Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nutrients ; 15(9)2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37432248

RESUMEN

The human gastrointestinal (GI) tract hosts complex and dynamic populations of microorganisms (gut microbiota) in advantageous symbiosis with the host organism through sophisticated molecular cross-talk. The balance and diversification within microbial communities (eubiosis) are crucial for the immune and metabolic homeostasis of the host, as well as for inhibiting pathogen penetration. In contrast, compositional dysregulation of the microbiota (dysbiosis) is blamed for the determinism of numerous diseases. Although further advances in the so-called 'omics' disciplines are needed, dietary manipulation of the gut microbial ecosystem through biomodulators (prebiotics, probiotics, symbionts, and postbiotics) represents an intriguing target to stabilize and/or restore eubiosis. Recently, new approaches have been developed for the production of infant formulas supplemented with prebiotics (human milk oligosaccharides [HMOs], galacto-oligosaccharides [GOS], fructo-oligosaccharides [FOS]), probiotics, and postbiotics to obtain formulas that are nutritionally and biologically equivalent to human milk (closer to the reference).


Asunto(s)
Microbiota , Prebióticos , Lactante , Humanos , Factores Inmunológicos , Reacciones Cruzadas , Suplementos Dietéticos
2.
Nutrients ; 15(7)2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-37049461

RESUMEN

Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with multifactorial etiology, characterized by impairment in two main functional areas: (1) communication and social interactions, and (2) skills, interests and activities. ASD patients often suffer from gastrointestinal symptoms associated with dysbiotic states and a "leaky gut." A key role in the pathogenesis of ASD has been attributed to the gut microbiota, as it influences central nervous system development and neuropsychological and gastrointestinal homeostasis through the microbiota-gut-brain axis. A state of dysbiosis with a reduction in the Bacteroidetes/Firmicutes ratio and Bacteroidetes level and other imbalances is common in ASD. In recent decades, many authors have tried to study and identify the microbial signature of ASD through in vivo and ex vivo studies. In this regard, the advent of metabolomics has also been of great help. Based on these data, several therapeutic strategies, primarily the use of probiotics, are investigated to improve the symptoms of ASD through the modulation of the microbiota. However, although the results are promising, the heterogeneity of the studies precludes concrete evidence. The aim of this review is to explore the role of intestinal barrier dysfunction, the gut-brain axis and microbiota alterations in ASD and the possible role of probiotic supplementation in these patients.


Asunto(s)
Trastorno del Espectro Autista , Enfermedades Gastrointestinales , Microbioma Gastrointestinal , Enfermedades Intestinales , Microbiota , Probióticos , Humanos , Eje Cerebro-Intestino , Trastorno del Espectro Autista/etiología , Trastorno del Espectro Autista/terapia , Probióticos/uso terapéutico , Disbiosis/terapia
3.
Nutrients ; 15(3)2023 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-36771322

RESUMEN

About 1 in 4 infants comes forward with prolonged crying, agitation, or infant colic (IC) during the first three months of life and is referred for medical evaluation. The pathogenesis remains poorly understood, as do its implications for future health. The aim of this narrative review was to critically examine and discuss the available literature on long-term consequences of excessive crying and/or colic. Most studies display an association between IC and the onset of functional gastrointestinal disorders (FGIDs) years later, probably related to the presence of common etiopathogenetic factors (environmental, dietary, intestinal dysmotility, visceral hypersensitivity). Although allergic disease in first-degree relatives may be a risk factor for IC, the latter does not appear to be a risk factor for subsequent atopic disease in the individual. Overall, there seems to be a relationship between IC and subsequent headaches, of the migraine type. Similarly, behavioral problems in children with a history of IC appear to be associated with higher parental stress scores. However, the current evidence is based on associations, and currently, a causal relationship between excessive crying and IC and long-term consequences remains not documented.


Asunto(s)
Cólico , Hipersensibilidad , Trastornos Migrañosos , Lactante , Niño , Humanos , Cólico/etiología , Factores de Riesgo , Trastornos Migrañosos/etiología , Padres
4.
Nutr Rev ; 81(3): 252-266, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35947766

RESUMEN

CONTEXT: Obesity is a significant risk factor for many pathological conditions. Whether a gluten-free diet (GFD) is a risk factor for overweight or obesity remains controversial. OBJECTIVE: The primary aim of this study was to assess the prevalence of body mass index (BMI) categories at disease presentation and the variation in BMI category from underweight/normal to overweight/obese and vice versa during a GFD. DATA SOURCES: PubMed, Scopus, and Web of Science databases were searched through February 2021 for retrospective, cross-sectional, and prospective studies reporting BMI categories at disease diagnosis and during a GFD. DATA EXTRACTION: Data were extracted by 2 reviewers independently. Disagreements were resolved by consensus; a third reviewer was consulted, if necessary. Risk of bias was assessed with the Cochrane ROBINS-I tool. DATA ANALYSIS: Subgroup analysis based on age (pediatric/adult patients), study design (prospective, cross-sectional, retrospective), and duration of GFD was performed.. Forty-five studies were selected (7959 patients with celiac disease and 20 524 healthy controls). The mean BMI of celiac patients at presentation was significantly lower than that of controls (P < 0.001). During a GFD, the mean BMI increased significantly (mean difference = 1.14 kg/m2 [95%CI, 0.68-1.60 kg/m2]; I2 = 82.8%; P < 0.001), but only 9% of patients (95%CI, 7%-12%; I2 = 80.0%) changed from the underweight/normal BMI category to the overweight/obese category, while 20% (95%CI, 11%-29%; I2 = 85.8%) moved into a lower BMI category. CONCLUSION: Most celiac patients had a normal BMI at presentation, although the mean BMI was significantly lower than that of controls. A GFD does not increase the risk of becoming overweight/obese, especially in children. The quality of several studies was suboptimal, with moderate or high overall risk of bias and heterogeneity.


Asunto(s)
Enfermedad Celíaca , Sobrepeso , Humanos , Niño , Adulto , Sobrepeso/epidemiología , Sobrepeso/complicaciones , Estudios Prospectivos , Delgadez/epidemiología , Delgadez/complicaciones , Estudios Retrospectivos , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/diagnóstico , Dieta Sin Gluten/efectos adversos , Estudios Transversales , Obesidad/epidemiología , Obesidad/etiología , Índice de Masa Corporal
5.
Nutrients ; 14(18)2022 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-36145098

RESUMEN

Adequate complementary feeding practices are important for short- and long-term child health. In industrialized countries, the formulation of several commercial baby foods (CBFs) and an increase in their consumption has been noticed. AIM: To update and analyze the nutritional composition of CBFs available in the Italian market. METHODS: Data collection carried out in two steps (July 2018-January 2019) and updated in May-September 2021. The information on CBFs was taken from the websites of the major CBF producers available in Italy. The collected information were: Suggested initial and final age of consumption; Ingredients; Energy value; Macronutrients (protein, lipids, and carbohydrates); Fiber; Micronutrients (sodium, iron, and calcium); Presence of salt and added sugars, flavorings, and other additives. RESULTS: Time-space for which CBFs are recommended starts too early and ends too late; protein content is adequate and even too high in some food; Amount of fats and their quality must be improved, keeping the intake of saturated fats low; Sugar content is too high in too many CBFs and salt is unnecessarily present in some of them. Finally, the texture of too many products is purée, and its use is recommended for too long, hindering the development of infants' chewing abilities.


Asunto(s)
Calcio , Evaluación Nutricional , Niño , Grasas de la Dieta/análisis , Fibras de la Dieta , Humanos , Lactante , Alimentos Infantiles/análisis , Fenómenos Fisiológicos Nutricionales del Lactante , Hierro , Micronutrientes , Valor Nutritivo , Sodio , Azúcares
7.
Dig Dis Sci ; 67(7): 2771-2791, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34333726

RESUMEN

Corona virus disease-19 (COVID-19) is the latest global pandemic. COVID-19 is mainly transmitted through respiratory droplets and, apart from respiratory symptoms, patients often present with gastrointestinal symptoms and liver involvement. Given the high percentage of COVID-19 patients that present with gastrointestinal symptoms (GIS), in this review, we report a practical up-to-date reference for the physician in their clinical practice with patients affected by chronic gastrointestinal (GI) diseases (inflammatory bowel disease, coeliac disease, chronic liver disease) at the time of COVID-19. First, we summarised data on the origin and pathogenetic mechanism of SARS-CoV-2. Then, we performed a literature search up to December 2020 examining clinical manifestations of GI involvement. Next, we illustrated and summarised the most recent guidelines on how to adhere to GI procedures (endoscopy, liver biopsy, faecal transplantation), maintaining social distance and how to deal with immunosuppressive treatment. Finally, we focussed on some special conditions such as faecal-oral transmission and gut microbiota. The rapid accumulation of information relating to this condition makes it particularly essential to revise the literature to take account of the most recent publications for medical consultation and patient care.


Asunto(s)
COVID-19 , Gastroenterólogos , Enfermedades Gastrointestinales , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/terapia , Humanos , Pandemias , SARS-CoV-2
9.
Nutrients ; 13(11)2021 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-34836010

RESUMEN

The association between eosinophilic esophagitis and celiac disease is still controversial and its prevalence is highly variable. We aimed to investigate the prevalence of esophageal eosinophilia and eosinophilic esophagitis in a large group of children with celiac disease, prospectively followed over 11 years. METHODS: Prospective observational study performed between 2008 and 2019. Celiac disease diagnosis was based on ESPGHAN criteria. At least four esophageal biopsies were sampled in patients who underwent endoscopy. The presence of at least 15 eosinophils/HPF on esophageal biopsies was considered suggestive of esophageal eosinophilia; at the same time, eosinophilic esophagitis was diagnosed according to the International Consensus Diagnostic Criteria for Eosinophilic Esophagitis. RESULTS: A total of 465 children (M 42% mean age 7.1 years (range: 1-16)) were diagnosed with celiac disease. Three hundred and seventy patients underwent endoscopy, and esophageal biopsies were available in 313. The prevalence of esophageal eosinophilia in children with celiac disease was 1.6% (95% CI: 0.54-2.9%). Only one child was diagnosed as eosinophilic esophagitis; we calculated a prevalence of 0.3% (95% CI: 0.2-0.5%). The odds ratio for an association between eosinophilic esophagitis and celiac disease was at least 6.5 times higher (95% CI: 0.89-47.7%; p = 0.06) than in the general population. CONCLUSION: The finding of an increased number of eosinophils (>15/HPF) in celiac patients does not have a clinical implication or warrant intervention, and therefore we do not recommend routine esophageal biopsies unless clinically indicated.


Asunto(s)
Enfermedad Celíaca/complicaciones , Eosinofilia/epidemiología , Esofagitis Eosinofílica/epidemiología , Enfermedades del Esófago/epidemiología , Adolescente , Biopsia , Enfermedad Celíaca/sangre , Enfermedad Celíaca/patología , Niño , Preescolar , Eosinofilia/etiología , Esofagitis Eosinofílica/etiología , Eosinófilos/patología , Enfermedades del Esófago/etiología , Esófago/patología , Femenino , Humanos , Lactante , Masculino , Oportunidad Relativa , Prevalencia , Estudios Prospectivos
10.
Medicina (Kaunas) ; 57(7)2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-34357014

RESUMEN

Helicobacter pylori (HP) is a Gram-negative bacterium which finds its suitable habitat in the stomach. The infection affects about half of the global population with high variability in prevalence among regions and for age. HP is the main causative agent of chronic active gastritis, peptic and duodenal ulcers, and may be the primary cause of gastric cancer or MALT lymphoma. Due to the high rate of failure of eradication therapy in various countries and the increase in antibiotic resistance reported in the literature, there is an ever wider need to seek alternative therapeutic treatments. Probiotics seem to be a promising solution. In particular, the Limosilactobacillus reuteri (L. reuteri) species is a Gram-positive bacterium and is commonly found in the microbiota of mammals. L. reuteri is able to survive the gastric acid environment and bile and to colonize the gastric mucosa. This species is able to inhibit the growth of several pathogenic bacteria through different mechanisms, keeping the homeostasis of the microbiota. In particular, it is able to secrete reuterin and reutericycline, substances that exhibit antimicrobial properties, among other molecules. Through the secretion of these and the formation of the biofilm, it has been found to strongly inhibit the growth of HP and, at higher concentrations, to kill it. Moreover, it reduces the expression of HP virulence factors. In clinical trials, L. reuteri has been shown to decrease HP load when used as a single treatment, but has not achieved statistical significance in curing infected patients. As an adjuvant of standard regimens with antibiotics and pump inhibitors, L. reuteri can be used not only to improve cure rates, but especially to decrease gastrointestinal symptoms, which are a common cause of lack of compliance and interruption of therapy, leading to new antibiotic resistance.


Asunto(s)
Gastritis , Infecciones por Helicobacter , Helicobacter pylori , Limosilactobacillus reuteri , Probióticos , Animales , Antibacterianos/uso terapéutico , Gastritis/tratamiento farmacológico , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/epidemiología , Humanos , Probióticos/uso terapéutico
11.
Nutrients ; 13(3)2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33804451

RESUMEN

Background: non-autoimmune thyroid disorder is a common finding in celiac patients, more frequent than in the general population. An impairment of iodine absorption has been hypothesized, but it has never been investigated so far. We aimed to evaluate the iodine absorption in children and adolescents with newly diagnosed celiac disease. Methods: 36 consecutive celiac patients (age 7.4 years, range 2.4-14.5 years) before starting a gluten-free diet (GFD) were enrolled. We assayed the urinary iodine concentration (UIC) in a 24-h urine sample, at baseline (T0) after 3 (T1) and 12 months (T2) of GFD. Results: UIC at T0 was 64 µg/L (IQR 45-93.25 µg/L) with an iodine deficiency rate of 77.8%. UIC was not different according to histological damage, clinical presentation (typical vs atypical); we found no correlation with the thyroid function tests and auxological parameters. UIC was not statistically different at T1 (76 µg/L) and T2 (89 µg/L) vs T0. UIC at T2 was similar between patients with positive and negative anti-transglutaminase antibodies at T2. No patients presented overt hypothyroidism during the study. Conclusions: We found that iodine absorption in celiac children is impaired compared to the general population; it increases slightly, but not significantly, during the GFD. We should regularly reinforce the need for a proper iodine intake in celiac disease patients to reduce iodine deficiency risk.


Asunto(s)
Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/fisiopatología , Dieta Sin Gluten , Absorción Gastrointestinal , Yodo/deficiencia , Adolescente , Enfermedad Celíaca/orina , Niño , Preescolar , Femenino , Humanos , Yodo/orina , Estudios Longitudinales , Masculino , Estado Nutricional , Proyectos Piloto , Pruebas de Función de la Tiroides , Glándula Tiroides/fisiopatología , Resultado del Tratamiento
12.
Front Immunol ; 12: 578386, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33717063

RESUMEN

Hosting millions of microorganisms, the digestive tract is the primary and most important part of bacterial colonization. On one side, in cases of opportunistic invasion, the abundant bacterial population inside intestinal tissues may face potential health problems such as inflammation and infections. Therefore, the immune system has evolved to sustain the host-microbiota symbiotic relationship. On the other hand, to maintain host immune homeostasis, the intestinal microflora often exerts an immunoregulatory function that cannot be ignored. A field of great interest is the association of either microbiota or probiotics with the immune system concerning clinical uses. This microbial community regulates some of the host's metabolic and physiological functions and drives early-life immune system maturation, contributing to their homeostasis throughout life. Changes in gut microbiota can occur through modification in function, composition (dysbiosis), or microbiota-host interplays. Studies on animals and humans show that probiotics can have a pivotal effect on the modulation of immune and inflammatory mechanisms; however, the precise mechanisms have not yet been well defined. Diet, age, BMI (body mass index), medications, and stress may confound the benefits of probiotic intake. In addition to host gut functions (permeability and physiology), all these agents have profound implications for the gut microbiome composition. The use of probiotics could improve the gut microbial population, increase mucus-secretion, and prevent the destruction of tight junction proteins by decreasing the number of lipopolysaccharides (LPSs). When LPS binds endothelial cells to toll-like receptors (TLR 2, 4), dendritic cells and macrophage cells are activated, and inflammatory markers are increased. Furthermore, a decrease in gut dysbiosis and intestinal leakage after probiotic therapy may minimize the development of inflammatory biomarkers and blunt unnecessary activation of the immune system. In turn, probiotics improve the differentiation of T-cells against Th2 and development of Th2 cytokines such as IL-4 and IL-10. The present narrative review explores the interactions between gut microflora/probiotics and the immune system starting from the general perspective of a biological plausibility to get to the in vitro and in vivo demonstrations of a probiotic-based approach up to the possible uses for novel therapeutic strategies.


Asunto(s)
Antiinflamatorios/farmacología , Gastroenteritis/etiología , Inmunomodulación/efectos de los fármacos , Probióticos/administración & dosificación , Animales , Antiinflamatorios/uso terapéutico , Dieta , Susceptibilidad a Enfermedades , Disbiosis , Gastroenteritis/tratamiento farmacológico , Gastroenteritis/metabolismo , Gastroenteritis/patología , Microbioma Gastrointestinal/inmunología , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Estilo de Vida
13.
Turk Arch Pediatr ; 56(5): 479-484, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35110118

RESUMEN

BACKGROUND: During the pandemic of SARS-Cov-2, among other clinical and public health issues, a major concern raised by SARS-CoV-2 is the possibility of transmission of the infection from mother to child in the perinatal period. This has placed a question mark on the safety of breastfeeding, with ambiguity on the joint management of SARS-CoV-2 positive or suspected mothers and their children. It was aimed to evaluate breastfeeding rates for newborns of asymptomatic SARS-CoV-2 positive mothers who were temporarily separated from their babies at birth, compared to those who were not separated. RESULTS: Babies who were not isolated from their mothers at delivery were significantly more likely to be breastfed and were at no higher risk of infection with SARS-CoV-2. CONCLUSION: Following the World Health Organization (WHO) recommendations and strict hand and mask hygiene measures, breastfeeding practices can be established and maintained through rooming-in, thus promoting the mother-child bond without compromising the safety of the newborn.

14.
Front Immunol ; 11: 567801, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33224137

RESUMEN

Gluten is a mixture of proteins highly resistant to hydrolysis, resulting in the emergence of toxic peptides responsible for gluten-related disorders. Currently, a gluten-free diet (GFD) is the unique proven therapy for celiac disease (CD). Several research groups and pharmaceutical companies are developing new nondietetic therapeutic strategies for CD. Probiotics are viable microorganisms thought to have a healthy effect on the host. The proteolytic mechanism of lactic acid bacteria comprises an extracellular serine protease, di- and oligopeptide-specific transport systems, and several intracellular peptidases that might affect gluten degradation. Therefore, probiotic supplementation is an attractive therapy because of its possible anti-inflammatory and immunomodulatory properties. Several studies have been performed to assess the effectiveness of various specific probiotic strains, showing positive effects on immune-modulation (inhibition of pro-inflammatory cytokine TNF-α) restoring gut microbiota and decrease of immunogenic peptides. The present review aims to summarize the current knowledge on the ability of probiotic strain (single or mixtures) to digest gliadin peptides in vitro and to modulate the inflammatory response in the gut.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Glútenes/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Animales , Enfermedad Celíaca/etiología , Enfermedad Celíaca/prevención & control , Dieta Sin Gluten , Glútenes/efectos adversos , Glútenes/inmunología , Humanos , Hidrólisis , Mucosa Intestinal/microbiología , Probióticos/administración & dosificación
15.
Adv Exp Med Biol ; 1125: 49-56, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30656551

RESUMEN

Colic is a common and distressing functional gastrointestinal disorder during infancy. It is a behavioral phenomenon in infants aged 1-4 months involving prolonged inconsolable crying and agitated status with multifactorial etiology. Colic can be considered as a benign, self-limited process because the baby normally grows and feeds even with transient irritable mood. Nevertheless, infantile colic is a common difficulty causing anxiety during parenthood and a recurrent reason for them to seek medical help, especially if it is the first child. The causes of colic can be classified as non-gastrointestinal or gastrointestinal. The former includes altered feeding techniques, modified child-parent relationship, immaturity of central nervous system, behavioral etiology, and maternal smoking or nicotine replacement therapy. Instead, the latter involves inadequate production of lactase enzyme, cow's milk protein intolerance, alteration of intestinal microbiota, gastrointestinal immaturity, or inflammation which causes intestinal hyperperistalsis due to increase in serotonin secretion and motilin receptor expression.Probiotics may play a crucial part in the manipulation of the microbiota. Probiotic administration is likely to maintain intestinal homeostasis through the modulation of permeability and peristalsis, influencing the gut-brain axis and inhibiting hypersensitivity. This is a decisive field in the development of preventive and therapeutic strategies for infantile colic. However, further studies are needed for each specific formulation in order to better characterize pharmacodynamic and pharmacokinetic properties and to evaluate their application as a possible preventive strategy if administered early during infancy against the later development of pain-related FGIDs.


Asunto(s)
Cólico/prevención & control , Cólico/terapia , Microbioma Gastrointestinal , Probióticos/uso terapéutico , Cólico/etiología , Intolerancia Alimentaria/fisiopatología , Humanos , Lactante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...