunología , Hiperglucemia/microbiología , Interferón gamma/sangre , Interleucina-6/sangre , Lipopolisacáridos/inmunología , Ganglios Linfáticos/citología , Linfocitos , Ratones , Ratones Endogámicos C57BL , Microbiota , Periodontitis/microbiología , Periodontitis/patología , Porphyromonas gingivalis/inmunología , Distribución Aleatoria , Bazo/citología , Vacunación
7.
Diabetologia ; 60(4): 690-700, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28105518

RESUMEN

AIMS/HYPOTHESIS: Despite the current pandemic of metabolic diseases, our understanding of the diverse nature of the development of metabolic alterations in people who eat a high-fat diet (HFD) is still poor. We recently demonstrated a cardio-metabolic adaptation in mice fed an HFD, which was characterised by a specific gut and periodontal microbiota profile. Since the severity of hepatic disease is characterised by specific microRNA (miRNA) signatures and the gut microbiota is a key driver of both hepatic disease and miRNA expression, we analysed the expression of three hepatic miRNA and studied their correlation with hepatic triacylglycerol content and gut microbiota. METHODS: Two cohorts of C57BL/6 4-week-old wild-type (WT) male mice (n = 62 and n = 96) were fed an HFD for 3 months to provide a model of metabolic adaptation. Additionally 8-week-old C57BL/6 mice, either WT or of different genotypes, with diverse gut microbiota (ob/ob, Nod1, Cd14 knockout [Cd14KO] and Nod2) or without gut microbiota (axenic mice) were fed a normal chow diet. Following which, glycaemic index, body weight, blood glucose levels and hepatic triacylglycerol levels were measured. Gut (caecum) microbiota taxa were analysed by pyrosequencing. To analyse hepatic miRNA expression, real-time PCR was performed on total extracted miRNA samples. Data were analysed using two-way ANOVA followed by the Dunnett's post hoc test, or by the unpaired Student's t test. A cluster analysis and multivariate analyses were also performed. RESULTS: Our results demonstrated that the expression of miR-181a, miR-666 and miR-21 in primary murine hepatocytes is controlled by lipopolysaccharide in a dose-dependent manner. Of the gut microbiota, Firmicutes were positively correlated and Proteobacteria and Bacteroides acidifaciens were negatively correlated with liver triacylglycerol levels. Furthermore, the relative abundance of Firmicutes was negatively correlated with hepatic expression of miR-666 and miR-21. In contrast, the relative abundance of B. acidifaciens was positively correlated with miR-21. CONCLUSIONS/INTERPRETATION: We propose the involvement of hepatic miRNA, liver triacylglycerols and gut microbiota as a new triad that underlies the molecular mechanisms by which gut microbiota governs hepatic pathophysiology during metabolic adaptation to HFD.


Asunto(s)
Hígado/metabolismo , MicroARNs/metabolismo , Triglicéridos/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Genotipo , Hepatocitos/metabolismo , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Reacción en Cadena de la Polimerasa
8.
Hepatology ; 64(6): 2015-2027, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27639192

RESUMEN

The early detection of liver fibrosis among patients with nonalcoholic fatty liver disease (NAFLD) is an important clinical need. In view of the suggested role played by bacterial translocation in liver disease and obesity, we sought to investigate the relationship between blood microbiota and liver fibrosis (LF) in European cohorts of patients with severe obesity. We carried out a cross-sectional study of obese patients, well characterized with respect to the severity of the NAFLD, in the cohort FLORINASH. This cohort has been divided into a discovery cohort comprising 50 Spanish patients and then in a validation cohort of 71 Italian patients. Blood bacterial DNA was analyzed both quantitatively by 16S ribosomal DNA (rDNA) quantitative polymerase chain reaction and qualitatively by 16S rDNA targeted metagenomic sequencing and functional metagenome prediction. Spanish plasma bile acid contents were analyzed by liquid chromatography/mass spectrometry. The 16S rDNA concentration was significantly higher in patients of the discovery cohort with LF. By 16S sequencing, we found specific differences in the proportion of several bacterial taxa in both blood and feces that correlate with the presence of LF, thus defining a specific signature of the liver disease. Several secondary/primary bile acid ratios were also decreased with LF in the discovery cohort. We confirmed, in the validation cohort, the correlation between blood 16S rDNA concentration and LF, whereas we did not confirm the specific bacterial taxa signature, despite a similar trend in patients with more-severe fibrosis. CONCLUSION: Changes in blood microbiota are associated with LF in obese patients. Blood microbiota analysis provides potential biomarkers for the detection of LF in this population. (Hepatology 2016;64:2015-2027).


