Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
BMJ Open ; 13(3): e067016, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-37001916

RESUMEN

INTRODUCTION: The gut microbiota develops from birth and matures significantly during the first 24 months of life, playing a major role in infant health and development. The composition of the gut microbiota is influenced by several factors including mode of delivery, gestational age, feed type and treatment with antibiotics. Alterations in the pattern of gut microbiota development and composition can be associated with illness and compromised health outcomes.Infants diagnosed with 'congenital heart disease' (CHD) often require surgery involving cardiopulmonary bypass (CPB) early in life. The impact of this type of surgery on the integrity of the gut microbiome is poorly understood. In addition, these infants are at significant risk of developing the potentially devastating intestinal condition necrotising enterocolitis. METHODS AND ANALYSIS: This study will employ a prospective cohort study methodology to investigate the gut microbiota and urine metabolome of infants with CHD undergoing surgery involving CPB. Stool and urine samples, demographic and clinical data will be collected from eligible infants based at the National Centre for Paediatric Cardiac Surgery in Ireland. Shotgun metagenome sequencing will be performed on stool samples and urine metabolomic analysis will identify metabolic biomarkers. The impact of the underlying diagnosis, surgery involving CPB, and the influence of environmental factors will be explored. Data from healthy age-matched infants from the INFANTMET study will serve as a control for this study. ETHICS AND DISSEMINATION: This study has received full ethical approval from the Clinical Research Ethics Committee of Children's Health Ireland, GEN/826/20.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Microbioma Gastrointestinal , Cardiopatías Congénitas , Recién Nacido , Lactante , Humanos , Niño , Puente Cardiopulmonar , Estudios Prospectivos , Cardiopatías Congénitas/cirugía
2.
Int J Mol Sci ; 23(10)2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-35628140

RESUMEN

Gastrointestinal toxicity (GIT) is a debilitating side effect of Irinotecan (CPT-11) and limits its clinical utility. Gut dysbiosis has been shown to mediate this side effect of CPT-11 by increasing gut bacterial ß-glucuronidase (GUSB) activity and impairing the intestinal mucosal barrier (IMB). We have recently shown the opposing effects of omega-6 (n-6) and omega-3 (n-3) polyunsaturated fatty acids (PUFA) on the gut microbiome. We hypothesized that elevated levels of tissue n-3 PUFA with a decreased n-6/n-3 PUFA ratio would reduce CPT-11-induced GIT and associated changes in the gut microbiome. Using a unique transgenic mouse (FAT-1) model combined with dietary supplementation experiments, we demonstrate that an elevated tissue n-3 PUFA status with a decreased n-6/n-3 PUFA ratio significantly reduces CPT-11-induced weight loss, bloody diarrhea, gut pathological changes, and mortality. Gut microbiome analysis by 16S rRNA gene sequencing and QIIME2 revealed that improvements in GIT were associated with the reduction in the CPT-11-induced increase in both GUSB-producing bacteria (e.g., Enterobacteriaceae) and GUSB enzyme activity, decrease in IMB-maintaining bacteria (e.g., Bifidobacterium), IMB dysfunction and systemic endotoxemia. These results uncover a host-microbiome interaction approach to the management of drug-induced gut toxicity. The prevention of CPT-11-induced gut microbiome changes by decreasing the tissue n-6/n-3 PUFA ratio could be a novel strategy to prevent chemotherapy-induced GIT.


Asunto(s)
Antineoplásicos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Ácidos Grasos Omega-3 , Enfermedades Gastrointestinales , Microbioma Gastrointestinal , Animales , Antineoplásicos/farmacología , Bacterias/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/tratamiento farmacológico , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-3/uso terapéutico , Ácidos Grasos Omega-6/farmacología , Enfermedades Gastrointestinales/tratamiento farmacológico , Irinotecán/farmacología , Ratones , ARN Ribosómico 16S/genética
3.
Neurobiol Pain ; 10: 100072, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34485761

RESUMEN

Gut microbiota play a role in certain pain states. Hence, these microbiota also influence somatic pain. We aimed to determine if there was an association between gut microbiota (composition and diversity) and postoperative pain. Patients (n = 20) undergoing surgical fixation of distal radius fracture under axillary brachial plexus block were studied. Gut microbiota diversity and abundance were analysed for association with: (i) a verbal pain rating scale of < 4/10 throughout the first 24 h after surgery (ii) a level of pain deemed "acceptable" by the patient during the first 24 h following surgery (iii) a maximum self-reported pain score during the first 24 h postoperatively and (iv) analgesic consumption during the first postoperative week. Analgesic consumption was inversely correlated with the Shannon index of alpha diversity. There were also significant differences, at the genus level (including Lachnospira), with respect to pain being "not acceptable" at 24 h postoperatively. Porphyromonas was more abundant in the group reporting an acceptable pain level at 24 h. An inverse correlation was noted between abundance of Collinsella and maximum self-reported pain score with movement. We have demonstrated for the first time that postoperative pain is associated with gut microbiota composition and diversity. Further work on the relationship between the gut microbiome and somatic pain may offer new therapeutic targets.

4.
Front Nutr ; 8: 667188, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34557508

RESUMEN

Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in preterm infants. The exact mechanism by which NEC develops is poorly understood however there is growing evidence to suggest that perturbations in the early-life gut microbiota composition increase the risk for NEC. Modulation of the gut microbiota with probiotics, prebiotics, or in combination (synbiotics) is an area which has attracted intense interest in recent years. In this narrative review, we present an overview of the role of the gut microbiota in the pathogenesis of NEC. We also examine the evidence currently available from randomized controlled trials, observational studies, systematic reviews, and meta-analysis examining the role of probiotics, prebiotics, and synbiotics in reducing the risk of or preventing NEC. Current clinical practice guidelines with recommendations on the routine administration of probiotics to preterm infants for NEC are also explored.

5.
Metabolites ; 11(8)2021 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-34436434

RESUMEN

Cardiovascular disease (CVD) has been classified as one of the leading causes of morbidity and mortality worldwide. CVD risk factors include smoking, hypertension, dyslipidaemia, obesity, inflammation and diabetes. The gut microbiota can influence human health through multiple interactions and community changes are associated with the development and progression of numerous disease states, including CVD. The gut microbiota are involved in the production of several metabolites, such as short-chain fatty acids (SCFAs), bile acids and trimethylamine-N-oxide (TMAO). These products of microbial metabolism are important modulatory factors and have been associated with an increased risk of CVD. Due to its association with CVD development, the gut microbiota has emerged as a target for therapeutic approaches. In this review, we summarise the current knowledge on the role of the gut microbiome in CVD development, and associated microbial communities, functions, and metabolic profiles. We also discuss CVD therapeutic interventions that target the gut microbiota such as probiotics and faecal microbiota transplantation.

6.
EBioMedicine ; 63: 103176, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33349590

RESUMEN

BACKGROUND: The human gut microbiota has emerged as a key factor in the development of obesity. Certain probiotic strains have shown anti-obesity effects. The objective of this study was to investigate whether Bifidobacterium longum APC1472 has anti-obesity effects in high-fat diet (HFD)-induced obese mice and whether B. longum APC1472 supplementation reduces body-mass index (BMI) in healthy overweight/obese individuals as the primary outcome. B. longum APC1472 effects on waist-to-hip ratio (W/H ratio) and on obesity-associated plasma biomarkers were analysed as secondary outcomes. METHODS: B. longum APC1472 was administered to HFD-fed C57BL/6 mice in drinking water for 16 weeks. In the human intervention trial, participants received B. longum APC1472 or placebo supplementation for 12 weeks, during which primary and secondary outcomes were measured at the beginning and end of the intervention. FINDINGS: B. longum APC1472 supplementation was associated with decreased bodyweight, fat depots accumulation and increased glucose tolerance in HFD-fed mice. While, in healthy overweight/obese adults, the supplementation of B. longum APC1472 strain did not change primary outcomes of BMI (0.03, 95% CI [-0.4, 0.3]) or W/H ratio (0.003, 95% CI [-0.01, 0.01]), a positive effect on the secondary outcome of fasting blood glucose levels was found (-0.299, 95% CI [-0.44, -0.09]). INTERPRETATION: This study shows a positive translational effect of B. longum APC1472 on fasting blood glucose from a preclinical mouse model of obesity to a human intervention study in otherwise healthy overweight and obese individuals. This highlights the promising potential of B. longum APC1472 to be developed as a valuable supplement in reducing specific markers of obesity. FUNDING: This research was funded in part by Science Foundation Ireland in the form of a Research Centre grant (SFI/12/RC/2273) to APC Microbiome Ireland and by a research grant from Cremo S.A.


Asunto(s)
Bifidobacterium longum/fisiología , Resistencia a la Enfermedad , Interacciones Microbiota-Huesped , Obesidad/metabolismo , Adiposidad , Corticoesteroides/sangre , Animales , Biomarcadores , Peso Corporal , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Metabolismo Energético , Glucosa/metabolismo , Leptina/sangre , Masculino , Ratones , Neuropéptidos/genética , Neuropéptidos/metabolismo , Obesidad/etiología , Probióticos , Roedores , Investigación Biomédica Traslacional
7.
Int J Obes (Lond) ; 45(3): 588-598, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33223517

RESUMEN

BACKGROUND: Early consumption of obesogenic diets, rich in saturated fat and added sugar, is associated with a plethora of biological dysfunctions, at both peripheral and brain levels. Obesity is also linked to decreased vitamin A bioavailability, an essential molecule for brain plasticity and memory function. METHODS: Here we investigated in mice whether dietary vitamin A supplementation (VAS) could prevent some of the metabolic, microbiota, neuronal and cognitive alterations induced by obesogenic, high-fat and high-sugar diet (HFSD) exposure from weaning to adulthood, i.e. covering periadolescent period. RESULTS: As expected, VAS was effective in enhancing peripheral vitamin A levels as well as hippocampal retinoic acid levels, the active metabolite of vitamin A, regardless of the diet. VAS attenuated HFSD-induced excessive weight gain, without affecting metabolic changes, and prevented alterations of gut microbiota α-diversity. In HFSD-fed mice, VAS prevented recognition memory deficits but had no effect on aversive memory enhancement. Interestingly, VAS alleviated both HFSD-induced higher neuronal activation and lower glucocorticoid receptor phosphorylation in the hippocampus after training. CONCLUSION: Dietary VAS was protective against the deleterious effects of early obesogenic diet consumption on hippocampal function, possibly through modulation of the gut-brain axis.


Asunto(s)
Cognición/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Microbioma Gastrointestinal/efectos de los fármacos , Vitamina A , Animales , Eje Cerebro-Intestino/efectos de los fármacos , Hipocampo/química , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Vitamina A/administración & dosificación , Vitamina A/farmacología
8.
Neuronal Signal ; 4(4): NS20200007, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33343931

RESUMEN

Visceral hypersensitivity is a hallmark of many functional and stress-related gastrointestinal disorders, and there is growing evidence that the gut microbiota may play a role in its pathophysiology. It has previously been shown that early life stress-induced visceral sensitivity is reduced by various probiotic strains of bacteria (including Lactobacillus rhamnosus GG (LGG)) alone or in combination with prebiotic fibres in rat models. However, the exact mechanisms underpinning such effects remain unresolved. Here, we investigated if soluble mediators derived from LGG can mimic the bacteria's effects on visceral hypersensitivity and the microbiota-gut-brain axis. Rats were exposed to maternal separation (MS) from postnatal days 2-12. From weaning onwards both non-separated (NS) and MS offspring were provided drinking water with or without supplementation of standardized preparations of the LGG soluble mediators (LSM). Our results show that MS led to increased visceral sensitivity and exaggerated corticosterone plasma levels following restraint stress in adulthood, and both of these effects were ameliorated through LSM supplementation. Differential regulation of various genes in the spinal cord of MS versus NS rats was observed, 41 of which were reversed by LSM supplementation. At the microbiota composition level MS led to changes in beta diversity and abundance of specific bacteria including parabacteroides, which were ameliorated by LSM. These findings support probiotic soluble mediators as potential interventions in the reduction of symptoms of visceral hypersensitivity.

10.
Eur J Neurosci ; 51(4): 1042-1058, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31339598

RESUMEN

Nutritional interventions targeting the microbiota-gut-brain axis are proposed to modulate stress-induced dysfunction of physiological processes and brain development. Maternal separation (MS) in rats induces long-term alterations to behaviour, pain responses, gut microbiome and brain neurochemistry. In this study, the effects of dietary interventions (milk fat globule membrane [MFGM] and a polydextrose/galacto-oligosaccharide prebiotic blend) were evaluated. Diets were provided from postnatal day 21 to both non-separated and MS offspring. Spatial memory, visceral sensitivity and stress reactivity were assessed in adulthood. Gene transcripts associated with cognition and stress and the caecal microbiota composition were analysed. MS-induced visceral hypersensitivity was ameliorated by MFGM and to greater extent with the combination of MFGM and prebiotic blend. Furthermore, spatial learning and memory were improved by prebiotics and MFGM alone and with the combination. The prebiotic blend and the combination of the prebiotics and MFGM appeared to facilitate return to baseline with regard to HPA axis response to the restraint stress, which can be beneficial in times where coping mechanisms to stressful events are required. Interestingly, the combination of MFGM and prebiotic reduced the long-term impact of MS on a marker of myelination in the prefrontal cortex. MS affected the microbiota at family level only, while MFGM, the prebiotic blend and the combination influenced abundance at family and genus level as well as influencing beta-diversity levels. In conclusion, intervention with MFGM and prebiotic blend significantly impacted the composition of the microbiota as well as ameliorating some of the long-term effects of early-life stress.


Asunto(s)
Microbioma Gastrointestinal , Privación Materna , Microbiota , Animales , Encéfalo , Glucolípidos , Glicoproteínas , Sistema Hipotálamo-Hipofisario , Gotas Lipídicas , Sistema Hipófiso-Suprarrenal , Prebióticos , Ratas , Estrés Fisiológico
11.
Microbiol Resour Announc ; 8(33)2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-31416857

RESUMEN

We report the draft genome sequence of Bacillus thuringiensis DPC6431, a producer of the anticlostridial bacteriocin thuricin CD and isolated from a human fecal sample. The assembly comprises 96 contigs for a total of 5,581,839 bp, with 32.5% G+C content.

12.
Arch Dis Child Fetal Neonatal Ed ; 104(2): F159-F164, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29925539

RESUMEN

OBJECTIVE: The objective of this study was to investigate the appropriate dosing interval of a probiotic (Infloran) given daily, biweekly and weekly in preterm infants <32 weeks' gestation. METHODS: There were 8 infants in the daily group, 8 infants in the biweekly group and 10 infants in the weekly group, all born between 25 and 32 weeks' gestation. The control group consisted of 12 preterm infants who did not receive the probiotic. Infloran (250 mg/capsule), containing Bifidobacterium bifidum (1×109 colony-forming unit (CFU)) and Lactobacillus acidophilus (1×109 CFU), was administered in 2.5 mL of breast milk per kilogram weight of the infant (2×109 CFU of bacteria in total), until 34 weeks postmenstrual age (PMA). Stool samples were collected at 31, 34, 41 and 44 weeks PMA and frozen at -20°C. RESULTS: After administration of the probiotic at 31 weeks PMA, Bifidobacterium were significantly higher in the daily group (45%) in comparison with the biweekly (17%) and weekly (9%) groups. At 34 weeks PMA, Bifidobacterium were significantly higher again in the daily (60%) group in comparison with the biweekly (21%), weekly (23%) and control (15%) groups. At 41 weeks PMA a decrease in the relative abundances of Streptococcaceae and Enterococcaceae was found in all three probiotic groups, and by 44 weeks PMA significantly higher levels of Lactobacillus were found in the biweekly group (16.5%) in comparison with the weekly group (2.1%). CONCLUSION: Our results indicate that a daily dose of Infloran is a suitable dosage for preterm infants in the neonatal intensive care unit, with significantly higher levels of Bifidobacterium found in the daily probiotic group up to 44 weeks PMA.


Asunto(s)
Recien Nacido Prematuro , Probióticos/administración & dosificación , Bifidobacterium/aislamiento & purificación , Esquema de Medicación , Enterocolitis Necrotizante/prevención & control , Heces/microbiología , Humanos , Lactobacillus acidophilus/aislamiento & purificación
13.
J Educ Perioper Med ; 20(3): E626, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30510974

RESUMEN

BACKGROUND: The use of epidural analgesia for laboring women is generally unavailable at public hospitals in Guyana despite favorable utilization rates in private institutions. In 2014, a healthcare team completed a targeted mission aimed at neuraxial analgesia training of providers at the preeminent public hospital in Georgetown, Guyana. This study evaluates the impact of the training, including provider attitudes, use, and barriers. METHODS: A prospective, mixed methods study of all obstetric, nursing, and anesthesiology providers at Georgetown Public Hospital Corporation was completed. Quantitative assessment of the posttraining use of epidural analgesia at 2 and 6 months was documented. Provider surveys were distributed anonymously at 2 months posttraining. Targeted interviews were completed from a random sampling of providers at 6 months; qualitative analysis of interviews formulated the basis for reporting limitations and barriers. RESULTS: Providers surveyed included 7 anesthesia providers and 24 obstetrics providers. Respondents believed Guyanese women should be offered epidural analgesia (93%), epidurals could be performed safely (87%), and Guyana has the resources necessary for routine use (81%). In assessing epidural knowledge, anesthesia providers achieved 60% correct response rate compared to 84% among obstetrics providers. Nurse anesthetists placed 16 epidurals following training. However, placement ceased after 2 months. The largest barriers to placement were unavailable anesthesia staff (63%), lack of supplies (16%), and insufficient nursing staff to monitor patients with epidurals (11%). CONCLUSIONS: A 1-week mission achieved widespread Guyanese provider acceptance despite a lack of previous experience. However, barriers proved insurmountable to achieving a sustainable, independently functioning epidural analgesia program.

14.
Microbiome ; 6(1): 205, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30424806

RESUMEN

BACKGROUND: Understanding the mechanism of the sexual dimorphism in susceptibility to obesity and metabolic syndrome (MS) is important for the development of effective interventions for MS. RESULTS: Here we show that gut microbiome mediates the preventive effect of estrogen (17ß-estradiol) on metabolic endotoxemia (ME) and low-grade chronic inflammation (LGCI), the underlying causes of MS and chronic diseases. The characteristic profiles of gut microbiome observed in female and 17ß-estradiol-treated male and ovariectomized mice, such as decreased Proteobacteria and lipopolysaccharide biosynthesis, were associated with a lower susceptibility to ME, LGCI, and MS in these animals. Interestingly, fecal microbiota-transplant from male mice transferred the MS phenotype to female mice, while antibiotic treatment eliminated the sexual dimorphism in MS, suggesting a causative role of the gut microbiome in this condition. Moreover, estrogenic compounds such as isoflavones exerted microbiome-modulating effects similar to those of 17ß-estradiol and reversed symptoms of MS in the male mice. Finally, both expression and activity of intestinal alkaline phosphatase (IAP), a gut microbiota-modifying non-classical anti-microbial peptide, were upregulated by 17ß-estradiol and isoflavones, whereas inhibition of IAP induced ME and LGCI in female mice, indicating a critical role of IAP in mediating the effects of estrogen on these parameters. CONCLUSIONS: In summary, we have identified a previously uncharacterized microbiome-based mechanism that sheds light upon sexual dimorphism in the incidence of MS and that suggests novel therapeutic targets and strategies for the management of obesity and MS in males and postmenopausal women.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Microbioma Gastrointestinal/fisiología , Síndrome Metabólico/prevención & control , Proteobacteria/metabolismo , Caracteres Sexuales , Fosfatasa Alcalina/biosíntesis , Animales , Carga Bacteriana , Trasplante de Microbiota Fecal , Femenino , Isoflavonas/farmacología , Lipopolisacáridos/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL
15.
Mar Drugs ; 16(6)2018 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-29925774

RESUMEN

Accumulating evidence demonstrates that dietary supplementation with functional food ingredients play a role in systemic and brain health as well as in healthy ageing. Conversely, deficiencies in calcium and magnesium as a result of the increasing prevalence of a high fat/high sugar "Western diet" have been associated with health problems such as obesity, inflammatory bowel diseases, and cardiovascular diseases, as well as metabolic, immune, and psychiatric disorders. It is now recognized that modulating the diversity of gut microbiota, the population of intestinal bacteria, through dietary intervention can significantly impact upon gut health as well as systemic and brain health. In the current study, we show that supplementation with a seaweed and seawater-derived functional food ingredient rich in bioactive calcium and magnesium (0.1% supplementation) as well as 70 other trace elements, significantly enhanced the gut microbial diversity in adult male rats. Given the significant impact of gut microbiota on health, these results position this marine multi-mineral blend (MMB) as a promising digestive-health promoting functional food ingredient.


Asunto(s)
Suplementos Dietéticos , Alimentos Funcionales , Microbioma Gastrointestinal/efectos de los fármacos , Minerales/farmacología , Algas Marinas/química , Animales , Conducta Animal/efectos de los fármacos , ADN Bacteriano/aislamiento & purificación , Microbioma Gastrointestinal/genética , Masculino , Minerales/química , Modelos Animales , ARN Ribosómico 16S/genética , Ratas , Ratas Sprague-Dawley
16.
Eur J Pediatr ; 177(6): 867-870, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29610991

RESUMEN

The aim of this study was to determine bacteriological stability of a probiotic mixture dispersed in various diluents. The commercially available probiotic (Infloran®), containing Bifidobacterium bifidum (109 CFU/250 mg tablet) and Lactobacillus acidophilus (109 CFU/250 mg tablet), was dispersed within expressed breast milk, sterile water, and infant formula and examined at temperatures of 4 and 21 °C. When stored at 4 °C, significant decreases (P < 0.05) in the level of L. acidophilus and B. bifidum were observed in expressed breast milk and sterile water after a 6-h period. However, when stored in infant formula, both strains remained stable over a 12-h period. When stored at 21 °C, a significant decrease (P < 0.05) was observed in the level of L. acidophilus in sterile water, expressed breast milk and infant formula throughout a 12-h period. However, no significant decrease was observed overtime in B. bifidum in all three diluents at this temperature. CONCLUSION: Our findings suggest that, when stored at 4 °C, this probiotic product can remain at a stable condition for 6 h in sterile water and infant formula; however, the viability of the probiotic decreases significantly after this period of time. Administration of this probiotic in sterile water can be an acceptable alternative to dispersion and administration in expressed breast milk. What is Known: • Administration of probiotics containing lactobacilli and bifidobacteria has become more widespread in neonatology, mainly as prophylaxis for the prevention of necrotising entercolitis in preterm infants. • Probiotic reconstitution, from its powder base, is not standardized and various diluents, including sterile water, breast milk, and infant formula, have been used. What is New: • When stored at 4 °C, a probiotic containing lactobacilli and bifidobacteria remains at a stable microbological condition for up to 6 h in sterile water. • Administration of this probiotic dispersed in sterile water, followed by an EBM feed, can be an acceptable alternative to dispersion and administration in EBM.


Asunto(s)
Bifidobacterium bifidum/fisiología , Fórmulas Infantiles/microbiología , Lactobacillus acidophilus/fisiología , Viabilidad Microbiana , Leche Humana/microbiología , Probióticos , Microbiología del Agua , Almacenamiento de Alimentos/métodos , Humanos , Lactante , Recién Nacido , Temperatura
17.
Br J Nutr ; 118(11): 959-970, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29173237

RESUMEN

n-3 PUFA are lipids that play crucial roles in immune-regulation, cardio-protection and neurodevelopment. However, little is known about the role that these essential dietary fats play in modulating caecal microbiota composition and the subsequent production of functional metabolites. To investigate this, female C57BL/6 mice were assigned to one of three diets (control (CON), n-3 supplemented (n3+) or n-3 deficient (n3-)) during gestation, following which their male offspring were continued on the same diets for 12 weeks. Caecal content of mothers and offspring were collected for 16S sequencing and metabolic phenotyping. n3- male offspring displayed significantly less % fat mass than n3+ and CON. n-3 Status also induced a number of changes to gut microbiota composition such that n3- offspring had greater abundance of Tenericutes, Anaeroplasma and Coriobacteriaceae. Metabolomics analysis revealed an increase in caecal metabolites involved in energy metabolism in n3+ including α-ketoglutaric acid, malic acid and fumaric acid. n3- animals displayed significantly reduced acetate, butyrate and total caecal SCFA production. These results demonstrate that dietary n-3 PUFA regulate gut microbiota homoeostasis whereby n-3 deficiency may induce a state of disturbance. Further studies are warranted to examine whether these microbial and metabolic disturbances are causally related to changes in metabolic health outcomes.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Ciego/microbiología , Ácidos Grasos Omega-3/deficiencia , Microbioma Gastrointestinal , Animales , Composición Corporal , ADN Bacteriano/aislamiento & purificación , Dieta , Suplementos Dietéticos , Ácidos Grasos/metabolismo , Ácidos Grasos Omega-3/sangre , Femenino , Fumaratos/metabolismo , Ácidos Cetoglutáricos/metabolismo , Malatos/metabolismo , Masculino , Metaboloma , Metabolómica , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/aislamiento & purificación , Análisis de Secuencia de ADN
18.
Sci Rep ; 7(1): 13523, 2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29051554

RESUMEN

ABSTACT: The gut hormone glucagon-like peptide (GLP)-1 and its analogues represent a new generation of anti-diabetic drugs, which have also demonstrated propensity to modulate host lipid metabolism. Despite this, drugs of this nature are currently limited to intramuscular administration routes due to intestinal degradation. The aim of this study was to design a recombinant microbial delivery vector for a GLP-1 analogue and assess the efficacy of the therapeutic in improving host glucose, lipid and cholesterol metabolism in diet induced obese rodents. Diet-induced obese animals received either Lactobacillus paracasei NFBC 338 transformed to express a long-acting analogue of GLP-1 or the isogenic control microbe which solely harbored the pNZ44 plasmid. Short-term GLP-1 microbe intervention in rats reduced serum low-density lipoprotein cholesterol, triglycerides and triglyceride-rich lipoprotein cholesterol substantially. Conversely, extended GLP-1 microbe intervention improved glucose-dependent insulin secretion, glucose metabolism and cholesterol metabolism, compared to the high-fat control group. Interestingly, the microbe significantly attenuated the adiposity associated with the model and altered the serum lipidome, independently of GLP-1 secretion. These data indicate that recombinant incretin-secreting microbes may offer a novel and safe means of managing cholesterol metabolism and diet induced dyslipidaemia, as well as insulin sensitivity in metabolic dysfunction.


Asunto(s)
Dieta Alta en Grasa , Péptido 1 Similar al Glucagón/genética , Lactobacillus/metabolismo , Obesidad/terapia , Animales , LDL-Colesterol/sangre , Modelos Animales de Enfermedad , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/microbiología , Lactobacillus/genética , Metaboloma , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Plásmidos/genética , Plásmidos/metabolismo , Ratas , Ratas Long-Evans , Triglicéridos/sangre
19.
EBioMedicine ; 24: 166-178, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28965876

RESUMEN

Autism spectrum disorder (ASD) is one of the most prevalent neurodevelopmental conditions worldwide. There is growing awareness that ASD is highly comorbid with gastrointestinal distress and altered intestinal microbiome, and that host-microbiome interactions may contribute to the disease symptoms. However, the paucity of knowledge on gut-brain axis signaling in autism constitutes an obstacle to the development of precision microbiota-based therapeutics in ASD. To this end, we explored the interactions between intestinal microbiota, gut physiology and social behavior in a BTBR T+Itpr3tf/J mouse model of ASD. Here we show that a reduction in the relative abundance of very particular bacterial taxa in the BTBR gut - namely, bile-metabolizing Bifidobacterium and Blautia species, - is associated with deficient bile acid and tryptophan metabolism in the intestine, marked gastrointestinal dysfunction, as well as impaired social interactions in BTBR mice. Together these data support the concept of targeted manipulation of the gut microbiota for reversing gastrointestinal and behavioral symptomatology in ASD, and offer specific plausible targets in this endeavor.


Asunto(s)
Trastorno del Espectro Autista/complicaciones , Bacterias/clasificación , Ácidos y Sales Biliares/metabolismo , Enfermedades Gastrointestinales/microbiología , Triptófano/metabolismo , Animales , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Bacterias/aislamiento & purificación , Bifidobacterium/clasificación , Bifidobacterium/aislamiento & purificación , Modelos Animales de Enfermedad , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/metabolismo , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Masculino , Ratones , Microbiota
20.
Biol Psychiatry ; 82(7): 472-487, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28242013

RESUMEN

BACKGROUND: The realization that the microbiota-gut-brain axis plays a critical role in health and disease, including neuropsychiatric disorders, is rapidly advancing. Nurturing a beneficial gut microbiome with prebiotics, such as fructo-oligosaccharides (FOS) and galacto-oligosaccharides (GOS), is an appealing but underinvestigated microbiota manipulation. Here we tested whether chronic prebiotic treatment modifies behavior across domains relevant to anxiety, depression, cognition, stress response, and social behavior. METHODS: C57BL/6J male mice were administered FOS, GOS, or a combination of FOS+GOS for 3 weeks prior to testing. Plasma corticosterone, microbiota composition, and cecal short-chain fatty acids were measured. In addition, FOS+GOS- or water-treated mice were also exposed to chronic psychosocial stress, and behavior, immune, and microbiota parameters were assessed. RESULTS: Chronic prebiotic FOS+GOS treatment exhibited both antidepressant and anxiolytic effects. Moreover, the administration of GOS and the FOS+GOS combination reduced stress-induced corticosterone release. Prebiotics modified specific gene expression in the hippocampus and hypothalamus. Regarding short-chain fatty acid concentrations, prebiotic administration increased cecal acetate and propionate and reduced isobutyrate concentrations, changes that correlated significantly with the positive effects seen on behavior. Moreover, FOS+GOS reduced chronic stress-induced elevations in corticosterone and proinflammatory cytokine levels and depression-like and anxiety-like behavior in addition to normalizing the effects of stress on the microbiota. CONCLUSIONS: Taken together, these data strongly suggest a beneficial role of prebiotic treatment for stress-related behaviors. These findings strengthen the evidence base supporting therapeutic targeting of the gut microbiota for brain-gut axis disorders, opening new avenues in the field of nutritional neuropsychopharmacology.


Asunto(s)
Ansiolíticos/uso terapéutico , Antidepresivos/uso terapéutico , Ansiedad/terapia , Depresión/terapia , Oligosacáridos/uso terapéutico , Prebióticos , Animales , Ansiedad/sangre , Ansiedad/etiología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Corticosterona/sangre , Depresión/sangre , Depresión/etiología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Expresión Génica/efectos de los fármacos , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neurotransmisores/sangre , Nocicepción/efectos de los fármacos , Estrés Psicológico/complicaciones , Triptófano/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...