Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
mSystems ; 8(5): e0033223, 2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37668401

RESUMEN

IMPORTANCE: The development of probiotic therapies targeted at the small intestinal microbiota represents a significant advancement in the field of probiotic interventions. This region poses unique opportunities due to its low number of gut microbiota, along with the presence of heightened immune and metabolic host responses. However, progress in this area has been hindered by a lack of detailed understanding regarding the molecular mechanisms through which probiotics exert their effects in the small intestine. Our study, utilizing a synthetic community of three small intestinal bacterial strains and the addition of two different probiotic species, and kynurenine as a representative dietary or endogenously produced compound, highlights the importance of selecting probiotic species with diverse genetic capabilities that complement the functional capacity of the resident microbiota, or alternatively, constructing a multispecies formula. This approach holds great promise for the development of effective probiotic therapies and underscores the need to consider the functional capacity of probiotic species when designing interventions.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Probióticos , Probióticos/farmacología , Intestino Delgado , Redes y Vías Metabólicas
2.
Gut Microbes ; 15(1): 2232506, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37417553

RESUMEN

The gut microbiota plays a pivotal role in health and disease. The use of probiotics as microbiota-targeted therapies is a promising strategy to improve host health. However, the molecular mechanisms involved in such therapies are often not well understood, particularly when targeting the small intestinal microbiota. In this study, we investigated the effects of a probiotic formula (Ecologic®825) on the adult human small intestinal ileostoma microbiota. The results showed that supplementation with the probiotic formula led to a reduction in the growth of pathobionts, such as Enterococcaceae and Enterobacteriaceae, and a decrease in ethanol production. These changes were associated with significant alterations in nutrient utilization and resistance to perturbations. These probiotic mediated alterations which coincided with an initial increase in lactate production and decrease in pH were followed by a sharp increase in the levels of butyrate and propionate. Moreover, the probiotic formula increased the production of multiple N-acyl amino acids in the stoma samples. The study demonstrates the utility of network theory in identifying novel microbiota-targeted therapies and improving existing ones. Overall, the findings provide insights into the dynamic molecular mechanisms underlying probiotic therapies, which can aid in the development of more effective treatments for a range of conditions.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Probióticos , Adulto , Humanos , Probióticos/farmacología , Propionatos/farmacología , Enterobacteriaceae
3.
Nutrients ; 15(11)2023 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-37299530

RESUMEN

Probiotic supplements are increasingly being used to target the gut microbiome with a view to improving cognitive and psychological function via the gut-brain axis. One possible mechanism behind the effect of probiotics is through alterations to microbially-derived metabolites including short-chain fatty acids (SCFA) and neurotransmitters. However, research to date has largely been conducted in animal models or under conditions irrelevant to the human gastrointestinal tract (GIT). The aim of the current work was therefore to use anaerobic, pH controlled in vitro batch cultures to (a) assess the production of neuroactive metabolites in human faecal microbiota under conditions relevant to the human GIT, and (b) to explore how several pre-selected probiotic strains may affect bacterial composition and metabolite production. Enumeration of bacteria was assessed using fluorescence in situ hybridisation with flow cytometry, and concentrations of SCFAs and neurotransmitters were measured using gas chromatography and liquid chromatography mass spectroscopy, respectively. GABA, serotonin, tryptophan, and dopamine were successfully detected, suggesting some level of microbial derivation. The addition of Lactococcus lactis W58 and Lactobacillus rhamnosus W198 resulted in a significant increase in lactate after 8 h of fermentation, while no significant effect of probiotics on bacterial composition or neurotransmitter production was found.


Asunto(s)
Microbiota , Probióticos , Humanos , Animales , Técnicas de Cultivo Celular por Lotes , Suplementos Dietéticos , Bacterias/genética , Bacterias/metabolismo , Heces/microbiología
4.
J Psychiatr Res ; 151: 507-515, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35636025

RESUMEN

The brain-gut axis is increasingly recognized as an important contributing factor in the onset and progression of severe mental illnesses such as schizophrenia spectrum disorders and bipolar disorder. This study investigates associations between levels of faecal metabolites identified using 1H-NMR, clinical parameters, and dietary components of forty-two individuals diagnosed in a transdiagnostic approach to have severe mental illness. Faecal levels of the amino acids; alanine, leucine, and valine showed a significant positive correlation with psychiatric symptom severity as well as with dairy intake. Overall, this study proposes a diet-induced link between the brain-gut axis and the severity of psychiatric symptoms, which could be valuable in the design of novel dietary or therapeutic interventions to improve psychiatric symptoms.


Asunto(s)
Trastorno Bipolar , Trastornos Mentales , Esquizofrenia , Aminoácidos , Trastorno Bipolar/tratamiento farmacológico , Dieta , Humanos , Trastornos Mentales/diagnóstico , Esquizofrenia/tratamiento farmacológico
5.
Front Psychiatry ; 13: 879491, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35573324

RESUMEN

Alzheimer's disease (AD) is a global public health priority as with aging populations, its prevalence is expected to rise even further in the future. The brain and gut are in close communication through immunological, nervous and hormonal routes, and therefore, probiotics are examined as an option to influence AD hallmarks, such as plaques, tangles, and low grade inflammation. This study aimed to provide an overview of the available animal evidence on the effect of different probiotics on gut microbiota composition, short chain fatty acids (SCFAs), inflammatory markers, Amyloid-ß (Aß), and cognitive functioning in AD animal models. A systematic literature search was performed in PubMed, SCOPUS, and APA PsychInfo. Articles were included up to May 2021. Inclusion criteria included a controlled animal study on probiotic supplementation and at least one of the abovementioned outcome variables. Of the eighteen studies, most were conducted in AD male mice models (n = 9). Probiotics of the genera Lactobacillus and Bifidobacterium were used most frequently. Probiotic administration increased species richness and/or bacterial richness in the gut microbiota, increased SCFAs levels, reduced inflammatory markers, and improved cognitive functioning in AD models in multiple studies. The effect of probiotic administration on Aß remains ambiguous. B. longum (NK46), C. butyricum, and the mixture SLAB51 are the most promising probiotics, as positive improvements were found on almost all outcomes. The results of this animal review underline the potential of probiotic therapy as a treatment option in AD.

6.
Neurosci Biobehav Rev ; 128: 311-327, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34171323

RESUMEN

Recently the scientific community has seen a growing interest in the role of the gut-brain axis and, in particular, how probiotic supplementation may influence neural function and behaviour via manipulation of the gut microbiota. The purpose of this review was to systematically review the current literature exploring the effect of probiotic intervention on cognitive function. PsychINFO, Web of Science, PubMed and Google Scholar were searched for human trials. Studies selected for inclusion administered a probiotic intervention and included at least one behavioural measure of cognitive performance. A total of 30 experimental papers were included, exploring the effect of probiotics across a variety of ages, populations and cognitive domains. The evidence suggests there may be potential for probiotics to enhance cognitive function or attenuate cognitive decline, particularly in clinically relevant adult populations for whom cognitive dysfunction may be present. However, the limited number of studies and the quality of the existing research makes it challenging to interpret the data. Further research is clearly warranted. PROSPERO: CRD42020164820.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Adulto , Encéfalo , Cognición , Humanos , Longevidad , Probióticos/uso terapéutico
7.
Transl Psychiatry ; 11(1): 300, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34016947

RESUMEN

Stress negatively affects cognitive performance. Probiotics remediate somatic and behavioral stress responses, hypothetically by acting on the gut microbiota. Here, in exploratory analyses, we assessed gut microbial alterations after 28-days supplementation of multi-strain probiotics (EcologicBarrier consisting of Lactobacilli, Lactococci, and Bifidobacteria in healthy, female subjects (probiotics group n = 27, placebo group n = 29). In an identical pre-session and post-session, subjects performed a working memory task before and after an acute stress intervention. Global gut microbial beta diversity changed over time, but we were not able to detect differences between intervention groups. At the taxonomic level, Time by Intervention interactions were not significant after multiple comparison correction; the relative abundance of eight genera in the probiotics group was higher (uncorrected) relative to the placebo group: Butyricimonas, Parabacteroides, Alistipes, Christensenellaceae_R-7_group, Family_XIII_AD3011_group, Ruminococcaceae_UCG-003, Ruminococcaceae_UCG-005, and Ruminococcaceae_UCG-010. In a second analysis step, association analyses were done only within this selection of microbial genera, revealing the probiotics-induced change in genus Ruminococcaceae_UCG-003 was significantly associated with probiotics' effect on stress-induced working memory changes (rspearman(27) = 0.565; pFDR = 0.014) in the probiotics group only and independent of potential confounders (i.e., age, BMI, and baseline dietary fiber intake). That is subjects with a higher increase in Ruminococcaceae_UCG-003 abundance after probiotics were also more protected from negative effects of stress on working memory after probiotic supplementation. The bacterial taxa showing an increase in relative abundance in the probiotics group are plant fiber degrading bacteria and produce short-chain fatty acids that are known for their beneficial effect on gut and brain health, e.g., maintaining intestinal-barrier and blood-brain-barrier integrity. This study shows that gut microbial alterations, modulated through probiotics use, are related to improved cognitive performance in acute stress circumstances.


Asunto(s)
Cognición , Microbioma Gastrointestinal , Probióticos , Bacterias , Femenino , Humanos , Estrés Psicológico , Adulto Joven
8.
Artículo en Inglés | MEDLINE | ID: mdl-33186639

RESUMEN

The endocannabinoid system is a metabolic pathway involved in the communication between the gut microbiota and the host. In the gut, the endocannabinoid system regulates the integrity of the intestinal barrier. A compromised integrity of the intestinal barrier is associated with several disorders such as inflammatory bowel disorder, obesity and major depressive disorder. Decreasing the integrity of the intestinal barrier results in an increased translocation of bacterial metabolites, including lipopolysaccharides, across the epithelial layer of the gut, causing the subsequent inflammation. Targeting the endocannabinoid system in the gut can improve the integrity of the intestinal barrier. Currently, microbial interventions in the form of probiotics are under investigation for the treatment of diseases related to a compromised integrity of the intestinal barrier. However, the role of the endocannabinoid system in the gut is ambiguous since activity of the endocannabinoid system is increased in obesity and decreased in inflammatory bowel disease, emphasizing the need for development of personalized microbial interventions. This review discusses the role of the endocannabinoid system in regulating the gut barrier integrity and highlights current efforts to develop new endocannabinoid-targeted microbial interventions.


Asunto(s)
Eje Cerebro-Intestino/fisiología , Trastorno Depresivo Mayor/metabolismo , Endocannabinoides/metabolismo , Microbioma Gastrointestinal/fisiología , Probióticos/administración & dosificación , Animales , Eje Cerebro-Intestino/efectos de los fármacos , Trastorno Depresivo Mayor/dietoterapia , Trastorno Depresivo Mayor/psicología , Endocannabinoides/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Humanos
9.
J Affect Disord ; 253: 317-326, 2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31078831

RESUMEN

BACKGROUND: Depression is the leading cause of disability worldwide; with evidence suggesting that decreased gut barrier function and inflammation are correlated with depressive symptoms. We conducted a clinical trial to determine the effect of consumption of probiotic supplements (Winclove's Ecologic® Barrier) on depressive symptoms in a sample of participants with mild to severe depression. METHOD: 71 participants were randomly allocated to either probiotic or placebo, which was, consumed daily over eight weeks. Pre- and post-intervention measures of symptoms and vulnerability markers of depression as well as gut microbiota composition were compared. Clinical trial participants were also compared on psychological variables and gut microbiota composition to a non-depressed group (n = 20). RESULTS: All clinical trial participants demonstrated improvement in symptoms, suggesting non-specific therapeutic effects associated with weekly monitoring visits. Participants in the probiotic group demonstrated a significantly greater reduction in cognitive reactivity compared with the placebo group, particularly in the mild/moderate subgroup. Probiotics did not significantly alter the microbiota of depressed individuals, however, a significant correlation was found between Ruminococcus gnavus and one depression metric. LIMITATIONS: There was a high attrition rate, which may be attributed to weekly monitoring visits. Additionally, modulation of the gut microbiota may need more specific testing to distinguish subtle changes. CONCLUSIONS: While microbiota composition was similar between all groups, probiotics did affect a psychological variable associated with susceptibility to depression. Further research is needed to investigate how probiotics can be utilised to modify mental wellbeing, and whether they can act as an adjunct to existing treatments.


Asunto(s)
Depresión/terapia , Suplementos Dietéticos , Probióticos/uso terapéutico , Adulto , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Masculino , Placebos/uso terapéutico , Probióticos/farmacología , Proyectos de Investigación
10.
Nutrients ; 10(6)2018 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-29914095

RESUMEN

During the postmenopausal period, the risk of cardiovascular diseases is increased in many obese women and is associated with a worse cardiometabolic profile and a sub-chronic low-grade systemic inflammation caused by a gut barrier permeability dysfunction. Here, we tested whether administration of two different dosages of the multispecies probiotic Ecologic® Barrier influenced the cardiometabolic biochemical parameters and lipopolysaccharide levels, the latter used as a marker of increased gut permeability in obese postmenopausal women. A total of 81 obese Caucasian postmenopausal women participated in the trial. The subjects were randomly assigned to three groups that received a placebo, a low dose (LD) (2.5 × 108 colony forming units (CFU) per day), or a high dose (HD) (1 × 1010 CFU per day) of lyophilisate powder containing live multispecies probiotic bacteria. The probiotic supplement was administered each day in two equal portions for 12 weeks. We found significant (p < 0.05) favorable changes (mostly large or medium effects) in the evaluated parameters in both the HD and LD groups but not in the placebo group. In the HD group, lipopolysaccharide, waist, fat mass, subcutaneous fat, uric acid, total cholesterol, triglycerides, low-density lipoprotein cholesterol, glucose, insulin, and insulin-resistant index (HOMA-IR) were improved. Similar changes were observed in the LD group, except for lipopolysaccharide, uric acid, triglycerides, and glucose levels. Additionally, significant differences were observed in both groups in terms of fat percentage and visceral fat. When the mean changes were compared between the three groups, statistically significant differences in lipopolysaccharide levels, uric acid, glucose, insulin, and HOMA-IR were found. Post hoc tests revealed significant differences in the mean changes (mostly medium effects) between the HD and LD groups for uric acid, glucose, insulin, and HOMA-IR. In the 12-week randomized, placebo-controlled, double-blind intervention, we observed that supplementation with the multispecies probiotic Ecologic® Barrier favorably affected the risk factors in a dose-dependent manner, showing beneficial effects on the cardiometabolic parameters and gut permeability of the patients. Our results suggest that this product can be effective in the prevention and treatment of cardiovascular diseases in obese postmenopausal women.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Lipopolisacáridos/metabolismo , Obesidad/metabolismo , Probióticos/administración & dosificación , Probióticos/farmacología , Adulto , Anciano , Enfermedades Cardiovasculares/prevención & control , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Persona de Mediana Edad , Posmenopausia
11.
Ann Transl Med ; 6(3): 39, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29610731

RESUMEN

Microscopic colitis (MC) is a chronic inflammatory bowel disease (IBD) with little in terms of endoscopic abnormalities and is frequently associated with other autoimmune diseases. The peak incidence of the disease is in middle aged or older populations, mostly females. The pathogenesis of MC is complex, multifactorial and poorly understood. Current concepts revolve around innate immunity or microbiome alterations as well as gut barrier dysfunction, all of which lead to the development of subtle inflammatory lesions in gut mucosa. The results of numerous basic and clinical studies involving molecular techniques as well as advanced endoscopic imaging revealed the important role of both intrinsic (e.g., hormonal) as well as extrinsic (e.g., NSAIDs and PPIs) factors in the modulation of gastrointestinal microbiome and MC pathogenesis. Capsule endoscopy as well confocal endomicroscopy imaging, alongside standard endoscopic techniques offer new tools in the evaluation of MC patients and allow their better stratification for novel treatment protocols based on modulation of gut microbiome and barrier function.

12.
Expert Rev Gastroenterol Hepatol ; 11(10): 961-978, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28618973

RESUMEN

INTRODUCTION: Over the last decade, remarkable progress has been made in the understanding of disease pathophysiology. Many new theories expound on the importance of emerging factors such as microbiome influences, genomics/omics, stem cells, innate intestinal immunity or mucosal barrier complexities. This has introduced a further dimension of uncertainty into clinical decision-making, but equally, may shed some light on less well-understood and difficult to manage conditions. Areas covered: Comprehensive review of the literature on gut barrier and microbiome relevant to small bowel pathology. A PubMed/Medline search from 1990 to April 2017 was undertaken and papers from this range were included. Expert commentary: The scenario of clinical uncertainty is well-illustrated by functional gastrointestinal disorders (FGIDs). The movement towards achieving a better understanding of FGIDs is expressed in the Rome IV guidelines. Novel diagnostic and therapeutic protocols focused on the GB and SB microbiome can facilitate diagnosis, management and improve our understanding of the underlying pathological mechanisms in FGIDs.


Asunto(s)
Permeabilidad de la Membrana Celular/fisiología , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/fisiopatología , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/fisiopatología , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/terapia , Humanos , Uniones Intercelulares/fisiología , Intestino Delgado/fisiopatología , Medicina de Precisión
13.
Gut Pathog ; 9: 12, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28286570

RESUMEN

BACKGROUND: One of the working mechanisms of probiotic bacteria is their ability to compete with pathogens. To define the probiotic properties of seven Lactic Acid Bacteria (LAB) strains, we tested them for survival in simulated gastro-intestinal conditions, antimicrobial activities, co-aggregative abilities, and interferences studies against five human intestinal pathogens (Salmonella enteritidis ATCC 13076, Listeria monocytogenes ATCC 7644, Escherichia coli O157: H7 ATCC 35150, Cronobacter sakazakii ATCC 29544 and Campylobacter jejuni ATCC 33291). RESULTS: The LAB strains were able to survive the stomach simulated conditions, and varied in their abilities to survive the small intestinal-simulated conditions. The strains showed antibiotic susceptibility profiles with values equal or below the breakpoints set by the European Food and Safety Authority. The LAB cell-free cultures supernatants showed antimicrobial activities, with inhibition zones ranging from 10.0 to 17.2 mm. All the LAB strains showed moderate auto-aggregation abilities while the greatest co-aggregation abilities were observed for Bifidobacterium bifidum W23, Lactobacillus plantarum W21 and Lactobacillus rhamnosus W71. The individual LAB strains showed strain-specific abilities to reduce the invasion of intestinal pathogens in an interference model with Caco-2 cells. Increased invasion inhibition was found when different combinations of LAB strains were used in the interference tests. CONCLUSION: The LAB strains examined in this study may protect the intestinal epithelium through a series of barriers (antimicrobial activity, co-aggregation with pathogens, adherence) and interference mechanisms. Consequently, these LAB strains may be considered candidates for prophylactic use to prevent intestinal infections.

14.
Microb Biotechnol ; 9(4): 452-65, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27231133

RESUMEN

Lactobacillus plantarum WCFS1 is one of the best studied Lactobacilli, notably as its genome was unravelled over 12 years ago. L. plantarum WCFS1 can be grown to high densities, is amenable to genetic transformation and highly robust with a relatively high survival rate during the gastrointestinal passage. In this review, we present and discuss the main insights provided by the functional genomics research on L. plantarum WCFS1 with specific attention for the molecular mechanisms related to its interaction with the human host and its potential to modify the immune system, and induce other health-related benefits. Whereas most insight has been gained in mouse and other model studies, only five human studies have been reported with L. plantarum WCFS1. Hence NCIMB 8826 (the parental strain of L. plantarum WCFS1) in human trials as to capitalize on the wealth of knowledge that is summarized here.


Asunto(s)
Tracto Gastrointestinal/microbiología , Lactobacillus plantarum/genética , Lactobacillus plantarum/fisiología , Probióticos/administración & dosificación , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Lactobacillus plantarum/crecimiento & desarrollo , Lactobacillus plantarum/inmunología , Ratones , Resultado del Tratamiento
15.
Brain Behav Immun ; 48: 258-64, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25862297

RESUMEN

BACKGROUND: Recent insights into the role of the human microbiota in cognitive and affective functioning have led to the hypothesis that probiotic supplementation may act as an adjuvant strategy to ameliorate or prevent depression. OBJECTIVE: Heightened cognitive reactivity to normal, transient changes in sad mood is an established marker of vulnerability to depression and is considered an important target for interventions. The present study aimed to test if a multispecies probiotic containing Bifidobacterium bifidum W23, Bifidobacterium lactis W52, Lactobacillus acidophilus W37, Lactobacillus brevis W63, Lactobacillus casei W56, Lactobacillus salivarius W24, and Lactococcus lactis (W19 and W58) may reduce cognitive reactivity in non-depressed individuals. DESIGN: In a triple-blind, placebo-controlled, randomized, pre- and post-intervention assessment design, 20 healthy participants without current mood disorder received a 4-week probiotic food-supplement intervention with the multispecies probiotics, while 20 control participants received an inert placebo for the same period. In the pre- and post-intervention assessment, cognitive reactivity to sad mood was assessed using the revised Leiden index of depression sensitivity scale. RESULTS: Compared to participants who received the placebo intervention, participants who received the 4-week multispecies probiotics intervention showed a significantly reduced overall cognitive reactivity to sad mood, which was largely accounted for by reduced rumination and aggressive thoughts. CONCLUSION: These results provide the first evidence that the intake of probiotics may help reduce negative thoughts associated with sad mood. Probiotics supplementation warrants further research as a potential preventive strategy for depression.


Asunto(s)
Afecto/efectos de los fármacos , Cognición/efectos de los fármacos , Trastorno Depresivo/psicología , Probióticos/farmacología , Adolescente , Bifidobacterium , Suplementos Dietéticos , Método Doble Ciego , Femenino , Humanos , Lactobacillus , Lactobacillus acidophilus , Masculino , Conducta Social , Adulto Joven
16.
Microb Ecol Health Dis ; 26: 25877, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25651995

RESUMEN

BACKGROUND: The intestinal microbiota composition varies between healthy and diseased individuals for numerous diseases. Although any cause or effect relationship between the alterations in the gut microbiota and disease is not always clear, targeting the intestinal microbiota might offer new possibilities for prevention and/or treatment of disease. OBJECTIVE: Here we review some examples of manipulating the intestinal microbiota by prebiotics, probiotics, and fecal microbial transplants. RESULTS: Prebiotics are best known for their ability to increase the number of bifidobacteria. However, specific prebiotics could potentially also stimulate other species they can also stimulate other species associated with health, like Akkermansia muciniphila, Ruminococcus bromii, the Roseburia/Enterococcus rectale group, and Faecalibacterium prausnitzii. Probiotics have beneficial health effects for different diseases and digestive symptoms. These effects can be due to the direct effect of the probiotic bacterium or its products itself, as well as effects of the probiotic on the resident microbiota. Probiotics can influence the microbiota composition as well as the activity of the resident microbiota. Fecal microbial transplants are a drastic intervention in the gut microbiota, aiming for total replacement of one microbiota by another. With numerous successful studies related to antibiotic-associated diarrhea and Clostridium difficile infection, the potential of fecal microbial transplants to treat other diseases like inflammatory bowel disease, irritable bowel syndrome, and metabolic and cardiovascular disorders is under investigation. CONCLUSIONS: Improved knowledge on the specific role of gut microbiota in prevention and treatment of disease will help more targeted manipulation of the intestinal microbiota. Further studies are necessary to see the (long term) effects for health of these interventions.

17.
Am J Reprod Immunol ; 73(1): 1-11, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25250861

RESUMEN

As studies uncover the breadth of microbes associated with human life, opportunities will emerge to manipulate and augment their functions in ways that improve health and longevity. From involvement in the complexities of reproduction and fetal/infant development, to delaying the onset of disease, and indeed countering many maladies, microbes offer hope for human well-being. Evidence is emerging to suggest that microbes may play a beneficial role in body sites traditionally viewed as being sterile. Although further evidence is required, we propose that much of medical dogma is about to change significantly through recognition and understanding of these hitherto unrecognized microbe-host interactions. A meeting of the International Scientific Association for Probiotics and Prebiotics held in Aberdeen, Scotland (June 2014), presented new views and challenged established concepts on the role of microbes in reproduction and health of the mother and infant. This article summarizes some of the main aspects of these discussions.


Asunto(s)
Interacciones Huésped-Patógeno/fisiología , Microbiota/fisiología , Reproducción , Femenino , Humanos/microbiología , Lactante , Madres , Prebióticos , Probióticos , Escocia
18.
Front Neurol ; 5: 241, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25484876

RESUMEN

Recent studies suggest that migraine may be associated with gastrointestinal (GI) disorders, including irritable bowel syndrome (IBS), inflammatory bowel syndrome, and celiac disease. Here, an overview of the associations between migraine and GI disorders is presented, as well as possible mechanistic links and clinical implications. People who regularly experience GI symptoms have a higher prevalence of headaches, with a stronger association with increasing headache frequency. Children with a mother with a history of migraine are more likely to have infantile colic. Children with migraine are more likely to have experienced infantile colic compared to controls. Several studies demonstrated significant associations between migraine and celiac disease, inflammatory bowel disease, and IBS. Possible underlying mechanisms of migraine and GI diseases could be increased gut permeability and inflammation. Therefore, it would be worthwhile to investigate these mechanisms further in migraine patients. These mechanisms also give a rationale to investigate the effects of the use of pre- and probiotics in migraine patients.

19.
J Bacteriol ; 194(23): 6638, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23144392

RESUMEN

We announce the draft genome sequence of Lactobacillus casei W56 in one contig. This strain shows immunomodulatory and probiotic properties. The strain is also an ingredient of commercially available probiotic products.


Asunto(s)
ADN Bacteriano/química , ADN Bacteriano/genética , Genoma Bacteriano , Lacticaseibacillus casei/genética , Análisis de Secuencia de ADN , Factores Inmunológicos/farmacología , Lacticaseibacillus casei/inmunología , Lacticaseibacillus casei/aislamiento & purificación , Lacticaseibacillus casei/fisiología , Datos de Secuencia Molecular , Probióticos/farmacología
20.
Gastroenterol Res Pract ; 2011: 161358, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22114588

RESUMEN

Gastrointestinal (GI) dysfunction has been reported in a substantial number of children with autism spectrum disorders (ASD). Activation of the mucosal immune response and the presence of abnormal gut microbiota are repeatedly observed in these children. In children with ASD, the presence of GI dysfunction is often associated with increased irritability, tantrums, aggressive behaviour, and sleep disturbances. Moreover, modulating gut bacteria with short-term antibiotic treatment can lead to temporary improvement in behavioral symptoms in some individuals with ASD. Probiotics can influence microbiota composition and intestinal barrier function and alter mucosal immune responses. The administration of probiotic bacteria to address changes in the microbiota might, therefore, be a useful novel therapeutic tool with which to restore normal gut microbiota, reduce inflammation, restore epithelial barrier function, and potentially ameliorate behavioural symptoms associated with some children with ASD. In this review of the literature, support emerges for the clinical testing of probiotics in ASD, especially in the context of addressing GI symptoms.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...