Asunto(s)
Heces/microbiología , Cirrosis Hepática/sangre , Cirrosis Hepática/complicaciones , Microbiota , Obesidad/sangre , Obesidad/complicaciones , Estudios Transversales , Femenino , Humanos , Cirrosis Hepática/microbiología , Masculino , Persona de Mediana Edad , Obesidad/microbiología , Proyectos Piloto
9.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G1091-101, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27033119

RESUMEN

Periodontitis and type 2 diabetes are connected pandemic diseases, and both are risk factors for cardiovascular complications. Nevertheless, the molecular factors relating these two chronic pathologies are poorly understood. We have shown that, in response to a long-term fat-enriched diet, mice present particular gut microbiota profiles related to three metabolic phenotypes: diabetic-resistant (DR), intermediate (Inter), and diabetic-sensitive (DS). Moreover, many studies suggest that a dysbiosis of periodontal microbiota could be associated with the incidence of metabolic and cardiac diseases. We investigated whether periodontitis together with the periodontal microbiota may also be associated with these different cardiometabolic phenotypes. We report that the severity of glucose intolerance is related to the severity of periodontitis and cardiac disorders. In detail, alveolar bone loss was more accentuated in DS than Inter, DR, and normal chow-fed mice. Molecular markers of periodontal inflammation, such as TNF-α and plasminogen activator inhibitor-1 mRNA levels, correlated positively with both alveolar bone loss and glycemic index. Furthermore, the periodontal microbiota of DR mice was dominated by the Streptococcaceae family of the phylum Firmicutes, whereas the periodontal microbiota of DS mice was characterized by increased Porphyromonadaceae and Prevotellaceae families. Moreover, in DS mice the periodontal microbiota was indicated by an abundance of the genera Prevotella and Tannerella, which are major periodontal pathogens. PICRUSt analysis of the periodontal microbiome highlighted that prenyltransferase pathways follow the cardiometabolic adaptation to a high-fat diet. Finally, DS mice displayed a worse cardiac phenotype, percentage of fractional shortening, heart rhythm, and left ventricle weight-to-tibia length ratio than Inter and DR mice. Together, our data show that periodontitis combined with particular periodontal microbiota and microbiome is associated with metabolic adaptation to a high-fat diet related to the severity of cardiometabolic alteration.


Asunto(s)
Adaptación Fisiológica , Enfermedades Cardiovasculares/metabolismo , Dieta Alta en Grasa , Intolerancia a la Glucosa , Microbiota , Periodontitis/microbiología , Función Ventricular , Animales , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/microbiología , Dimetilaliltranstransferasa/metabolismo , Disbiosis/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Periodontitis/complicaciones , Inhibidor 1 de Activador Plasminogénico/metabolismo , Prevotella/aislamiento & purificación , Streptococcaceae/aislamiento & purificación , Factor de Necrosis Tumoral alfa/metabolismo
10.
Pediatr Res ; 80(6): 777-784, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27490741

RESUMEN

BACKGROUND: The human microbiota is a modulator of the immune system. Variations in the placental microbiota could be related with pregnancy disorders. We profiled the placental microbiota and microbiome in women with gestational diabetes (GDM) and studied its relation to maternal metabolism and placental expression of anti-inflammatory cytokines. METHODS: Placental microbiota and microbiome and expression of anti-inflammatory cytokines (IL10, TIMP3, ITGAX, and MRC1MR) were analyzed in placentas from women with GDM and from control women. Fasting insulin, glucose, O'Sullivan glucose, lipids, and blood cell counts were assessed at second and third trimester of pregnancy. RESULTS: Bacteria belonging to the Pseudomonadales order and Acinetobacter genus showed lower relative abundance in women with GDM compared to control (P < 0.05). In GDM, lower abundance of placental Acinetobacter associated with a more adverse metabolic (higher O'Sullivan glucose) and inflammatory phenotype (lower blood eosinophil count and lower placental expression of IL10 and TIMP3) (P < 0.05 to P = 0.001). Calcium signaling pathway was increased in GDM placental microbiome. CONCLUSION: A distinct microbiota profile and microbiome is present in GDM. Acinetobacter has been recently shown to induce IL-10 in mice. GDM could constitute a state of placental microbiota-driven altered immunologic tolerance, making placental microbiota a new target for therapy in GDM.


Asunto(s)
Diabetes Gestacional/microbiología , Microbiota/genética , Microbiota/inmunología , Placenta/microbiología , Acinetobacter/genética , Acinetobacter/inmunología , Acinetobacter/aislamiento & purificación , Adulto , Señalización del Calcio , Estudios de Casos y Controles , Citocinas/genética , Citocinas/metabolismo , ADN Bacteriano/genética , Diabetes Gestacional/inmunología , Diabetes Gestacional/metabolismo , Femenino , Gammaproteobacteria/genética , Gammaproteobacteria/inmunología , Gammaproteobacteria/aislamiento & purificación , Expresión Génica , Humanos , Placenta/inmunología , Placenta/metabolismo , Embarazo , ARN Bacteriano/genética , ARN Ribosómico 16S/genética
11.
BMC Gastroenterol ; 14: 189, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25407511

RESUMEN

Data are accumulating that emphasize the important role of the intestinal barrier and intestinal permeability for health and disease. However, these terms are poorly defined, their assessment is a matter of debate, and their clinical significance is not clearly established. In the present review, current knowledge on mucosal barrier and its role in disease prevention and therapy is summarized. First, the relevant terms 'intestinal barrier' and 'intestinal permeability' are defined. Secondly, the key element of the intestinal barrier affecting permeability are described. This barrier represents a huge mucosal surface, where billions of bacteria face the largest immune system of our body. On the one hand, an intact intestinal barrier protects the human organism against invasion of microorganisms and toxins, on the other hand, this barrier must be open to absorb essential fluids and nutrients. Such opposing goals are achieved by a complex anatomical and functional structure the intestinal barrier consists of, the functional status of which is described by 'intestinal permeability'. Third, the regulation of intestinal permeability by diet and bacteria is depicted. In particular, potential barrier disruptors such as hypoperfusion of the gut, infections and toxins, but also selected over-dosed nutrients, drugs, and other lifestyle factors have to be considered. In the fourth part, the means to assess intestinal permeability are presented and critically discussed. The means vary enormously and probably assess different functional components of the barrier. The barrier assessments are further hindered by the natural variability of this functional entity depending on species and genes as well as on diet and other environmental factors. In the final part, we discuss selected diseases associated with increased intestinal permeability such as critically illness, inflammatory bowel diseases, celiac disease, food allergy, irritable bowel syndrome, and--more recently recognized--obesity and metabolic diseases. All these diseases are characterized by inflammation that might be triggered by the translocation of luminal components into the host. In summary, intestinal permeability, which is a feature of intestinal barrier function, is increasingly recognized as being of relevance for health and disease, and therefore, this topic warrants more attention.


Asunto(s)
Mucosa Intestinal/metabolismo , Animales , Biomarcadores/metabolismo , Enfermedad Crítica , Dieta , Hígado Graso/metabolismo , Histamina/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/inmunología , Intestinos/inmunología , Intestinos/microbiología , Transporte Iónico , Obesidad/metabolismo , Péptido Hidrolasas/metabolismo , Permeabilidad , Prebióticos , Probióticos/administración & dosificación , Receptor Cannabinoide CB1/metabolismo , Serotonina/metabolismo
12.
Curr Cardiol Rep ; 16(11): 540, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25303894

RESUMEN

These days, the gut microbiota is universally recognized as an active organ that can modulate the overall host metabolism by promoting multiple functions, from digestion to the systemic maintenance of overall host physiology. Dysbiosis, the alteration of the complex ecologic system of gut microbes, is associated with and causally responsible for multiple types of pathologies. Among the latters, metabolic diseases such as type 2 diabetes and obesity are each distinguishable by a unique gut microbiota profile. Interestingly, the specific microbiota typically found in the blood of diabetic patients also has been observed at the level of atherosclerotic plaque. Here, we report evidence from the literature, as well as a few controversial reports, regarding the putative role of gut microbiota dysbiosis-induced cardiovascular diseases, such as atherosclerosis, which are common comorbidities of metabolic dysfunction.


Asunto(s)
Aterosclerosis/etiología , Disbiosis/complicaciones , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Inflamación/inmunología , Inflamación/microbiología , Microbiota/inmunología , Periodontitis/complicaciones , Animales , Aterosclerosis/inmunología , Modelos Animales de Enfermedad , Disbiosis/inmunología , Disbiosis/microbiología , Medicina Basada en la Evidencia , Humanos , Inmunidad Innata , Ratones , Periodontitis/inmunología , Probióticos/uso terapéutico , Ratas
13.
Microorganisms ; 12(6)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38930480

RESUMEN

Upper urinary tract urolithiasis is an emerging disease in cats, with 98% of kidney stones composed of calcium oxalate. In humans, disturbances in the intestinal and urinary microbiota are suspected to contribute to the formation of calcium oxalate stones. We hypothesized that similar mechanisms may be at play in cats. This study examines the intestinal and urinary microbiota of nine cats with kidney stones compared to nine healthy cats before, during, and after treatment with the antibiotic cefovecin, a cephalosporin. Initially, cats with kidney stones displayed a less diverse intestinal microbiota. Antibiotic treatment reduced microbiota diversity in both groups. The absence of specific intestinal bacteria could lead to a loss of the functions these bacteria perform, such as oxalate degradation, which may contribute to the formation of calcium oxalate stones. This study confirms the presence of a distinct urobiome in cats with kidney stones, characterized by greater richness and diversity compared to healthy cats. These findings highlight the potential of microbiota modulation as a strategy to prevent renal lithiasis in cats.

14.
Diabetologia ; 56(11): 2524-37, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23963324

RESUMEN

AIMS/HYPOTHESIS: Circulating lipopolysaccharide-binding protein (LBP) is an acute-phase reactant known to be increased in obesity. We hypothesised that LBP is produced by adipose tissue (AT) in association with obesity. METHODS: LBP mRNA and LBP protein levels were analysed in AT from three cross-sectional (n = 210, n = 144 and n = 28) and three longitudinal (n = 8, n = 25, n = 20) human cohorts; in AT from genetically manipulated mice; in isolated adipocytes; and in human and murine cell lines. The effects of a high-fat diet and exposure to lipopolysaccharide (LPS) and peroxisome proliferator-activated receptor (PPAR)γ agonist were explored. Functional in vitro and ex vivo experiments were also performed. RESULTS: LBP synthesis and release was demonstrated to increase with adipocyte differentiation in human and mouse AT, isolated adipocytes and human and mouse cell lines (Simpson-Golabi-Behmel syndrome [SGBS], human multipotent adipose-derived stem [hMAD] and 3T3-L1 cells). AT LBP expression was robustly associated with inflammatory markers and increased with metabolic deterioration and insulin resistance in two independent cross-sectional human cohorts. AT LBP also increased longitudinally with weight gain and excessive fat accretion in both humans and mice, and decreased with weight loss (in two other independent cohorts), in humans with acquired lipodystrophy, and after ex vivo exposure to PPARγ agonist. Inflammatory agents such as LPS and TNF-α led to increased AT LBP expression in vivo in mice and in vitro, while this effect was prevented in Cd14-knockout mice. Functionally, LBP knockdown using short hairpin (sh)RNA or anti-LBP antibody led to increases in markers of adipogenesis and decreased adipocyte inflammation in human adipocytes. CONCLUSIONS/INTERPRETATION: Collectively, these findings suggest that LBP might have an essential role in inflammation- and obesity-associated AT dysfunction.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Adipocitos/metabolismo , Tejido Adiposo/patología , Proteínas Portadoras/metabolismo , Inflamación/metabolismo , Glicoproteínas de Membrana/metabolismo , Obesidad/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Adulto , Animales , Humanos , Técnicas In Vitro , Resistencia a la Insulina/fisiología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Rosiglitazona , Tiazolidinedionas/farmacología , Factor de Necrosis Tumoral alfa/farmacología
15.
Diabetes Obes Metab ; 15 Suppl 3: 61-70, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24003922

RESUMEN

Over the last decade, the research community has revealed the role of a new organ: the intestinal microbiota. It is considered as a symbiont that is part of our organism since, at birth, it educates the immune system and contributes to the development of the intestinal vasculature and most probably the nervous system. With the advent of new generation sequencing techniques, a catalogue of genes that belong to this microbiome has been established that lists more than 5 million non-redundant genes called the metagenome. Using germ free mice colonized with the microbiota from different origins, it has been formally demonstrated that the intestinal microbiota causes the onset of metabolic diseases. Further to the role of point mutations in our genome, the microbiota can explain the on-going worldwide pandemic of obesity and diabetes, its dissemination and family inheritance, as well as the diversity of the associated metabolic phenotypes. More recently, the discovery of bacterial DNA within host tissues, such as the liver, the adipose tissue and the blood, which establishes a tissue microbiota, introduces new opportunities to identify targets and predictive biomarkers based on the host to microbiota interaction, as well as to define new strategies for pharmacological, immunomodulatory vaccines and nutritional applications.


Asunto(s)
Metabolismo/fisiología , Metagenoma/fisiología , Microbiota/fisiología , Animales , Comunicación Celular/fisiología , Especificidad del Huésped/inmunología , Humanos , Intestinos/inmunología , Intestinos/microbiología , Enfermedades Metabólicas/microbiología , Ratones
16.
Med Sci (Paris) ; 29(8-9): 800-6, 2013.
Artículo en Francés | MEDLINE | ID: mdl-24005637

RESUMEN

A new organ has emerged over the course of the last century: the intestinal microbiota. It is characterized by numerous functions provided by several billions of bacteria inhabiting and living in harmony in the lumen and in the mucosal layer of the intestinal epithelium. More than 4 million genes composed by more than 1 500 species interact with each other, with the host and the environment to set up a mutualistic ecological group. A nutritional stress will modify the terms of the symbiosis between the host and the microbiota for the control of energy homeostasis. It is now thought that the pandemic of diabetes and obesity, not being due to the sole variations of our genome, would be due to changes in our metagenome: our intestinal bacteria. This organ which genomic varies on an everyday basis is inherited from our mother and the closed environment at birth. The corresponding diversity, the rapid evolution of gene expression, its influence on metabolism, as well as the very recent discovery of the existence of an tissue microbiota within the host, open new therapeutic pharmacological and nutritional opportunities as well as the identification of very accurate biomarkers constituting a personalized metagenomic identity card. Hence, individualized medicine foresees its origin within the metagenome.


Asunto(s)
Intestinos/microbiología , Enfermedades Metabólicas/microbiología , Enfermedades Metabólicas/terapia , Metagenoma/genética , Metagenoma/fisiología , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/microbiología , Diabetes Mellitus Tipo 2/terapia , Dieta , Metabolismo Energético/fisiología , Homeostasis , Humanos , Enfermedades Metabólicas/genética , Fenómenos Fisiológicos de la Nutrición , Obesidad/genética , Obesidad/microbiología , Obesidad/terapia
17.
Gut ; 61(4): 543-53, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22110050

RESUMEN

OBJECTIVE: The gut microbiota, which is considered a causal factor in metabolic diseases as shown best in animals, is under the dual influence of the host genome and nutritional environment. This study investigated whether the gut microbiota per se, aside from changes in genetic background and diet, could sign different metabolic phenotypes in mice. METHODS: The unique animal model of metabolic adaptation was used, whereby C57Bl/6 male mice fed a high-fat carbohydrate-free diet (HFD) became either diabetic (HFD diabetic, HFD-D) or resisted diabetes (HFD diabetes-resistant, HFD-DR). Pyrosequencing of the gut microbiota was carried out to profile the gut microbial community of different metabolic phenotypes. Inflammation, gut permeability, features of white adipose tissue, liver and skeletal muscle were studied. Furthermore, to modify the gut microbiota directly, an additional group of mice was given a gluco-oligosaccharide (GOS)-supplemented HFD (HFD+GOS). RESULTS: Despite the mice having the same genetic background and nutritional status, a gut microbial profile specific to each metabolic phenotype was identified. The HFD-D gut microbial profile was associated with increased gut permeability linked to increased endotoxaemia and to a dramatic increase in cell number in the stroma vascular fraction from visceral white adipose tissue. Most of the physiological characteristics of the HFD-fed mice were modulated when gut microbiota was intentionally modified by GOS dietary fibres. CONCLUSIONS: The gut microbiota is a signature of the metabolic phenotypes independent of differences in host genetic background and diet.


Asunto(s)
Adaptación Fisiológica/fisiología , Dieta Alta en Grasa , Intestinos/microbiología , Metagenoma/fisiología , Animales , Ciego/microbiología , Citocinas/sangre , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/fisiopatología , Ácidos Grasos no Esterificados/sangre , Prueba de Tolerancia a la Glucosa , Absorción Intestinal/fisiología , Lipopolisacáridos/sangre , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Permeabilidad , Fenotipo
18.
Commun Biol ; 6(1): 1168, 2023 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-37968381

RESUMEN

Opioid-dependent immune-mediated analgesic effects have been broadly reported upon inflammation. In preclinical mouse models of intestinal inflammatory diseases, the local release of enkephalins (endogenous opioids) by colitogenic T lymphocytes alleviate inflammation-induced pain by down-modulating gut-innervating nociceptor activation in periphery. In this study, we wondered whether this immune cell-derived enkephalin-mediated regulation of the nociceptor activity also operates under steady state conditions. Here, we show that chimeric mice engrafted with enkephalin-deficient bone marrow cells exhibit not only visceral hypersensitivity but also an increase in both epithelial paracellular and transcellular permeability, an alteration of the microbial topography resulting in increased bacteria-epithelium interactions and a higher frequency of IgA-producing plasma cells in Peyer's patches. All these alterations of the intestinal homeostasis are associated with an anxiety-like behavior despite the absence of an overt inflammation as observed in patients with irritable bowel syndrome. Thus, our results show that immune cell-derived enkephalins play a pivotal role in maintaining gut homeostasis and normal behavior in mice. Because a defect in the mucosal opioid system remarkably mimics some major clinical symptoms of the irritable bowel syndrome, its identification might help to stratify subgroups of patients.


Asunto(s)
Síndrome del Colon Irritable , Humanos , Animales , Ratones , Analgésicos Opioides , Encefalinas/genética , Inflamación , Dolor
19.
J Clin Gastroenterol ; 46 Suppl: S27-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22955352

RESUMEN

Each individual can be distinguished by the heterogeneity of the trillions of microbes inhabiting his gastrointestinal tract. This concept, together with the role that gut microbiota is considered to play in the induction of metabolic diseases, paves the way for the development of personalized medicine. By exploiting our unique animal model of metabolic adaptation to a high-fat diet, we have recently shown that differential gut microbiota lead to different metabolic phenotypes--metabotypes. Moreover, we have also reported that a given metabotype can be distinguished by different profiles of gut microbes, symptomatic of the complexity of the regulation of host physiology by gut microbiota. Furthermore, in an effort to find bacterial predictors of type 2 diabetes (T2D), we discovered that in a healthy population, subjects who subsequently developed T2D had increased blood levels of bacterial 16S rDNA well before. In addition, tissue (blood) microbiota, mainly characterized by Proteobacteria (up to 90%), has been discovered both in healthy individuals and in diabetic patients. Altogether, our results confirm the presence of gut microbes and propose tissue microbiota as new targets for the innovative treatment of T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/microbiología , Diabetes Mellitus Tipo 2/fisiopatología , Tracto Gastrointestinal/microbiología , Metagenoma/fisiología , Adaptación Fisiológica , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa , Humanos , Inflamación , Ratones
20.
Sci Rep ; 12(1): 1415, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35082330

RESUMEN

Intestinal gluconeogenesis (IGN), gastric bypass (GBP) and gut microbiota positively regulate glucose homeostasis and diet-induced dysmetabolism. GBP modulates gut microbiota, whether IGN could shape it has not been investigated. We studied gut microbiota and microbiome in wild type and IGN-deficient mice, undergoing GBP or not, and fed on either a normal chow (NC) or a high-fat/high-sucrose (HFHS) diet. We also studied fecal and urine metabolome in NC-fed mice. IGN and GBP had a different effect on the gut microbiota of mice fed with NC and HFHS diet. IGN inactivation increased abundance of Deltaproteobacteria on NC and of Proteobacteria such as Helicobacter on HFHS diet. GBP increased abundance of Firmicutes and Proteobacteria on NC-fed WT mice and of Firmicutes, Bacteroidetes and Proteobacteria on HFHS-fed WT mice. The combined effect of IGN inactivation and GBP increased abundance of Actinobacteria on NC and the abundance of Enterococcaceae and Enterobacteriaceae on HFHS diet. A reduction was observed in the amounf of short-chain fatty acids in fecal (by GBP) and in both fecal and urine (by IGN inactivation) metabolome. IGN and GBP, separately or combined, shape gut microbiota and microbiome on NC- and HFHS-fed mice, and modify fecal and urine metabolome.


Asunto(s)
Derivación Gástrica/métodos , Microbioma Gastrointestinal/fisiología , Gluconeogénesis/fisiología , Intestinos/metabolismo , Metaboloma , Estómago/metabolismo , Actinobacteria/clasificación , Actinobacteria/genética , Actinobacteria/aislamiento & purificación , Animales , ADN Bacteriano/genética , Enterobacteriaceae/clasificación , Enterobacteriaceae/genética , Enterobacteriaceae/aislamiento & purificación , Enterococcaceae/clasificación , Enterococcaceae/genética , Enterococcaceae/aislamiento & purificación , Ácidos Grasos Volátiles/metabolismo , Firmicutes/clasificación , Firmicutes/genética , Firmicutes/aislamiento & purificación , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Filogenia , Proteobacteria/clasificación , Proteobacteria/genética , Proteobacteria/aislamiento & purificación , Estómago/microbiología , Estómago/cirugía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA