Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 812
Filtrar
1.
Arch. argent. pediatr ; 122(5): e202310168, oct. 2024.
Artículo en Inglés, Español | LILACS, BINACIS | ID: biblio-1571504

RESUMEN

Los postbióticos fueron definidos en 2021 por la Asociación Científica Internacional de Probióticos y Prebióticos (ISAPP) como "una preparación de microorganismos inanimados y/o sus componentes celulares capaces de conferir un efecto benéfico al hospedador". El campo de los postbióticos es un área nueva dentro de la familia de los bióticos; se han desarrollado ya numerosos productos con aplicaciones clínicas, como la estimulación inmunológica, el manejo de diarreas en niños y adultos, el abordaje del intestino irritable, además de tres fórmulas infantiles. En particular, las fórmulas infantiles con postbióticos obtenidos a partir de la fermentación de la leche con Bifidobacterium breve C50 y Streptococcus thermophilus O65, y sus metabolitos, incluido el oligosacárido 3'-GL, han demostrado seguridad y contribución al desarrollo de la microbiota intestinal y el sistema inmune asociado al intestino. Estas modificaciones contribuyen a la prevención y el manejo de los trastornos funcionales digestivos del lactante.


Postbiotics were defined in 2021 by the International Scientific Association for Probiotics and Prebiotics (ISAPP) as a "preparation of inanimate microorganisms and/or their cellular components that confers a health benefit to the host." The field of postbiotics is a new area within the biotics family; numerous products have already been developed for clinical applications, such as immune stimulation, the management of diarrhea in children and adults, the management of irritable bowel syndrome, and 3 infant formulas. In particular, infant formulas with postbiotics obtained from milk fermented with Bifidobacterium breve C50 and Streptococcus thermophilus O65 ­and their metabolites­, including the oligosaccharide 3'-GL, have demonstrated to be safe and to contribute to the development of the gut microbiota and the gutassociated immune system. These modifications help to prevent and manage functional gastrointestinal disorders in infants.


Asunto(s)
Humanos , Lactante , Probióticos , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/terapia , Fórmulas Infantiles , Streptococcus thermophilus , Diarrea/microbiología , Diarrea/terapia , Prebióticos/administración & dosificación , Microbioma Gastrointestinal , Bifidobacterium breve , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/terapia
2.
Curr Microbiol ; 81(11): 386, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39358608

RESUMEN

High-density stress can lead to dysbiotic microbiota, affecting the organism's metabolic, and protective functions. Agavin is a fructan with prebiotic properties that regulate the gut microbiota by promoting the growth of beneficial bacteria. This study evaluated the effect of agavin on the gut microbiota using Next-Generation Sequencing (NGS) and its correlation with the growth parameters. Four groups of fish were fed different diets: a control diet (negative and positive control), without agavin supplementation, and two experimental diets supplemented with agavin at 20 g kg-1 and 40 g kg-1. Nile tilapias (1.04 g ± 0.01 g) were fed for 110 days. After 90 days of feeding, fish were subjected to high-density stress (63 kg m-3) for 20 days, except for the negative control. NGS detected 1579 different operational taxonomic units in the samples. In the correlation analysis of growth parameters, the families Vibrionaceae and Methyloligillaceae showed a positive correlation with fish growth parameters, these results may serve to know the relation of agavin and microbiota on the growth performance, as well as the metabolic activities of families in tilapia. Furthermore, high-density stress and agavin supplementation modify the gut microbiota in tilapia. At a low-density, supplementation with 20 g kg-1 agavin promoted the growth of the potentially beneficial families Sphingomonadaceae, Oxalobacteriaceae, and Chitinophagaceae; at high densities, reduced the abundance of pathogenic families (Vibrionaceae and Aeromonadaceae). These results suggest that, under stress conditions, agavin can stimulate the growth of potentially beneficial bacteria and reduce the growth of potentially pathogenic bacteria, suggesting its potential use as a prebiotic in aquaculture.


Asunto(s)
Alimentación Animal , Bacterias , Cíclidos , Suplementos Dietéticos , Fructanos , Microbioma Gastrointestinal , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Cíclidos/microbiología , Cíclidos/crecimiento & desarrollo , Alimentación Animal/análisis , Fructanos/farmacología , Fructanos/metabolismo , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/efectos de los fármacos , Dieta/veterinaria , Prebióticos/administración & dosificación , Secuenciación de Nucleótidos de Alto Rendimiento
3.
Int J Mol Sci ; 25(19)2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39408940

RESUMEN

Type 1 Diabetes Mellitus (T1DM) is a chronic autoimmune disease that results in the destruction of pancreatic ß cells, leading to hyperglycaemia and the need for lifelong insulin therapy. Although genetic predisposition and environmental factors are considered key contributors to T1DM, the exact causes of the disease remain partially unclear. Recent evidence has focused on the relationship between the gut, the oral cavity, immune regulation, and systemic inflammation. In individuals with T1DM, changes in the gut and oral microbial composition are commonly observed, indicating that dysbiosis may contribute to immune dysregulation. Gut dysbiosis can influence the immune system through increased intestinal permeability, altered production of short chain fatty acids (SCFAs), and interactions with the mucosal immune system, potentially triggering the autoimmune response. Similarly, oral dysbiosis may contribute to the development of systemic inflammation and thus influence the progression of T1DM. A comprehensive understanding of these relationships is essential for the identification of biomarkers for early diagnosis and monitoring, as well as for the development of therapies aimed at restoring microbial balance. This review presents a synthesis of current research on the connection between T1DM and microbiome dysbiosis, with a focus on the gut and oral microbiomes in pediatric populations. It explores potential mechanisms by which microbial dysbiosis contributes to the pathogenesis of T1DM and examines the potential of microbiome-based therapies, including probiotics, prebiotics, synbiotics, and faecal microbiota transplantation (FMT). This complex relationship highlights the need for longitudinal studies to monitor microbiome changes over time, investigate causal relationships between specific microbial species and T1DM, and develop personalised medicine approaches.


Asunto(s)
Diabetes Mellitus Tipo 1 , Disbiosis , Microbioma Gastrointestinal , Humanos , Diabetes Mellitus Tipo 1/microbiología , Disbiosis/microbiología , Niño , Boca/microbiología , Probióticos/uso terapéutico , Trasplante de Microbiota Fecal , Prebióticos/administración & dosificación , Microbiota
4.
Nutrients ; 16(19)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39408300

RESUMEN

BACKGROUND/OBJECTIVES: Prebiotics and probiotics have been reported to improve symptoms of irritable bowel syndrome (IBS). Nevertheless, the effects of prebiotics/probiotics can vary depending on the IBS subtypes. The purpose of this study was to investigate the effects of personalized prebiotic and probiotic supplements based on intestinal microbiota and IBS subtypes in patients. METHODS: Patients with diarrhea-type IBS (IBS-D), constipation-type IBS (IBS-C), and mixed-type IBS (IBS-M) were enrolled (n = 40 per group; total: n = 120). Personalized prebiotic and probiotic supplements were determined according to the IBS subtypes and intestinal microbiota. The patients received supplements for 4 weeks. The primary outcome was the change in the IBS-severity scoring system from baseline to week 4. RESULTS: The IBS-severity scoring system significantly decreased in all patients (-38.0 [95% confidence interval (CI): -53.6, -22.4]; p < 0.001), in patients with IBS-D (-44.5 [95% CI: -70.6, -18.5]; p = 0.004) and IBS-C (-51.2 [95% CI: -79.4, -22.9]; p = 0.002), but not in those with IBS-M (-20.0 [95% CI: -48.0, 8.1]; p = 0.47). In this study, no serious adverse events were observed that had a causal relationship with the intervention. CONCLUSIONS: In conclusion, personalized prebiotic and probiotic supplements selected according to individual intestinal microbiota and IBS subtype may alleviate the severity of IBS symptoms, particularly in patients with IBS-C and IBS-D.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal , Síndrome del Colon Irritable , Prebióticos , Probióticos , Humanos , Síndrome del Colon Irritable/terapia , Síndrome del Colon Irritable/dietoterapia , Probióticos/administración & dosificación , Probióticos/uso terapéutico , Prebióticos/administración & dosificación , Femenino , Masculino , Adulto , Persona de Mediana Edad , Resultado del Tratamiento , Estreñimiento/terapia , Diarrea , Índice de Severidad de la Enfermedad , Adulto Joven
5.
Clin Med Res ; 22(2): 97-106, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39231625

RESUMEN

Background: Although various treatments are developed against COVID-19 variants, probiotic, and synbiotic adjunct therapy with several benefits such as safety, low cost, and availability could be needed for preventing or treating COVID-19 infection.Objective: The present systematic review aimed to assess the prophylactic efficacy of the probiotic, prebiotic, and synbiotic administration against COVID-19.Methods: The protocol of this systematic review was registered at the PROSPERO (Code number: CRD42023418900). The Scopus, Cochrane Library, Web of Sciences, and PubMed databases were systematically searched to define the clinical trials published up to November 2022 in the English language. The comparison of the incidence of COVID-19 disease and levels of specific antibodies against SARS-cov2 between the intervention and placebo groups were evaluated in this systematic review.Results: According to the five included trials, four indicated the incidence of COVID-19, and no significant differences were observed between the probiotic and placebo groups during 1, 2, or 3 months of follow-up in the mentioned studies. Regarding the antibody assays against SARS-Cov2 including IgM, IgG, or IgA reported by three eligible trials, there were no significant differences between the intervention and control groups.Conclusion: It seems that the administration of single or multi-strain probiotics or synbiotics had no prophylactic effects in different populations such as high-risk staff exposed to COVID-19, elderly nursing home residents, healthy adults, and household contact with COVID-19 patients during 1-to-3-months of intervention.


Asunto(s)
COVID-19 , Prebióticos , Probióticos , Simbióticos , Humanos , COVID-19/prevención & control , Prebióticos/administración & dosificación , Probióticos/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Simbióticos/administración & dosificación
6.
Gut Microbes ; 16(1): 2402547, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39287045

RESUMEN

Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory demyelination occurring in the central nervous system (CNS). Inulin is a common prebiotic that can improve metabolic disorders by modulating the gut microbiota. However, its capacity to affect CNS autoimmunity is poorly recognized. Experimental autoimmune encephalomyelitis (EAE) is a classical mouse model of MS. Herein, we found that oral administration of inulin ameliorated the severity EAE in mice, accompanied by reductions in inflammatory cell infiltration and demyelination in the CNS. These reductions were associated with decreased proportion and numbers of Th17 cells in brain and spleen. Consistent with the findings, the serum concentrations of IL-17, IL-6, and TNF-α were reduced in inulin treated EAE mice. Moreover, the proliferation of auto-reactive lymphocytes, against MOG35-55 antigen, was attenuated ex vivo. Mechanistically, inulin treatment altered the composition of gut microbiota. It increased Lactobacillus and Dubosiella whereas decreased g_Prevotellaceae_NK3B31_group at the genus level, alongside with elevated concentration of butyric acid in fecal content and serum. In vitro, butyrate, but not inulin, could inhibit the activation of MOG35-55 stimulated lymphocytes. Furthermore, fecal microbiota transplantation assay confirmed that fecal contents of inulin-treated normal mice had an ameliorative effect on EAE mice. In contrast, antibiotic cocktail (ABX) treatment diminished the therapeutic effect of inulin in EAE mice as well as the reduction of Th17 cells, while supplementation with Lactobacillus reuteri restored the amelioration effect. These results confirmed that the attenuation of inulin on Th17 cells and inflammatory demyelination in EAE mice was dependent on its modulation on gut microbiota and metabolites. Our findings provide a potential therapeutic regimen for prebiotic inulin supplementation in patients with multiple sclerosis.


Asunto(s)
Autoinmunidad , Encefalomielitis Autoinmune Experimental , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Inulina , Ratones Endogámicos C57BL , Esclerosis Múltiple , Prebióticos , Células Th17 , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Inulina/administración & dosificación , Inulina/farmacología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/microbiología , Células Th17/inmunología , Ratones , Prebióticos/administración & dosificación , Femenino , Ácidos Grasos Volátiles/metabolismo , Autoinmunidad/efectos de los fármacos , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/microbiología , Sistema Nervioso Central/inmunología , Bacterias/clasificación , Bacterias/aislamiento & purificación
7.
Pediatr Allergy Immunol ; 35(9): e14226, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39221598

RESUMEN

BACKGROUND: Immunomodulatory proteins in human milk (HM) can shape infant immune development. However, strategies to modulate their levels are currently unknown. This study investigated whether maternal prebiotic supplementation alters the levels of immunomodulatory proteins in HM. METHODS: The study was nested within the SYMBA double-blind randomized controlled trial (ACTRN12615001075572), which investigated the effects of maternal prebiotic (short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides) supplementation from <21 weeks gestation during pregnancy until 6 months postnatal during lactation on child allergic disease risk. Mother-child dyads receiving prebiotics (n = 46) or placebo (n = 54) were included in this study. We measured the levels of 24 immunomodulatory proteins in HM collected at 2, 4, and 6 months. RESULTS: Cluster analysis showed that the overall immunomodulatory protein composition of milk samples from both groups was similar. At 2 months, HM of prebiotic-supplemented women had decreased levels of TGF-ß1 and TSLP (95% CI: -17.4 [-29.68, -2.28] and -57.32 [-94.22, -4.7] respectively) and increased levels of sCD14 (95% CI: 1.81 [0.17, 3.71]), when compared to the placebo group. At 4 months, IgG1 was lower in the prebiotic group (95% CI: -1.55 [-3.55, -0.12]) compared to placebo group. CONCLUSION: This exploratory study shows that prebiotic consumption by lactating mothers selectively alters specific immunomodulatory proteins in HM. This finding is crucial for understanding how prebiotic dietary recommendations for pregnant and lactating women can modify the immune properties of HM and potentially influence infant health outcomes through immune support from breastfeeding.


Asunto(s)
Suplementos Dietéticos , Leche Humana , Prebióticos , Humanos , Leche Humana/inmunología , Leche Humana/química , Prebióticos/administración & dosificación , Femenino , Método Doble Ciego , Embarazo , Lactante , Adulto , Masculino , Lactancia/inmunología , Oligosacáridos/administración & dosificación , Recién Nacido , Lactancia Materna , Citocinas/metabolismo
8.
Int J Mol Sci ; 25(17)2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39273662

RESUMEN

The gut microbiota is one of the most critical factors in human health. It involves numerous physiological processes impacting host health, mainly via immune system modulation. A balanced microbiome contributes to the gut's barrier function, preventing the invasion of pathogens and maintaining the integrity of the gut lining. Dysbiosis, or an imbalance in the gut microbiome's composition and function, disrupts essential processes and contributes to various diseases. This narrative review summarizes key findings related to the gut microbiota in modern multifactorial inflammatory conditions such as ulcerative colitis or Crohn's disease. It addresses the challenges posed by antibiotic-driven dysbiosis, particularly in the context of C. difficile infections, and the development of novel therapies like fecal microbiota transplantation and biotherapeutic drugs to combat these infections. An emphasis is given to restoration of the healthy gut microbiome through dietary interventions, probiotics, prebiotics, and novel approaches for managing gut-related diseases.


Asunto(s)
Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Obesidad , Probióticos , Humanos , Disbiosis/microbiología , Disbiosis/terapia , Obesidad/microbiología , Probióticos/uso terapéutico , Animales , Inflamación/microbiología , Prebióticos/administración & dosificación
9.
Nutrients ; 16(18)2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39339649

RESUMEN

Stress, unhealthy lifestyle, and sleep disturbance worsen cognitive function in mood disorders, prompting a rise in the development of integrative health approaches. The recent investigations in the gut-brain axis field highlight the strong interplay among microbiota, inflammation, and mental health. Thus, this study aimed to investigate a new nutraceutical formulation comprising prebiotics, minerals, and silymarin's impact on microbiota, inflammation, mood, and sleep quality. The study evaluated the LL1 + silymarin capsule supplementation over 180 days in overweight adults. We analyzed the fecal gut microbiota using partial 16S rRNA sequences, measured cytokine expression via CBA, collected anthropometric data, quality of life, and sleep questionnaire responses, and obtained plasma samples for metabolic and hormonal analysis at baseline (T0) and 180 days (T180) post-supplementation. Our findings revealed significant reshaping in gut microbiota composition at the phylum, genus, and species levels, especially in the butyrate-producer bacteria post-supplementation. These changes in gut microbiota were linked to enhancements in sleep quality, mood perception, cytokine expression, and anthropometric measures which microbiota-derived short-chain fatty acids might enhance. The supplementation tested in this study seems to be able to improve microbiota composition, reflecting anthropometrics and inflammation, as well as sleep quality and mood improvement.


Asunto(s)
Afecto , Eje Cerebro-Intestino , Suplementos Dietéticos , Microbioma Gastrointestinal , Silimarina , Calidad del Sueño , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Proyectos Piloto , Afecto/efectos de los fármacos , Masculino , Femenino , Silimarina/farmacología , Adulto , Eje Cerebro-Intestino/efectos de los fármacos , Persona de Mediana Edad , Calidad de Vida , Heces/microbiología , Cápsulas , Citocinas/metabolismo , Citocinas/sangre , Sobrepeso , Prebióticos/administración & dosificación , ARN Ribosómico 16S
11.
Int J Mol Sci ; 25(18)2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39337515

RESUMEN

Diabetic cardiomyopathy is a major etiological factor in heart failure in diabetic patients, characterized by mitochondrial oxidative metabolism dysfunction, myocardial fibrosis, and marked glycogen elevation. The aim of the present study is to evaluate the effect of endurance training and prebiotic xylooligosaccharide (XOS) on the activity of key oxidative enzymes, myocardial collagen, and glycogen distribution as well as some serum biochemical risk markers in streptozotocin-induced type 1 diabetic rats. Male Wistar rats (n = 36) were divided into four diabetic groups (n = 9): sedentary diabetic rats on a normal diet (SDN), trained diabetic rats on a normal diet (TDN), trained diabetic rats on a normal diet with an XOS supplement (TD-XOS), and sedentary diabetic rats with an XOS supplement (SD-XOS). The results show that aerobic training managed to increase the enzyme activity of respiratory Complex I and II and the lactate dehydrogenase in the cardiomyocytes of the diabetic rats. Furthermore, the combination of exercise and XOS significantly decreased the collagen and glycogen content. No significant effects on blood pressure, heart rate or markers of inflammation were detected. These results demonstrate the beneficial effects of exercise, alone or in combination with XOS, on the cardiac mitochondrial enzymology and histopathology of diabetic rats.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Entrenamiento Aeróbico , Glucuronatos , Oligosacáridos , Condicionamiento Físico Animal , Prebióticos , Ratas Wistar , Animales , Ratas , Masculino , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/complicaciones , Glucuronatos/farmacología , Prebióticos/administración & dosificación , Entrenamiento Aeróbico/métodos , Glucógeno/metabolismo , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/prevención & control , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/etiología
12.
Ann N Y Acad Sci ; 1540(1): 61-88, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39283061

RESUMEN

The gut microbiome may be related to the prevalence of overweight and obesity, but high interindividual variability of the human microbiome complicates our understanding. Obesity often occurs concomitantly with micronutrient deficiencies that impair energy metabolism. Microbiota composition is affected by diet. Host-microbiota interactions are bidirectional. We propose three pathways whereby these interactions may modulate the gut microbiome and obesity: (1) ingested compounds or derivatives affecting small intestinal transit, endogenous secretions, digestion, absorption, microbiome balance, and gut barrier function directly affect host metabolism; (2) substrate availability affecting colonic microbial composition and contact with the gut barrier; and (3) microbial end products affecting host metabolism. The quantity/concentration, duration, and/or frequency (circadian rhythm) of changes in these pathways can alter the gut microbiome, disrupt the gut barrier, alter host immunity, and increase the risk of and progression to overweight and obesity. Host-specific characteristics (e.g., genetic variations) may further affect individual sensitivity and/or resilience to diet- and microbiome-associated perturbations in the colonic environment. In this narrative review, the effects of selected interventions, including fecal microbiota transplantation, dietary calorie restriction, dietary fibers and prebiotics, probiotics and synbiotics, vitamins, minerals, and fatty acids, on the gut microbiome, body weight, and/or adiposity are summarized to help identify mechanisms of action and research opportunities.


Asunto(s)
Microbioma Gastrointestinal , Obesidad , Sobrepeso , Humanos , Microbioma Gastrointestinal/fisiología , Obesidad/microbiología , Obesidad/metabolismo , Sobrepeso/microbiología , Probióticos , Prebióticos/administración & dosificación , Trasplante de Microbiota Fecal , Dieta
13.
Benef Microbes ; 15(6): 589-608, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39242081

RESUMEN

The neuropsychiatric effects of probiotics, prebiotics, and synbiotics have been gaining attention since the rise of microbial-gut-brain axis research. Nevertheless, some of the findings are inconsistent, and few studies have analysed the similarities and differences in the neuropsychiatric effects of the three comprehensively. To reveal the respective neuropsychiatric effects of probiotics, prebiotics, and synbiotics and synthesise the similarities and differences among the three effects, 47 meta-analyses with 12 types of neuropsychiatric results were integrated under an umbrella review. Probiotics, prebiotics, and synbiotics intake might all be associated with improvements in some neuropsychiatric outcomes, including neuropsychological test outcomes (probiotic and prebiotic), hepatic encephalopathy outcomes (probiotic, prebiotic, and synbiotic), instant memory in patients with Alzheimer's disease (probiotic), depressive symptoms (probiotic, prebiotic and synbiotic), mood states and psychiatric distress (probiotic), overall mental health (probiotic), neurological function (probiotic), brain-derived neurotrophic factor (BDNF) concentration (probiotic and synbiotic), and Pittsburgh Sleep Quality Index (PSQI) score (probiotic). All three are similar in that the intake of probiotics, prebiotics, and synbiotics might be associated with improvements in hepatic encephalopathy outcomes and depressive symptoms, both probiotic and synbiotic intake might be associated with elevated BDNF concentrations, and both probiotic and prebiotic intake might be associated with improved neuropsychological test results. The difference between the three is that the neuropsychiatric effects of probiotics might be more widespread and be reflected in the fact that probiotic intake might also be associated with improvements in mood states and psychiatric distress, overall mental health, neurological function, Alzheimer's disease patients' instant memory, and PSQI score. Probiotics might be the best and most promising option for improving neuropsychiatric outcomes. In the future, in addition to requiring more high-quality meta-analyses, further preclinical studies are needed to explore specific relevant mechanisms and determine true causal relationships.


Asunto(s)
Prebióticos , Probióticos , Simbióticos , Probióticos/administración & dosificación , Probióticos/uso terapéutico , Humanos , Simbióticos/administración & dosificación , Prebióticos/administración & dosificación , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/dietoterapia , Encefalopatía Hepática/terapia , Encefalopatía Hepática/dietoterapia , Trastornos Mentales/terapia , Metaanálisis como Asunto , Depresión/terapia , Factor Neurotrófico Derivado del Encéfalo
14.
J Clin Invest ; 134(18)2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39286985

RESUMEN

BACKGROUNDFrailty significantly affects morbidity and mortality rates in the older population (age >65 years). Age-related degenerative diseases are influenced by the intestinal microbiota. However, limited research exists on alterations in the intestinal microbiota in frail older individuals, and the effectiveness of prebiotic intervention for treating frailty remains uncertain.OBJECTIVEWe sought to examine the biological characteristics of the intestinal microbiome in frail older individuals and assess changes in both frailty status and gut microbiota following intervention with a prebiotic blend consisting of inulin and oligofructose.METHODSThe study consisted of 3 components: an observational analysis with a sample size of 1,693, a cross-sectional analysis (n = 300), and a multicenter double-blind, randomized, placebo-controlled trial (n = 200). Body composition, commonly used scales, biochemical markers, intestinal microbiota, and metabolites were examined in 3 groups of older individuals (nonfrail, prefrail, and frail). Subsequently, changes in these indicators were reevaluated after a 3-month intervention using the prebiotic mixture for the prefrail and frail groups.RESULTSThe intervention utilizing a combination of prebiotics significantly improved frailty and renal function among the older population, leading to notable increases in protein levels, body fat percentage, walking speed, and grip strength. Additionally, it stimulated an elevation in gut probiotic count and induced alterations in microbial metabolite expression levels as well as corresponding metabolic pathways.CONCLUSIONSThe findings suggest a potential link between changes in the gut microbiota and frailty in older adults. Prebiotics have the potential to modify the gut microbiota and metabolome, resulting in improved frailty status and prevention of its occurrence.TRIAL REGISTRATIONClinicalTrials.gov NCT03995342.


Asunto(s)
Anciano Frágil , Fragilidad , Microbioma Gastrointestinal , Prebióticos , Humanos , Prebióticos/administración & dosificación , Anciano , Masculino , Método Doble Ciego , Femenino , Anciano de 80 o más Años , Vida Independiente , Oligosacáridos/administración & dosificación , Inulina/administración & dosificación , Estudios Transversales
15.
BMC Res Notes ; 17(1): 249, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39237963

RESUMEN

OBJECTIVES: Apples are one of the most frequently consumed fruits and are effective in preventing lifestyle-related and other diseases. However, few studies have been conducted to evaluate health benefits of processed apple products such as juice. In this study, we analyzed the health benefits of consuming apple juice, focusing on changes in the gut microbiota, which plays an important role in maintaining human health. RESULTS: Rats were fed apple juice ad libitum, and the relative abundances of various gut microbiota in fecal samples were analyzed. In addition, rats treated apple juice were fed with a high-fat diet, and body weight, plasma triglyceride, glucose, and cholesterol levels were measured. The relative abundance of Clostridium cluster XIV did not change with the treatment of apple juice, but the relative abundance of Clostridium cluster IV was significantly decreased. In contrast, the relative abundances of Lactobacillus and Bifidobacterium, which provide benefits to the human body, were significantly increased by 3-fold and 10-fold, respectively, with apple juice consumption. When apple juice-treated rats were fed a high-fat diet, the increase in body weight, liver fat, and blood lipid parameters were all suppressed compared to high-fat alone group. CONCULUSION: This study suggests that the consumption of apple juice changes the gut microbiota, exerts a prebiotic effect, and is effective in improving lifestyle-related diseases.


Asunto(s)
Dieta Alta en Grasa , Jugos de Frutas y Vegetales , Microbioma Gastrointestinal , Malus , Prebióticos , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratas , Masculino , Prebióticos/administración & dosificación , Peso Corporal/efectos de los fármacos , Ratas Sprague-Dawley , Heces/microbiología , Triglicéridos/sangre , Bifidobacterium
16.
J Control Release ; 375: 495-512, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39284524

RESUMEN

The process of microencapsulation and the development of microparticle-based drug formulations have gained increased pharmaceutical interest, particularly for drug delivery and bacterial-encapsulation purposes for probiotic delivery. Existing studies have examined microcomposite (MC) responses to gastrointestinal (GI) conditions with the aim of controlling disintegration, and thus release, across the small and large bowel. However, the delivery of MCs which remain intact, without degrading, could act as bacterial growth scaffolds or materials providing a prebiotic support, conferring potentially beneficial GI health properties. This present study employs prilling as a method to produce a portfolio of MCs using a variety of biopolymers (alginate, chitosan, pectin and gellan gum) with a range of MC diameters and density compositions. Fluorescent probes are co-encapsulated within each MC to enable flow-cytometry directed release profile assessments following exposure to chemical simulated gastric and intestinal digestion conditions. We observe that MC size, gel-strength, density, and biopolymer material all influence response to gastric and intestinal conditions. Gellan gum (GG) MCs demonstrated complete resistance to disintegration throughout GI-simulation in the stomach and small intestine. Considering these MCs could reach the colon intact, we then examined how such MCs, doped with prebiotic growth supporting carboxymethyl cellulose (CMC) polymers, could impact microbial communities using a bioreactor model of the colonic microbiome. Following supplementation with GGCMC MCs, mucosal bacterial diversity (using 16 s rRNA sequencing and Shannon entropy and observed feature diversity metrics) and taxonomic composition changes were observed. Concentrations of short chain fatty acid (SCFA) metabolites were also found to be altered. This is the first study to comprehensivelyexamine how MC physicochemistry can be manipulated to tailor MCs to have the desired GI release performance and subsequently, how GI-resistant MCs could have influential microbial altering properties and be adopted in novel prebiotic strategies.


Asunto(s)
Microbioma Gastrointestinal , Prebióticos , Prebióticos/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Bacterias , Biopolímeros/química , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Humanos , Polisacáridos Bacterianos/química
17.
Brain Behav Immun ; 122: 167-184, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39142421

RESUMEN

Ageing is characterised by a progressive increase in systemic inflammation and especially neuroinflammation. Neuroinflammation is associated with altered brain states that affect behaviour, such as an increased level of anxiety with a concomitant decline in cognitive abilities. Although multiple factors play a role in the development of neuroinflammation, microglia have emerged as a crucial target. Microglia are the only macrophage population in the CNS parenchyma that plays a crucial role in maintaining homeostasis and in the immune response, which depends on the activation and subsequent deactivation of microglia. Therefore, microglial dysfunction has a major impact on neuroinflammation. The gut microbiota has been shown to significantly influence microglia from birth to adulthood in terms of development, proliferation, and function. Diet is a key modulating factor that influences the composition of the gut microbiota, along with prebiotics that support the growth of beneficial gut bacteria. Although the role of diet in neuroinflammation and behaviour has been well established, its relationship with microglia functionality is less explored. This article establishes a link between diet, animal behaviour and the functionality of microglia. The results of this research stem from experiments on mouse behaviour, i.e., memory, anxiety, and studies on microglia functionality, i.e., cytochemistry (phagocytosis, cellular senescence, and ROS assays), gene expression and protein quantification. In addition, shotgun sequencing was performed to identify specific bacterial families that may play a crucial role in the brain function. The results showed negative effects of long-term consumption of a high fat diet on ageing mice, epitomised by increased body weight, glucose intolerance, anxiety, cognitive impairment and microglia dysfunction compared to ageing mice on a control diet. These effects were a consequence of the changes in gut microbiota modulated by the diet. However, by adding the prebiotics fructo- and galacto-oligosaccharides, we were able to mitigate the deleterious effects of a long-term high-fat diet.


Asunto(s)
Envejecimiento , Ansiedad , Dieta Alta en Grasa , Microbioma Gastrointestinal , Memoria , Microglía , Prebióticos , Animales , Microglía/metabolismo , Dieta Alta en Grasa/efectos adversos , Ratones , Ansiedad/metabolismo , Microbioma Gastrointestinal/fisiología , Prebióticos/administración & dosificación , Masculino , Memoria/fisiología , Encéfalo/metabolismo , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias/metabolismo , Inflamación/metabolismo
18.
Med Oncol ; 41(9): 231, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39162936

RESUMEN

Colorectal cancer (CRC), the third most common cancer worldwide, develops mainly due to the accumulation of genetic and epigenetic changes over many years. Substantial evidence suggests that gut microbiota plays a significant role in the initiation, progression, and control of CRC, depending on the balance between beneficial and pathogenic microorganisms. Nonetheless, gut microbiota composition by regulating the host immune response may either promote or inhibit CRC. Thus, modification of gut microbiota potentially impacts clinical outcomes of immunotherapy. Previous studies have indicated that therapeutic strategies such as probiotics, prebiotics, and postbiotics enhance the intestinal immune system and improve the efficacy of immunotherapeutic agents, potentially serving as a complementary strategy in cancer immunotherapy. This review discusses the role of the gut microbiota in the onset and development of CRC in relation to the immune response. Additionally, we focus on the effect of strategies manipulating gut microbiome on the immune response and efficacy of immunotherapy against CRC. We demonstrate that manipulation of gut microbiome can enhance immune response and outcomes of immunotherapy through downregulating Treg cells and other immunosuppressive cells while improving the function of T cells within the tumor; however, further research, especially clinical trials, are needed to evaluate its efficacy in cancer treatment.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Inmunomodulación , Inmunoterapia , Probióticos , Humanos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/terapia , Microbioma Gastrointestinal/inmunología , Probióticos/uso terapéutico , Inmunomodulación/inmunología , Inmunoterapia/métodos , Prebióticos/administración & dosificación
19.
J Clin Psychopharmacol ; 44(5): 457-461, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39146178

RESUMEN

BACKGROUND: Preliminary evidence suggests that people with schizophrenia have decreased relative abundance of butyrate-producing bacteria in the gut microbiota. Butyrate plays a critical role in maintaining the integrity of the gut-blood barrier and has a number of anti-inflammatory effects. This proof-of-concept study was designed to assess whether the addition of the oligofructose-enriched inulin (OEI) prebiotic: Prebiotin could increase the production of butyrate. METHODS: Twenty-seven people who met the criteria for either Diagnostic and Statistical Manual of Mental Disorders, 5th Edition, schizophrenia or schizoaffective disorder were entered into a 10-day, double-blind, placebo-controlled, randomized clinical trial. The study was conducted on an inpatient unit to standardize the participant diet and environment. Participants were randomized to either OEI (4 g, 3 times a day) or a placebo (4 g of maltodextrin, 3 times a day). In order to assess the effect of OEI treatment on butyrate levels, participants underwent pretreatment and posttreatment OEI challenges. The primary outcome measure was relative change in postchallenge plasma butyrate levels after 10 days of OEI treatment. RESULTS: In both the intent-to-treat and completer analyses, OEI treatment was associated with a greater number of participants who met the OEI challenge responder criteria than those treated with placebo. OEI treatment was also associated with an increase in baseline butyrate levels (effect size for the group difference in the change of baseline butyrate levels was 0.58). CONCLUSIONS: We were able to demonstrate that treatment with the prebiotic OEI selectively increased the level of plasma butyrate in people with schizophrenia.Trial registration:ClinicalTrials.gov identifier NCT03617783.


Asunto(s)
Butiratos , Oligosacáridos , Prebióticos , Esquizofrenia , Humanos , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/sangre , Prebióticos/administración & dosificación , Método Doble Ciego , Masculino , Femenino , Adulto , Persona de Mediana Edad , Oligosacáridos/administración & dosificación , Oligosacáridos/farmacología , Inulina/administración & dosificación , Inulina/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Prueba de Estudio Conceptual , Trastornos Psicóticos/tratamiento farmacológico , Trastornos Psicóticos/dietoterapia , Trastornos Psicóticos/sangre , Adulto Joven
20.
Benef Microbes ; 15(6): 609-641, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39147376

RESUMEN

Microbiota changes throughout infancy and can be modified by biotic supplementation, which includes probiotics, prebiotics, synbiotics, and post-biotics. Given the potential influence of the microbiome on infant sleep, this systematic review and meta-analysis aimed to determine the effect of biotic supplementation on sleep behaviours in full-term infants aged 0-12 months. In June 2023, we searched seven databases for randomised controlled trials (RCTs) of biotic supplementation intervention studies using synonymous terms for 'infant' AND 'biotic' AND 'sleep' (PROSPERO registration ID: CRD42022358822). Title/abstracts and the full texts were independently screened. Data on infant sleep and settling behaviour outcomes, reported adverse/side effects, and co-morbid conditions were extracted for analysis. Using the modified Cochrane Collaboration tool, two independent reviewers judged the risk of bias. Meta-analyses were conducted using RevMan5. Our search yielded 453 unique studies and 23 RCTs are included in this review. Probiotic supplementation was the most common biotic supplementation (provided in 53% of studies), while 28% and 19% offered prebiotic and synbiotic supplementation, respectively. Sleep duration was the most common (95%) reported outcome for probiotics. No significant differences were reported in sleep duration during the 1st to 4th week of probiotic supplementation. However, in the 5th week of probiotic supplementation, infants who received placebo slept significantly longer (MD = -35.17 min, 95% CI [-69.72, -0.62]), suggesting a borderline significance that is clinically relevant. There were limited studies and timeframe alignment to analyse prebiotics, synbiotics, post-biotics, and para-probiotics effects on infant sleep duration. The study suggests probiotic supplementation does not increase infant sleep duration within the first 4 postpartum weeks and may contribute to reduced sleep duration in the fifth week. Limited studies were available to assess the effects of biotic supplementation over the first 12 postpartum months. Future research should assess the full range of sleep behaviours, infant feeding type, and microbiome.


Asunto(s)
Suplementos Dietéticos , Prebióticos , Probióticos , Ensayos Clínicos Controlados Aleatorios como Asunto , Sueño , Humanos , Lactante , Probióticos/administración & dosificación , Prebióticos/administración & dosificación , Recién Nacido , Simbióticos/administración & dosificación , Microbioma Gastrointestinal , Femenino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA