Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 425
Filtrar
1.
Nutrients ; 16(17)2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39275271

RESUMEN

Normal and optimal functioning of the gastrointestinal tract is paramount to ensure optimal nutrition through digestion, absorption and motility function. Disruptions in these functions can lead to adverse physiological symptoms, reduced quality of life and increased nutritional risk. When disruption or dysfunction of neuromuscular function occurs, motility disorders can be classified depending on whether coordination or strength/velocity of peristalsis are predominantly impacted. However, due to their nonspecific presenting symptoms and overlap with sensory disruption, they are frequently misdiagnosed as disorders of the gut-brain interaction. Motility disorders are a prevalent issue in the pediatric population, with management varying from medical therapy to psychological therapy, dietary manipulation, surgical intervention or a multimodal approach. This narrative review aims to discuss the dietary management of common pediatric motility disorders including gastroesophageal reflux, esophageal atresia, achalasia, gastroparesis, constipation, and the less common but most severe motility disorder, pediatric intestinal pseudo-obstruction.


Asunto(s)
Enfermedades Gastrointestinales , Motilidad Gastrointestinal , Humanos , Motilidad Gastrointestinal/fisiología , Niño , Enfermedades Gastrointestinales/dietoterapia , Enfermedades Gastrointestinales/terapia , Enfermedades Gastrointestinales/fisiopatología , Preescolar
3.
J Int Soc Sports Nutr ; 21(1): 2388085, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39193818

RESUMEN

BACKGROUND: Fatigue and gastrointestinal (GI) distress are common among athletes with an estimated 30-90% of athletes participating in marathons, triathlons, or similar events experiencing GI complaints. Intense exercise can lead to increased intestinal permeability, potentially allowing members of the gut microbiota to permeate into the bloodstream, resulting in an inflammatory response and cascade of performance-limiting outcomes. Probiotics, through their capacity to regulate the composition of the gut microbiota, may act as an adjunctive therapy by enhancing GI and immune function while mitigating inflammatory responses. This review investigates the effectiveness of probiotic supplementation on fatigue, inflammatory markers, and exercise performance based on randomized controlled trials (RCTs). METHODS: This review follows the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines and PICOS (Population, Intervention, Comparison, Outcome, Study design) framework. A comprehensive search was conducted in Sportdiscus, PubMed, and Scopus databases, and the screening of titles, abstracts, and full articles was performed based on pre-defined eligibility criteria. Of the 3505 records identified, 1884 were screened using titles and abstracts, of which 450 studies were selected for full-text screening. After final screening, 13 studies met the eligibility criteria and were included for review. The studies contained 513 participants, consisting of 351 males and 115 females, however, two studies failed to mention the sex of the participants. Among the participants, 246 were defined as athletes, while the remaining participants were classified as recreationally active (n = 267). All trials were fully described and employed a double- or triple-blind placebo-controlled intervention using either a single probiotic strain or a multi-strain synbiotic (containing both pro- and pre-biotics). RESULTS: This review assesses the effects of daily probiotic supplementation, ranging from 13 to 90 days, on physical performance and physiological markers in various exercise protocols. Ten studies reported improvements in various parameters, such as, enhanced endurance performance, improved anxiety and stress levels, decreased GI symptoms, and reduced upper respiratory tract infections (URTI). Moreover, despite no improvements in maximal oxygen uptake (VO2), several studies demonstrated that probiotic supplementation led to amelioration in lactate, creatine kinase (CK), and ammonia concentrations, suggesting beneficial effects on mitigating exercise-induced muscular stress and damage. CONCLUSION: Probiotic supplementation, specifically at a minimum dosage of 15 billion CFUs daily for a duration of at least 28 days, may contribute to the reduction of perceived or actual fatigue.


Asunto(s)
Rendimiento Atlético , Fatiga , Enfermedades Gastrointestinales , Inflamación , Probióticos , Humanos , Atletas , Rendimiento Atlético/fisiología , Fatiga/inmunología , Fatiga/prevención & control , Enfermedades Gastrointestinales/complicaciones , Enfermedades Gastrointestinales/dietoterapia , Enfermedades Gastrointestinales/inmunología , Enfermedades Gastrointestinales/microbiología , Microbioma Gastrointestinal/inmunología , Inflamación/complicaciones , Inflamación/dietoterapia , Inflamación/inmunología , Inflamación/microbiología , Probióticos/administración & dosificación , Ensayos Clínicos Controlados Aleatorios como Asunto , Fenómenos Fisiológicos en la Nutrición Deportiva/inmunología
4.
Nutrients ; 16(16)2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39203877

RESUMEN

The rise in food intolerances and celiac disease, along with advanced diagnostic techniques, has prompted health professionals to seek effective and economical testing methods. This study evaluates combining genetic tests with routine carbohydrate-absorption breath tests to classify patients with chronic gastrointestinal disorders into therapeutic groups, enhancing dietary management and improving gut health and quality of life. Forty-nine patients with suspected carbohydrate intolerance underwent genetic testing for lactase non-persistence, hereditary fructose intolerance, and celiac disease risk. Simultaneously, breath tests assessed lactose and fructose absorption. The lactase non-persistence genotype appeared in 36.7% of cases, with one hereditary fructose-intolerance case in a heterozygous condition. Celiac disease risk markers (HLA-DQ2/8 haplotypes) were found in 49.0% of the population. Secondary lactose and/or fructose malabsorption was present in 67.3% of patients, with 66.1% of lactase non-persistence individuals showing secondary lactose malabsorption. Fructose malabsorption was prevalent in 45.8% of patients at risk for celiac disease. Two main treatment groups were defined based on genetic results, indicating primary and irreversible gastrointestinal disorder causes, followed by a sub-classification using breath test results. Genetic testing is a valuable tool for designing dietary management plans, avoiding unnecessary diet restrictions, and reducing recovery times.


Asunto(s)
Pruebas Respiratorias , Enfermedad Celíaca , Intolerancia a la Fructosa , Enfermedades Gastrointestinales , Pruebas Genéticas , Lactasa , Intolerancia a la Lactosa , Humanos , Femenino , Masculino , Adulto , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/genética , Persona de Mediana Edad , Pruebas Respiratorias/métodos , Pruebas Genéticas/métodos , Intolerancia a la Fructosa/genética , Intolerancia a la Fructosa/dietoterapia , Intolerancia a la Fructosa/diagnóstico , Enfermedades Gastrointestinales/dietoterapia , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/diagnóstico , Intolerancia a la Lactosa/genética , Intolerancia a la Lactosa/dietoterapia , Intolerancia a la Lactosa/diagnóstico , Enfermedad Crónica , Lactasa/genética , Lactasa/deficiencia , Lactasa/metabolismo , Anciano , Intolerancia Alimentaria/genética , Adulto Joven , Adolescente , Fructosa
5.
Adv Exp Med Biol ; 1449: 157-174, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39060737

RESUMEN

The most frequent functional gastrointestinal disorders (FGID) in children include infantile colic, constipation, functional abdominal pain (FAP), and irritable bowel syndrome (IBS). Unfortunately, treatment options for FGID in children are limited, therefore many dietary interventions have been evaluated, including probiotics. This chapter summarizes currently available evidence and recommendations for probiotic use in the treatment of frequent FGIDs in children. The strongest evidence exists for the use of Limosilactobacillus (L.) reuteri DSM 17938 and Bifidobacterium animalis subsp. lactis BB-12 for the treatment of infantile colic in breastfed infants. Limited but yet encouraging evidence exists for Lacticaseibacillus rhamnosus GG (LGG) for the treatment of IBS and L. reuteri DSM 17938 for FAP.


Asunto(s)
Enfermedades Gastrointestinales , Probióticos , Probióticos/uso terapéutico , Humanos , Enfermedades Gastrointestinales/terapia , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/dietoterapia , Niño , Lactante , Síndrome del Colon Irritable/terapia , Síndrome del Colon Irritable/dietoterapia , Síndrome del Colon Irritable/microbiología , Limosilactobacillus reuteri/fisiología
6.
Dig Dis Sci ; 69(9): 3344-3360, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39001958

RESUMEN

Elemental diets have been employed for the management of various diseases for over 50 years, with several mechanisms mediating their beneficial effects. Yet, they are underutilized due to poor palatability, access, cost, and lack of awareness regarding their clinical efficacy. Therefore, in this review, we aimed to systematically search and review the literature to summarize the formulation variability, mechanisms of action, clinical applications, and tolerability of the elemental diets in gastrointestinal diseases. While large prospective trials are lacking, elemental diets appear to exhibit objective and subjective clinical benefit in several diseases, including eosinophilic esophagitis, eosinophilic gastroenteritis, inflammatory bowel diseases, small intestinal bacterial overgrowth, intestinal methanogen overgrowth, chemoradiotherapy-associated mucositis, and celiac disease. Although some data support the long-term use of elemental diets as an add-on supplement for chronic pancreatitis and Crohn's disease, most of the literature on exclusive elemental diets focuses on inducing remission. Therefore, subsequent treatment strategies for maintaining remission need to be adopted in chronic/relapsing diseases. Several mechanistic pathways were identified to mediate the effects of elemental diets, including food additive and allergen-free content, high passive absorption rate, and anti-inflammatory properties. High rates of intolerance up to 40% are seen in the trials where exclusive elemental diets were administered orally due to poor organoleptic acceptability; however, when tolerated, adverse events were rare. Other limitations of elemental diets are cost, access, and lifestyle/social restrictions. Moreover, judicious use is advised in presence of a concomitant restrictive food intake disorders. Elemental diets offer a potentially highly efficacious dietary intervention with minor side effects. Palatability, cost, access, and social restrictions are common barriers of use. Prospective clinical trials are needed to elucidate the role of elemental formulas in the management of individual diseases.


Asunto(s)
Alimentos Formulados , Humanos , Enfermedades Gastrointestinales/dietoterapia , Enfermedades Gastrointestinales/terapia
9.
An Pediatr (Engl Ed) ; 101(1): 36-45, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38906802

RESUMEN

In this article we present a protocol for the use of the low-FODMAP diet in paediatric patients and review of the current evidence on its efficacy. These short-chain carbohydrates, which can be fermented by the intestinal microbiota, are found in a wide variety of foods, mainly of plant origin. The low-FODMAP diet is a therapeutic tool used for the management of gastrointestinal disorders such as irritable bowel syndrome. The sources we used were PubMed, Web of Science, Google Scholar and institutional websites. Following consumption of FODMAP-rich foods, a series of end products are generated that are not absorbed, giving rise to symptoms. Before starting a low-FODMAP diet, it is important to carry out a diagnostic evaluation including any applicable tests. Treatment is structured in 3 phases: elimination, reintroduction and personalization phase. In the first phase, FODMAP-rich foods are eliminated for 2-3 weeks. In the second phase, lasting 8 weeks, FODMAP-rich foods are gradually reintroduced. The last phase consists in customizing the diet according to individual tolerance. This article details which foods contain FODMAPs and possible substitutes. In addition, specific food diary/intake tracking and educational materials are provided in a series of appendices to facilitate adherence to the diet. Although most studies have been conducted in adults, there is also some evidence on the beneficial effects in the paediatric age group, with a reduction of symptoms, especially in patients with functional gastrointestinal disorders. Nevertheless, more research is required on the subject.


Asunto(s)
Enfermedades Gastrointestinales , Humanos , Niño , Enfermedades Gastrointestinales/dietoterapia , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/terapia , Dieta Baja en Carbohidratos/métodos , Síndrome del Colon Irritable/dietoterapia , Síndrome del Colon Irritable/diagnóstico , Carbohidratos de la Dieta/administración & dosificación , Dieta FODMAP
11.
J Gastroenterol Hepatol ; 39(9): 1728-1736, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38780349

RESUMEN

In recent years, gut microbiota has become a hot topic in the fields of medicine and life sciences. Short-chain fatty acids (SCFAs), the main metabolites of gut microbiota produced by microbial fermentation of dietary fiber, play a vital role in healthy and ill hosts. SCFAs regulate the process of metabolism, immune, and inflammation and have therapeutic effects on gastrointestinal and neurological disorders, as well as antitumor properties. This review summarized the production, distribution, and molecular mechanism of SCFAs, as well as their mechanisms of action in healthy and ill hosts. In addition, we also emphasized the negative effects of SCFAs, aiming to provide the public with a more comprehensive understanding of SCFAs.


Asunto(s)
Dieta , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Microbioma Gastrointestinal/fisiología , Humanos , Ácidos Grasos Volátiles/metabolismo , Fibras de la Dieta/administración & dosificación , Animales , Fermentación , Enfermedades Gastrointestinales/microbiología , Enfermedades Gastrointestinales/dietoterapia
12.
Rev. Hosp. Ital. B. Aires (2004) ; 43(3): 153-159, sept. 2023.
Artículo en Español | LILACS, UNISALUD, BINACIS | ID: biblio-1519056

RESUMEN

El consumo de probióticos, prebióticos y posbióticos, o su combinación, puede contribuir a mantener una microbiota intestinal saludable ya que permite la regulación de su disbiosis en el caso de algunas enfermedades o trastornos, principalmente en los trastornos gastrointestinales funcionales (TGIF). El microbioma intestinal es protagonista esencial en la fisiopatología de los TGIF a través de sus funciones metabólicas y nutricionales, el mantenimiento de la integridad de la mucosa intestinal y la regulación de la respuesta inmunitaria. Las investigaciones realizadas hasta la fecha indican que los probióticos, prebióticos y posbióticos pueden tener efectos inmunomoduladores directos y clínicamente relevantes. Existen pruebas del uso de esta familia de bióticos en individuos sanos para mejorar la salud general y aliviar los síntomas en una serie de enfermedades como los cólicos infantiles. La colonización y establecimiento de la microbiota comienza en el momento del nacimiento; los primeros 2-3 años de vida son fundamentales para el desarrollo de una comunidad microbiana abundante y diversa. Diversos estudios científicos realizados mediante técnicas tradicionales dependientes de cultivo y más recientemente por técnicas moleculares han observado diferencias en las poblaciones bacterianas de bebés sanos y aquellos que sufren TGIF, estos últimos caracterizados por un aumento de especies patógenas y una menor población de bifidobacterias y lactobacilos, en comparación con los primeros. En tal contexto, se considera que la microbiota intestinal como protagonista en el desarrollo de esos trastornos, entre ellos los cólicos infantiles, a través de sus funciones metabólicas, nutricionales, de mantenimiento de la integridad de la mucosa intestinal y regulación de la respuesta inmunitaria. Esto ha abierto la puerta al estudio de la utilización de prebióticos, probióticos y posbióticos en el tratamiento y/o prevención de los TGIF infantiles. El parto vaginal y de término así como la lactancia son fundamentales en la constitución de una microbiota saludable. Como herramientas de apoyo, existen estudios de eficacia que sustentan la administración de esta familia de bióticos, principalmente en los casos en que la lactancia no sea posible o esté limitada. (AU)


The consumption of probiotics, prebiotics, and postbiotics, or a combination of them, can contribute to maintaining a healthy intestinal microbiota as it allows the regulation of its dysbiosis in the case of some diseases or disorders, mainly in functional gastrointestinal disorders (FGIDs). The gut microbiome is an essential player in the pathophysiology of FGIDs through its metabolic and nutritional functions, the maintenance of intestinal mucosal integrity, and the regulation of the immune response. Research results thus far indicate that probiotics, prebiotics, and postbiotics may have direct and clinically relevant immunomodulatory effects. There is evidence regarding the prescription of this family of biotics in healthy individuals to improve overall health and alleviate symptoms in many conditions like infantile colic. The colonization and microbiota establishment begins at birth; the first 2-3 years of life are critical for developing an abundant and diverse microbial community. Several scientific studies performed by traditional culture-dependent techniques and more recently by molecular techniques have observed differences in the bacterial populations of healthy infants and those suffering from FGIDs, the latter characterized by an increase in pathogenic species and a lower population of bifidobacteria and lactobacilli, compared to the former. In this context, the intestinal microbiota plays a leading role in the onset of these disorders, including infantile colic, through its metabolic and nutritional functions, maintenance of the integrity of the intestinal mucosa, and regulation of the immune response. That has opened the door to the study of prebiotics, probiotics, and postbiotics usage in the treatment and or prevention of infantile FGIDs. Vaginal and term delivery and breastfeeding are fundamental in the constitution of a healthy microbiota. As supportive tools, there are efficacy studies that support the administration of this family of biotics, mainly in cases where lactation is not possible or is limited.


Asunto(s)
Humanos , Cólico/microbiología , Probióticos , Prebióticos , Simbióticos , Microbioma Gastrointestinal , Enfermedades Gastrointestinales/microbiología , Lactancia , Cólico/dietoterapia , Cólico/fisiopatología , Cólico/prevención & control , Alimentos Funcionales , Enfermedades Gastrointestinales/dietoterapia , Enfermedades Gastrointestinales/fisiopatología , Enfermedades Gastrointestinales/prevención & control
13.
Food Funct ; 13(3): 1062-1077, 2022 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-35073567

RESUMEN

Food digestion and absorption in infants are closely related to early growth and long-term health. Human milk and infant formula are the main food sources for 0-6 month-old infants. Due to the immature gastrointestinal tract of newborns, mild digestive problems, such as inefficient digestion and impaired absorption of proteins, lipids and lactose, and gut dysbiosis, are often seen in infancy. The differences in composition between infant formula and human milk make mild digestive problems more likely to occur in formula-fed infants. In recent years, several types of infant formulas have been developed to treat or reduce gastrointestinal digestive problems in infants. This review summarizes the gastrointestinal environment of infants and the digestion of human milk and different infant formulas. We particularly focus on the common digestive problems and appropriate nutritional solutions that may occur in healthy term infants during the first six months of life.


Asunto(s)
Sistema Digestivo/fisiopatología , Enfermedades Gastrointestinales/dietoterapia , Fórmulas Infantiles , Fenómenos Fisiológicos Nutricionales del Lactante/fisiología , Leche Humana , Lactancia Materna , Femenino , Enfermedades Gastrointestinales/fisiopatología , Humanos , Lactante , Recién Nacido
14.
Cambios rev. méd ; 20(2): 116-128, 30 Diciembre 2021. ilus, tabs.
Artículo en Español | LILACS | ID: biblio-1368472

RESUMEN

La aparición del coronavirus tipo 2 del Síndrome Respiratorio Agudo grave (SARS-CoV-2) y su afectación como nuevo Coronavirus de 2019 (Covid-19), ha sido sorprendente por su rápido contagio y progresión de la enfermedad. Todos los grupos etarios pueden ser afectados, pero la población vulnerable es aquella que tiene más de 50 años, inmunosupresión, malnutridos o enfermedades degenerativas no transmisibles como hipertensión, diabetes y enfermedades pulmonares. Aquellos pacientes con múltiples morbilidades se encuentran asociados a procesos de malnutrición y son ellos los que podrían llegar a desarrollar complicaciones por la enfermedad y necesitar de cuidados intensivos1. El correcto manejo del estado nutricional busca prevenir el riesgo de complicaciones relacionadas con el déficit calórico proteico y el aumento de la actividad catabólica; la bibliografía detalla cómo la malnutrición genera compromiso al disminuir la musculatura respiratoria, cardíaca, esquelética y coadyuvar a la falla orgánica. El reto para la nutrición en esta pandemia es poder identificar a la población en riesgo y crear un soporte nutricional adecuado para cada paciente2. Las guías de manejo proporcionan recomendaciones útiles y rápidas para el diagnóstico y tratamiento nutrioterapéutico especializado.


The emergence of the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), and its affectation as the new Coronavirus of 2019 (Covid- 19), has been surprising due to its rapid transmission and disease progression. All age groups can be affected, but the vulnerable population is those over 50 years of age, immunosuppression, malnourished or non-communicable degenerative diseases such as hypertension, diabetes and pulmonary diseases. Those patients with multiple morbidities are associated with malnutrition processes and they are the ones who could develop complications due to the disease and require intensive care1. The correct management of nutritional status seeks to prevent the risk of complications related to protein caloric deficit and increased catabolic activity; the literature details how malnutrition generates compromise by decreasing respiratory, cardiac and skeletal muscle and contributing to organ failure. The challenge for nutrition in this pandemic is to identify the population at risk and create adequate nutritional support for each patient2. Management guidelines provide useful and rapid recommendations for diagnosis and specialized nutriotherapeutic treatment.


Asunto(s)
Humanos , Masculino , Femenino , Estado Nutricional , Enfermedades Gastrointestinales/dietoterapia , COVID-19/dietoterapia , Necesidades Nutricionales , Apoyo Nutricional , Enfermedades Gastrointestinales/tratamiento farmacológico , COVID-19/tratamiento farmacológico
16.
JCI Insight ; 6(22)2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34618688

RESUMEN

Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) are carbohydrates thought to contribute to the symptoms of IBS. A diet in high in FODMAPs (HFM) induces gastrointestinal symptoms in patients with irritable bowel syndrome (IBS), and a diet low in FODMAPs (LFM) improves symptoms in up to 60% of patients with IBS. However, the mechanism by which FODMAPs affect IBS symptoms is unclear. We showed that mice fed on a HFM diet have mast cell activation and colonic barrier loss. Using mast cell-deficient mice with and without mast cell reconstitution, we showed that HFM-mediated colonic barrier loss is dependent on TLR4-dependent mast cell activation. In in vitro studies, we demonstrated that IBS fecal supernatant stimulates mast cells significantly more compared with fecal supernatant from healthy controls. This effect of IBS fecal supernatant on mast cell stimulation is ameliorated in the absence of the TLR4 receptor and after a LFM diet. We found that a LFM diet improves colonic barrier function and reduces mast cell activation while decreasing fecal LPS levels. Our findings indicate that a HFM diet causes mast cell activation via LPS, which in turn leads to colonic barrier loss, and a LFM diet reverses these pathophysiologic mucosal changes.


Asunto(s)
Fermentación/fisiología , Enfermedades Gastrointestinales/dietoterapia , Síndrome del Colon Irritable/dietoterapia , Lipopolisacáridos/metabolismo , Mastocitos/metabolismo , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Resultado del Tratamiento
17.
Nutrients ; 13(9)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34579034

RESUMEN

The pivotal role of childhood nutrition has always roused a growing interest from the scientific community. Plant extracts and bioactive dietary components play a significant role in the maintenance of human health and wellness, with the potential to modulate risk factors and manage symptoms for a large number of common childhood disorders such as memory impairment, respiratory illnesses, gastrointestinal disorders, metabolic derangements, and pathologies related to the oral cavity. This review is designed to highlight the health benefits of botanical extracts and bioactive dietary components in children as evidenced by clinical trials, considering their safety with regards to childhood sensibilities. The supplementation of children with the herbal extracts or bioactive components mentioned in this review leads to the conclusion that they are useful for treating various ailments, with no serious adverse events being reported. However, for the limited number of investigations specifically focused on the safety of such products in children, time is needed to expand the literature data covering the safety of childhood supplementation with botanical extract and bioactive food components.


Asunto(s)
Fenómenos Fisiológicos Nutricionales Infantiles , Suplementos Dietéticos , Fitoquímicos/administración & dosificación , Extractos Vegetales/administración & dosificación , Adolescente , Niño , Preescolar , Dieta/métodos , Enfermedades Gastrointestinales/dietoterapia , Humanos , Lactante , Recién Nacido , Fitoterapia/métodos , Ensayos Clínicos Controlados Aleatorios como Asunto , Enfermedades Respiratorias/dietoterapia
18.
Nutrients ; 13(6)2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205445

RESUMEN

In the last decade, the role of nutritional management in pediatric gastrointestinal diseases has gained increasing popularity. Disease-specific diets have been introduced as conventional treatments by international guidelines. Patients tend to more willingly accept food-based therapies than drugs because of their relatively "harmless" nature. Apart from a diet's therapeutic role, nutritional support is crucial in maintaining growth and improving clinical outcomes in pediatric patients. Despite the absence of classical "side effects", however, it should be emphasized that any dietary modification might have negative consequences on children's growth and development. Hence, expert supervision is always advised, in order to support adequate nutritional requirements. Unfortunately, the media provide an inaccurate perception of the role of diet for gastrointestinal diseases, leading to misconceptions by patients or their caregivers that tends to overestimate the beneficial role of diets and underestimate the potential adverse effects. Moreover, not only patients, but also healthcare professionals, have a number of misconceptions about the nutritional benefits of diet modification on gastrointestinal diseases. The aim of this review is to highlight the role of diet in pediatric gastrointestinal diseases, to detect misconceptions and to give a practical guide for physicians on the basis of current scientific evidence.


Asunto(s)
Enfermedades Gastrointestinales/dietoterapia , Terapia Nutricional , Dolor Abdominal , Animales , Bovinos , Niño , Preescolar , Dieta , Enteritis/dietoterapia , Enteritis/fisiopatología , Eosinofilia/dietoterapia , Eosinofilia/fisiopatología , Hipersensibilidad a los Alimentos , Gastritis/dietoterapia , Gastritis/fisiopatología , Enfermedades Gastrointestinales/fisiopatología , Microbioma Gastrointestinal/fisiología , Conocimientos, Actitudes y Práctica en Salud , Humanos , Lactante , Recién Nacido , Enfermedades Inflamatorias del Intestino/dietoterapia , Enfermedades Inflamatorias del Intestino/fisiopatología , Leche/efectos adversos , Leche/inmunología , Necesidades Nutricionales , Guías de Práctica Clínica como Asunto , Probióticos
19.
Vet Clin North Am Equine Pract ; 37(1): 63-87, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33820610

RESUMEN

Many horses are fed differently than their wild ancestors. They often have limited access to pasture and are fed conserved forage and concentrates rich in starch and sugars, in only 2 meals per day. Feeding practices in contrast to natural feeding behavior can lead to gastrointestinal issues. Standard nutritional evaluation is warranted because of its important role in prevention and in treatment and management of diseases. When medical and nutritional treatments are combined, success rates are higher. New techniques to characterize equine microbiota have been used, allowing for microbiota manipulation to prevent and treat intestinal diseases.


Asunto(s)
Alimentación Animal/análisis , Dieta/veterinaria , Enfermedades Gastrointestinales/veterinaria , Enfermedades de los Caballos/dietoterapia , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Enfermedades Gastrointestinales/dietoterapia , Enfermedades de los Caballos/etiología , Caballos
20.
Nutrients ; 13(3)2021 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-33801020

RESUMEN

Decreased serum vitamin D (VD) levels have been associated with gastrointestinal (GI) disorders, including irritable bowel syndrome (IBS). VD can also modulate the intestinal barrier. Given the link between the GI barrier's alterations and diet, attention has aroused the positive effects of the Low FODMAP Diet (LFD) on IBS patients' symptom profile. We evaluated the GI symptoms and the urinary and circulating markers of GI barrier function, the markers of inflammation and intestinal dysbiosis in 36 IBS patients with predominant diarrhea (IBS-D) (5 men and 31 women, 43.1 ± 1.7 years) categorized for their circulating VD levels in low (L-VD) and normal (N-VD) (cutoff = 20 ng/mL). Evaluations were performed before and after 12 weeks of LFD. At the baseline, L-VD patients showed a significantly worse symptom profile and altered small intestinal permeability (s-IP) than N-VD. After LFD, a significant increase in the circulating VD levels in both the subgroups and a significant improvement of s-IP in L-VD patients occurred. Finally, VD levels negatively correlated with the symptom score and fecal zonulin. These data highlight the close relationship between VD and the intestinal barrier and support their involvement in IBS-D pathophysiology. Moreover, the potentially positive role of LFD in the management of IBS-D was confirmed.


Asunto(s)
Diarrea/dietoterapia , Dieta Baja en Carbohidratos/métodos , Enfermedades Gastrointestinales/dietoterapia , Síndrome del Colon Irritable/dietoterapia , Vitamina D/sangre , Adulto , Biomarcadores/sangre , Índice de Masa Corporal , Diarrea/complicaciones , Heces/química , Femenino , Fermentación , Enfermedades Gastrointestinales/complicaciones , Haptoglobinas , Humanos , Síndrome del Colon Irritable/fisiopatología , Masculino , Persona de Mediana Edad , Precursores de Proteínas/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA