Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 625(7993): 157-165, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38093016

RESUMEN

Evidence is accumulating that perturbed postnatal development of the gut microbiome contributes to childhood malnutrition1-4. Here we analyse biospecimens from a randomized, controlled trial of a microbiome-directed complementary food (MDCF-2) that produced superior rates of weight gain compared with a calorically more dense conventional ready-to-use supplementary food in 12-18-month-old Bangladeshi children with moderate acute malnutrition4. We reconstructed 1,000 bacterial genomes (metagenome-assembled genomes (MAGs)) from the faecal microbiomes of trial participants, identified 75 MAGs of which the abundances were positively associated with ponderal growth (change in weight-for-length Z score (WLZ)), characterized changes in MAG gene expression as a function of treatment type and WLZ response, and quantified carbohydrate structures in MDCF-2 and faeces. The results reveal that two Prevotella copri MAGs that are positively associated with WLZ are the principal contributors to MDCF-2-induced expression of metabolic pathways involved in utilizing the component glycans of MDCF-2. The predicted specificities of carbohydrate-active enzymes expressed by their polysaccharide-utilization loci are correlated with (1) the in vitro growth of Bangladeshi P. copri strains, possessing varying degrees of polysaccharide-utilization loci and genomic conservation with these MAGs, in defined medium containing different purified glycans representative of those in MDCF-2, and (2) the levels of faecal carbohydrate structures in the trial participants. These associations suggest that identifying bioactive glycan structures in MDCFs metabolized by growth-associated bacterial taxa will help to guide recommendations about their use in children with acute malnutrition and enable the development of additional formulations.


Asunto(s)
Alimentos , Microbioma Gastrointestinal , Desnutrición , Polisacáridos , Humanos , Lactante , Bacterias/genética , Bangladesh , Peso Corporal/genética , Heces/microbiología , Microbioma Gastrointestinal/fisiología , Genoma Bacteriano/genética , Desnutrición/microbiología , Metagenoma/genética , Polisacáridos/metabolismo , Aumento de Peso
2.
N Engl J Med ; 384(16): 1517-1528, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33826814

RESUMEN

BACKGROUND: More than 30 million children worldwide have moderate acute malnutrition. Current treatments have limited effectiveness, and much remains unknown about the pathogenesis of this condition. Children with moderate acute malnutrition have perturbed development of their gut microbiota. METHODS: In this study, we provided a microbiota-directed complementary food prototype (MDCF-2) or a ready-to-use supplementary food (RUSF) to 123 slum-dwelling Bangladeshi children with moderate acute malnutrition between the ages of 12 months and 18 months. The supplementation was given twice daily for 3 months, followed by 1 month of monitoring. We obtained weight-for-length, weight-for-age, and length-for-age z scores and mid-upper-arm circumference values at baseline and every 2 weeks during the intervention period and at 4 months. We compared the rate of change of these related phenotypes between baseline and 3 months and between baseline and 4 months. We also measured levels of 4977 proteins in plasma and 209 bacterial taxa in fecal samples. RESULTS: A total of 118 children (59 in each study group) completed the intervention. The rates of change in the weight-for-length and weight-for-age z scores are consistent with a benefit of MDCF-2 on growth over the course of the study, including the 1-month follow-up. Receipt of MDCF-2 was linked to the magnitude of change in levels of 70 plasma proteins and of 21 associated bacterial taxa that were positively correlated with the weight-for-length z score (P<0.001 for comparisons of both protein and bacterial taxa). These proteins included mediators of bone growth and neurodevelopment. CONCLUSIONS: These findings provide support for MDCF-2 as a dietary supplement for young children with moderate acute malnutrition and provide insight into mechanisms by which this targeted manipulation of microbiota components may be linked to growth. (Supported by the Bill and Melinda Gates Foundation and the National Institutes of Health; ClinicalTrials.gov number, NCT04015999.).


Asunto(s)
Suplementos Dietéticos , Alimentos Formulados , Microbioma Gastrointestinal , Fenómenos Fisiológicos Nutricionales del Lactante , Desnutrición/dietoterapia , Antropometría , Bangladesh , Proteínas Sanguíneas/análisis , Peso Corporal , Heces/microbiología , Femenino , Crecimiento , Humanos , Lactante , Masculino , Desnutrición/microbiología , Proteoma , Aumento de Peso
3.
Exp Physiol ; 109(5): 662-671, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38156734

RESUMEN

Childhood stunting and wasting, or decreased linear and ponderal growth associated with undernutrition, continue to be a major global public health challenge. Although many of the current therapeutic and dietary interventions have significantly reduced childhood mortality caused by undernutrition, there remain great inefficacies in improving childhood stunting. Longitudinal bone growth in children is governed by different genetic, nutritional and other environmental factors acting systemically on the endocrine system and locally at the growth plate. Recent studies have shown that this intricate interplay between nutritional and hormonal regulation of the growth plate could involve the gut microbiota, highlighting the importance of a holistic approach in tackling childhood undernutrition. In this review, I focus on the mechanistic insights provided by these recent advances in gut microbiota research and discuss ongoing development of microbiota-based therapeutics in humans, which could be the missing link in solving undernutrition and childhood stunting.


Asunto(s)
Desarrollo Óseo , Microbioma Gastrointestinal , Trastornos del Crecimiento , Humanos , Microbioma Gastrointestinal/fisiología , Desarrollo Óseo/fisiología , Niño , Trastornos del Crecimiento/microbiología , Trastornos del Crecimiento/fisiopatología , Animales , Desnutrición/microbiología , Desnutrición/fisiopatología , Desarrollo Infantil/fisiología
4.
Proc Natl Acad Sci U S A ; 116(24): 11988-11996, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31138692

RESUMEN

Undernutrition in children is a pressing global health problem, manifested in part by impaired linear growth (stunting). Current nutritional interventions have been largely ineffective in overcoming stunting, emphasizing the need to obtain better understanding of its underlying causes. Treating Bangladeshi children with severe acute malnutrition with therapeutic foods reduced plasma levels of a biomarker of osteoclastic activity without affecting biomarkers of osteoblastic activity or improving their severe stunting. To characterize interactions among the gut microbiota, human milk oligosaccharides (HMOs), and osteoclast and osteoblast biology, young germ-free mice were colonized with cultured bacterial strains from a 6-mo-old stunted infant and fed a diet mimicking that consumed by the donor population. Adding purified bovine sialylated milk oligosaccharides (S-BMO) with structures similar to those in human milk to this diet increased femoral trabecular bone volume and cortical thickness, reduced osteoclasts and their bone marrow progenitors, and altered regulators of osteoclastogenesis and mediators of Th2 responses. Comparisons of germ-free and colonized mice revealed S-BMO-dependent and microbiota-dependent increases in cecal levels of succinate, increased numbers of small intestinal tuft cells, and evidence for activation of a succinate-induced tuft cell signaling pathway linked to Th2 immune responses. A prominent fucosylated HMO, 2'-fucosyllactose, failed to elicit these changes in bone biology, highlighting the structural specificity of the S-BMO effects. These results underscore the need to further characterize the balance between, and determinants of, osteoclastic and osteoblastic activity in stunted infants/children, and suggest that certain milk oligosaccharides may have therapeutic utility in this setting.


Asunto(s)
Huesos/efectos de los fármacos , Vida Libre de Gérmenes/efectos de los fármacos , Desnutrición/tratamiento farmacológico , Leche Humana/metabolismo , Oligosacáridos/administración & dosificación , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Animales , Bacterias/efectos de los fármacos , Bovinos , Dieta , Modelos Animales de Enfermedad , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Lactante , Intestino Delgado/microbiología , Masculino , Desnutrición/microbiología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
5.
Lancet ; 395(10217): 75-88, 2020 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-31852605

RESUMEN

Malnutrition has historically been researched and addressed within two distinct silos, focusing either on undernutrition, food insecurity, and micronutrient deficiencies, or on overweight, obesity, and dietary excess. However, through rapid global nutrition transition, an increasing proportion of individuals are exposed to different forms of malnutrition during the life course and have the double burden of malnutrition (DBM) directly. Long-lasting effects of malnutrition in early life can be attributed to interconnected biological pathways, involving imbalance of the gut microbiome, inflammation, metabolic dysregulation, and impaired insulin signalling. Life-course exposure to early undernutrition followed by later overweight increases the risk of non-communicable disease, by imposing a high metabolic load on a depleted capacity for homoeostasis, and in women increases the risk of childbirth complications. These life-course trajectories are shaped both by societal driving factors-ie, rapidly changing diets, norms of eating, and physical activity patterns-and by broader ecological factors such as pathogen burden and extrinsic mortality risk. Mitigation of the DBM will require major societal shifts regarding nutrition and public health, to implement comprehensive change that is sustained over decades, and scaled up into the entire global food system.


Asunto(s)
Desnutrición/metabolismo , Obesidad/metabolismo , Sobrepeso/metabolismo , África del Sur del Sahara/epidemiología , Edad de Inicio , Ejercicio Físico , Femenino , Microbioma Gastrointestinal , Humanos , Indonesia/epidemiología , Masculino , Desnutrición/epidemiología , Desnutrición/microbiología , Redes y Vías Metabólicas , Estado Nutricional , Obesidad/epidemiología , Obesidad/microbiología , Sobrepeso/epidemiología , Sobrepeso/microbiología , Prevalencia
6.
Nature ; 527(7578): S155-60, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26580321

RESUMEN

We define neurodevelopment as the dynamic inter-relationship between genetic, brain, cognitive, emotional and behavioural processes across the developmental lifespan. Significant and persistent disruption to this dynamic process through environmental and genetic risk can lead to neurodevelopmental disorders and disability. Research designed to ameliorate neurodevelopmental disorders in low- and middle-income countries, as well as globally, will benefit enormously from the ongoing advances in understanding their genetic and epigenetic causes, as modified by environment and culture. We provide examples of advances in the prevention and treatment of, and the rehabilitation of those with, neurodevelopment disorders in low- and middle-income countries, along with opportunities for further strategic research initiatives. Our examples are not the only possibilities for strategic research, but they illustrate problems that, when solved, could have a considerable impact in low-resource settings. In each instance, research in low- and middle-income countries led to innovations in identification, surveillance and treatment of a neurodevelopmental disorder. These innovations have also been integrated with genotypic mapping of neurodevelopmental disorders, forming important preventative and rehabilitative interventions with the potential for high impact. These advances will ultimately allow us to understand how epigenetic influences shape neurodevelopmental risk and resilience over time and across populations. Clearly, the most strategic areas of research opportunity involve cross-disciplinary integration at the intersection between the environment, brain or behaviour neurodevelopment, and genetic and epigenetic science. At these junctions a robust integrative cross-disciplinary scientific approach is catalysing the creation of technologies and interventions for old problems. Such approaches will enable us to achieve and sustain the United Nations moral and legal mandate for child health and full development as a basic global human right.


Asunto(s)
Investigación Biomédica , Trastornos del Neurodesarrollo , Niño , Epistasis Genética , Interacción Gen-Ambiente , Infecciones por VIH/epidemiología , Infecciones por VIH/psicología , Humanos , Hidrocefalia/psicología , Malaria/epidemiología , Malaria/psicología , Desnutrición/microbiología , Desnutrición/psicología , Trastornos del Neurodesarrollo/epidemiología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/prevención & control , Trastornos del Neurodesarrollo/rehabilitación , Neurotoxinas/efectos adversos , Síndrome del Cabeceo/psicología , Trauma Psicológico/psicología
7.
Int J Food Sci Nutr ; 72(8): 1071-1082, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33840335

RESUMEN

The number of elderlies is increasing but prevalence of malnutrition has been reported. The aim of the study was to determine the significance of short-term nutritional deficiencies in mice. Immune status was assessed through flow cytometry of leucocytes in Peyer's patches (PP) and mesenteric lymph nodes (MLN), and intestinal microbiota was evaluated by terminal restriction fragment length polymorphism (T-RFLP). C57BL/6NCrl mice fed standard diet (StD) or experimental diet high in fat, and low in carbohydrates, protein, fibre, vitamins, and minerals (ExpD) for 2 or 4 weeks. ExpD-animals gained less weight, increased liver lipids, and developed splenomegaly. Diet affected regulatory T-cells, gut homing receptors and TLR2 and TLR4 in PP and MLN and the microbiota was influenced. Partial least squares models on flow cytometry- and T-RFLP data demonstrated correlations between microbial communities and immune phenotyping. Our model shows similarities to malnourished elderly and interactions between intestinal bacteria and the immune system.


Asunto(s)
Dieta , Microbioma Gastrointestinal , Inmunidad , Desnutrición , Animales , Inmunofenotipificación , Desnutrición/inmunología , Desnutrición/microbiología , Ratones , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados
8.
Appl Microbiol Biotechnol ; 104(13): 5973-5984, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32415319

RESUMEN

The objective of this study was to evaluate the effect of undernutrition on colonic microbiota and fermentation in pregnant ewes. Sixteen ewes bearing multiple fetuses for 115 days in the control (CON) and severe feed restriction (SFR) groups were fed 100% and 30% level of ad libitum feed intake, respectively. After 15-day treatment, all ewes were sacrificed to collect colonic digesta samples to extract DNA for 16S rRNA sequencing and to detect fermentation parameters. Our data showed that SFR increased (P < 0.05) the levels of colonic propionate, isobutyrate, butyrate, isovalerate, and valerate, and slightly decreased (P < 0.1) colonic pH. The mole proportions of isobutyrate, butyrate, and isovalerate were increased (P < 0.05) upon SFR while that of acetate was decreased (P < 0.05). Hematoxylin-eosin staining sections exhibited the disorderly, irregular, and loose arrangement and part sloughing of colonic epithelial cells. Furthermore, SFR decreased (P < 0.05) the diversity of colonic microbiota and changed the microbial communities. At the genus level, SFR increased (P < 0.05) the abundance of unclassified Peptococcaceae and decreased (P < 0.05) the abundances of Ruminococcus, unclassified Ruminococcaceae, and unclassified VadinBB60. Additionally, the abundances of Ruminococcus and unclassified Ruminococcaceae were positively correlated (P < 0.05) with the acetate proportion while the abundance of unclassified Peptococcaceae was negatively correlated (P < 0.05) with the percentages of isobutyrate, butyrate, and isovalerate. In summary, SFR diminished the diversity of bacteria, affected the composition of bacterial communities, and finally changed the colonic fermentation pattern and epithelial histomorphology in pregnant ewes. KEY POINTS: • Undernutrition changed colonic bacterial diversity and composition in pregnant ewes. • Microbial alteration affected colonic fermentation pattern and parameters. • Alteration of colonic microbiota and fermentation damaged epithelium histomorphology. Graphical abstract.


Asunto(s)
Colon/microbiología , Microbioma Gastrointestinal/fisiología , Desnutrición/microbiología , Complicaciones del Embarazo/microbiología , Alimentación Animal/efectos adversos , Alimentación Animal/análisis , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Colon/metabolismo , Colon/patología , Ácidos Grasos Volátiles/análisis , Ácidos Grasos Volátiles/metabolismo , Femenino , Fermentación , Concentración de Iones de Hidrógeno , Mucosa Intestinal/patología , Desnutrición/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo , Ovinos
9.
PLoS Pathog ; 13(7): e1006471, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28750066

RESUMEN

Diverse enteropathogen exposures associate with childhood malnutrition. To elucidate mechanistic pathways whereby enteric microbes interact during malnutrition, we used protein deficiency in mice to develop a new model of co-enteropathogen enteropathy. Focusing on common enteropathogens in malnourished children, Giardia lamblia and enteroaggregative Escherichia coli (EAEC), we provide new insights into intersecting pathogen-specific mechanisms that enhance malnutrition. We show for the first time that during protein malnutrition, the intestinal microbiota permits persistent Giardia colonization and simultaneously contributes to growth impairment. Despite signals of intestinal injury, such as IL1α, Giardia-infected mice lack pro-inflammatory intestinal responses, similar to endemic pediatric Giardia infections. Rather, Giardia perturbs microbial host co-metabolites of proteolysis during growth impairment, whereas host nicotinamide utilization adaptations that correspond with growth recovery increase. EAEC promotes intestinal inflammation and markers of myeloid cell activation. During co-infection, intestinal inflammatory signaling and cellular recruitment responses to EAEC are preserved together with a Giardia-mediated diminishment in myeloid cell activation. Conversely, EAEC extinguishes markers of host energy expenditure regulatory responses to Giardia, as host metabolic adaptations appear exhausted. Integrating immunologic and metabolic profiles during co-pathogen infection and malnutrition, we develop a working mechanistic model of how cumulative diet-induced and pathogen-triggered microbial perturbations result in an increasingly wasted host.


Asunto(s)
Coinfección/microbiología , Coinfección/parasitología , Infecciones por Escherichia coli/microbiología , Escherichia coli/fisiología , Giardia lamblia/fisiología , Giardiasis/parasitología , Desnutrición/microbiología , Desnutrición/parasitología , Animales , Niño , Coinfección/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/inmunología , Giardiasis/inmunología , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/parasitología , Masculino , Desnutrición/inmunología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/inmunología
10.
Int J Med Microbiol ; 309(2): 151-158, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30733116

RESUMEN

Shigella/Enteroinvasive Escherichia coli (EIEC) pathotype is a major enteropathogen associated with diarrhea and malnutrition in children from developing countries. This study aimed to correlate Shigella/EIEC virulence-related genes (VRGs) with clinical symptoms, nutritional status and coenteropathogens in children from the Brazilian semiarid region. We designed a case-control study of community diarrhea in six cities of the Brazil semiarid region with 1200 children aging 2-36 months. Standardized questionnaire was applied for collecting sociodemographic, nutritional status and clinical information of the children. DNA samples were extracted from stools and diagnosed for Shigella/EIEC using PCR-based approaches. Positive samples were tested for 28 VRGs using four multiplex PCRs. Intestinal inflammation was determined by measuring fecal myeloperoxidase (MPO). Shigella/EIEC pathotype was detected in 5% of the children and was significantly associated with diarrhea. The genes sen (encoding Shigella enterotoxin 2), ipgB2, ipgB1 (both encoding type 3 secretion system-T3SS effectors that modulate actin filament), and ospF (encoding a T3SS effector involved in suppression of host responses) were further associated with diarrhea in Shigella/EIEC positive children. Among children presenting diarrhea, virA gene (encoding a T3SS effector that promotes microtubule destabilization) was associated with fever, while virB (encoding a major transcriptional activator) was associated with low height-for-age z-score. In addition, these VRGs were associated with increased fecal MPO, and coinfection with Salmonella spp. was associated with increased abdominal pain. These data reinforce the impact of Shigella/EIEC on diarrhea in children from Brazilian semiarid region and highlighted the contributions of specific virulence genes for its pathobiology.


Asunto(s)
Diarrea/patología , Disentería Bacilar/patología , Infecciones por Escherichia coli/patología , Escherichia coli/aislamiento & purificación , Desnutrición/patología , Shigella/aislamiento & purificación , Factores de Virulencia/genética , Brasil/epidemiología , Estudios de Casos y Controles , Preescolar , Ciudades/epidemiología , Estudios Transversales , Clima Desértico , Diarrea/epidemiología , Diarrea/microbiología , Disentería Bacilar/epidemiología , Disentería Bacilar/microbiología , Escherichia coli/genética , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/microbiología , Femenino , Genes Bacterianos , Humanos , Lactante , Masculino , Desnutrición/epidemiología , Desnutrición/microbiología , Reacción en Cadena de la Polimerasa , Shigella/genética , Shigella/patogenicidad , Encuestas y Cuestionarios , Resultado del Tratamiento
11.
BMC Microbiol ; 19(1): 277, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31823731

RESUMEN

BACKGROUND: Childhood malnutrition is a global health challenge associated with multiple adverse consequences, including delayed maturation of the gut microbiota (GM) which might induce long-term immune dysfunction and stunting. To understand GM dynamics during malnutrition and subsequent re-feeding, we used a piglet model with a malnutrition-induced phenotype similar to humans. Piglets were weaned at the age of 4 weeks, fed a nutritionally optimal diet for 1 week post-weaning before being fed a pure maize diet for 7 weeks to induce symptoms of malnutrition. After malnourishment, the piglets were re-fed using different regimes all based on general food aid products, namely Corn-Soy blend (CSB) fortified with phosphorus (CSB+), CSB fortified with phosphorus and skim milk powder (CSB++) and CSB fortified with phosphorus and added whey permeate (CSB + P). RESULTS: Malnourishment had profound impact on the GM of the piglets leading to a less diverse GM dominated especially by Akkermansia spp. as determined by 16S rRNA gene amplicon sequencing. All three re-feeding regimes partly restored GM, leading to a more diverse GM compositionally closer to that of well-nourished piglets. This effect was even more pronounced for CSB++ compared to CSB+ and CSB + P. CONCLUSION: The GM of piglets were profoundly disturbed by malnourishment resulting in significantly increased abundance of Akkermansia spp. CSB++ may have superior effect on recovering GM diversity compared to the two other food aid products used in this study.


Asunto(s)
Alimentación Animal/análisis , Disbiosis , Microbioma Gastrointestinal , Desnutrición/complicaciones , Factores de Edad , Animales , Bacterias/clasificación , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Desnutrición/microbiología , Leche , Fósforo/administración & dosificación , ARN Ribosómico 16S/genética , Glycine max , Porcinos , Destete , Proteína de Suero de Leche/administración & dosificación , Zea mays
12.
Eur J Nutr ; 58(3): 909-930, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29644395

RESUMEN

Nutritional restrictions during the first 1000 days of life can impair or delay the physical and cognitive development of the individual and have long-term consequences for their health. Metabolic phenotyping (metabolomics/metabonomics) simultaneously measures a diverse range of low molecular weight metabolites in a sample providing a comprehensive assessment of the individual's biochemical status. There are a growing number of studies applying such approaches to characterize the metabolic derangements induced by various forms of early-life malnutrition. This includes acute and chronic undernutrition and specific micronutrient deficiencies. Collectively, these studies highlight the diverse and dynamic metabolic disruptions resulting from various forms of nutritional deficiencies. Perturbations were observed in many pathways including those involved in energy, amino acid, and bile acid metabolism, the metabolic interactions between the gut microbiota and the host, and changes in metabolites associated with gut health. The information gleaned from such studies provides novel insights into the mechanisms linking malnutrition with developmental impairments and assists in the elucidation of candidate biomarkers to identify individuals at risk of developmental shortfalls. As the metabolic profile represents a snapshot of the biochemical status of an individual at a given time, there is great potential to use this information to tailor interventional strategies specifically to the metabolic needs of the individual.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Desnutrición/diagnóstico , Desnutrición/microbiología , Metaboloma/fisiología , Metabolómica/métodos , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Fenotipo
13.
Gastroenterol Hepatol ; 42(1): 1-10, 2019 Jan.
Artículo en Inglés, Español | MEDLINE | ID: mdl-30197248

RESUMEN

BACKGROUND: Available evidence assessing the impact of small intestinal bacterial overgrowth (SIBO) following gastrectomy is limited. OBJECTIVES: To evaluate the prevalence of SIBO after gastrectomy and its association with malnutrition. To describe the antibiotic treatment required to correct it and if nutritional status improves. MATERIAL AND METHODS: A prospective cohort study was performed at the Agencia Sanitaria Costa del Sol (Costa del Sol Health Agency) from 2012 to 2015. A hydrogen-methane breath test with oral glucose overload was performed. Demographic variables and nutritional parameters were collected at baseline and one month after effective treatment of SIBO. The antibiotic regimens and the number of treatment lines used were assessed. RESULTS: Sixty gastrectomy patients were analysed, 58.3% of which were male. A sub-analysis of the curve was performed at 45min to minimise possible false positives, and SIBO was identified in 61.6% of cases. SIBO patients tended to have a lower BMI, although this trend was not statistically significant. After treatment with rifaximin, 94.6% of patients were still positive for SIBO, which fell to 85.7% after metronidazole. The rate of total antibiotic treatment failure was 67.6%. No statistically significant changes were found in nutritional parameters after treatment. CONCLUSIONS: SIBO was identified in 61.6% of patients after gastrectomy. No correlation was found with any malnutrition parameter. Rifaximin and metronidazole were found to be largely ineffective in eradicating SIBO. When treatment was effective, the impact on malnutrition was negligible and may have been associated with other factors.


Asunto(s)
Gastrectomía , Intestino Delgado/microbiología , Estado Nutricional , Femenino , Humanos , Masculino , Desnutrición/epidemiología , Desnutrición/microbiología , Persona de Mediana Edad , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/microbiología , Estudios Prospectivos
14.
Biol Reprod ; 98(4): 579-592, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29324977

RESUMEN

Malnutrition is a global threat to pregnancy health and impacts offspring development. Establishing an optimal pregnancy environment requires the coordination of maternal metabolic and immune pathways, which converge at the gut. Diet, metabolic, and immune dysfunctions have been associated with gut dysbiosis in the nonpregnant individual. In pregnancy, these states are associated with poor pregnancy outcomes and offspring development. However, the impact of malnutrition on maternal gut microbes, and their relationships with maternal metabolic and immune status, has been largely underexplored. To determine the impact of undernutrition and overnutrition on maternal metabolic status, inflammation, and the microbiome, and whether relationships exist between these systems, pregnant mice were fed either a normal, calorically restricted (CR), or a high fat (HF) diet. In late pregnancy, maternal inflammatory and metabolic biomarkers were measured and the cecal microbiome was characterized. Microbial richness was reduced in HF mothers although they did not gain more weight than controls. First trimester weight gain was associated with differences in the microbiome. Microbial abundance was associated with altered plasma and gut inflammatory phenotypes and peripheral leptin levels. Taxa potentially protective against elevated maternal leptin, without the requirement of a CR diet, were identified. Suboptimal dietary conditions common during pregnancy adversely impact maternal metabolic and immune status and the microbiome. HF nutrition exerts the greatest pressures on maternal microbial dynamics and inflammation. Key gut bacteria may mediate local and peripheral inflammatory events in response to maternal nutrient and metabolic status, with implications for maternal and offspring health.


Asunto(s)
Peso Corporal/fisiología , Ciego/microbiología , Microbioma Gastrointestinal/fisiología , Desnutrición/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Animales , Restricción Calórica , Dieta Alta en Grasa , Femenino , Desnutrición/inmunología , Desnutrición/microbiología , Ratones , Embarazo
15.
Curr Opin Clin Nutr Metab Care ; 21(3): 179-183, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29432296

RESUMEN

PURPOSE OF REVIEW: This review focuses on the recent discoveries about the impact of intestinal microbiota on mammalian host juvenile growth. RECENT FINDINGS: Intestinal microbiota is a powerful modulator of many facets of multicellular host's physiology. Recent results from human field studies and animal research have clearly shown that not only the nutrition, but also the intestinal microbiota impacts host postnatal growth kinetics. Absence of microbiome leads to stunted growth in mammalian gnotobiotic models and changes in the composition of the intestinal microbiota can impact the postnatal growth kinetics both positively and negatively under normal nutritional conditions as well as in undernutrition. Strikingly, specific bacterial strains are able to interact with GH/IGF-1 somatotropic axis activity, thus directly impacting host juvenile development. SUMMARY: Intestinal microbiota dictates the pace of host postnatal growth. This newly described role envisages that therapy with specific bacterial strains, together with re-nutritional strategies, might successfully alleviate the long-term sequelae of undernutrition during childhood in humans.


Asunto(s)
Microbioma Gastrointestinal , Trastornos del Crecimiento/microbiología , Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Intestinos/microbiología , Desnutrición/microbiología , Animales , Bacterias , Crecimiento , Trastornos del Crecimiento/etiología , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/prevención & control , Humanos , Desnutrición/complicaciones , Desnutrición/prevención & control , Mamíferos/microbiología , Probióticos/uso terapéutico
16.
Eur J Clin Microbiol Infect Dis ; 37(10): 2011-2020, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30051355

RESUMEN

Campylobacter spp. have been associated with anthropometric Z-score decrements, but the role of specific virulence genes associated with these outcomes has not been explored. This study aimed to investigate whether specific Campylobacter jejuni virulence-related gene and immune-inflammatory biomarkers are associated with malnutrition in children from Northeastern Brazil. A case-control study was performed in Fortaleza, Brazil. Children aging 6-24 months were characterized as malnourished (cases) if weight-for-age Z-score (WAZ) = 2 and as nourished (controls) if WAZ ≥ 1. DNA samples were extracted from stools and screened for C. jejuni/coli by real-time PCR. A subsequent C. jejuni-specific PCR was employed and positive samples were evaluated for 18 C. jejuni virulence genes by using four multiplex PCRs. C. jejuni was detected in 9.71% (33/340) of the children's samples, being 63.63% (21/33) from nourished and 37.37% (12/33) from malnourished children. The cadF, iamA, cheW, and sodB genes were the most frequent genes (100%, 90.9%, 87.9%, and 75.8%, respectively), while some others (ceuE, jlpA, pldA, and pVir) showed low rates (all below 6%). Malnourished children were significantly associated with infection with C. jejuni strains lacking cdtB gene (active subunit of cytolethal distending toxin) and harboring flgE gene (flagellar hook protein). These strains were also associated with children presenting increased serum SAA and sCD-14, but decreased IgG anti-LPS. These data reinforce the impact of Campylobacter jejuni infection on children without diarrhea and highlight the contribution of a specific virulence gene profile, cdtB(-)flgE(+) and increased systemic response in malnutrition children.


Asunto(s)
Infecciones por Campylobacter/epidemiología , Campylobacter jejuni/genética , Campylobacter jejuni/patogenicidad , Desnutrición/microbiología , Toxinas Bacterianas/genética , Biomarcadores/análisis , Biomarcadores/orina , Brasil , Infecciones por Campylobacter/complicaciones , Infecciones por Campylobacter/microbiología , Preescolar , Diarrea/microbiología , Heces/microbiología , Femenino , Trastornos del Crecimiento/microbiología , Humanos , Lactante , Receptores de Lipopolisacáridos/sangre , Receptores de Lipopolisacáridos/inmunología , Masculino , Desnutrición/inmunología , Virulencia/genética
17.
BMC Gastroenterol ; 18(1): 93, 2018 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-29929472

RESUMEN

BACKGROUND: Human rotavirus (HRV) is a major cause of viral gastroenteritis in infants; particularly in developing countries where malnutrition is prevalent. Malnutrition perturbs the infant gut microbiota leading to sub-optimal functioning of the immune system and further predisposing infants to enteric infections. Therefore, we hypothesized that malnutrition exacerbates rotavirus disease severity in infants. METHODS: In the present study, we used a neonatal germ free (GF) piglets transplanted with a two-month-old human infant's fecal microbiota (HIFM) on protein deficient and sufficient diets. We report the effects of malnourishment on the HRV infection and the HIFM pig microbiota in feces, intestinal and systemic tissues, using MiSeq 16S gene sequencing (V4-V5 region). RESULTS: Microbiota analysis indicated that the HIFM transplantation resulted in a microbial composition in pigs similar to that of the original infant feces. This model was then used to understand the interconnections between microbiota diversity, diet, and HRV infection. Post HRV infection, HIFM pigs on the deficient diet had lower body weights, developed more severe diarrhea and increased virus shedding compared to HIFM pigs on sufficient diet. However, HRV induced diarrhea and shedding was more pronounced in non-colonized GF pigs compared to HIFM pigs on either sufficient or deficient diet, suggesting that the microbiota alone moderated HRV infection. HRV infected pigs on sufficient diet showed increased microbiota diversity in intestinal tissues; whereas, greater diversity was observed in systemic tissues of HRV infected pigs fed with deficient diet. CONCLUSIONS: These results suggest that proper nourishment improves the microbiota quality in the intestines, alleviates HRV disease and lower probability of systemic translocation of potential opportunistic pathogens/pathobionts. In conclusion, our findings further support the role for microbiota and proper nutrition in limiting enteric diseases.


Asunto(s)
Gastroenteritis/complicaciones , Gastroenteritis/microbiología , Microbioma Gastrointestinal , Desnutrición/complicaciones , Desnutrición/microbiología , Infecciones por Rotavirus/complicaciones , Infecciones por Rotavirus/microbiología , Animales , Diarrea/microbiología , Diarrea/virología , Susceptibilidad a Enfermedades , Heces/microbiología , Gastroenteritis/virología , Humanos , Lactante , Intestinos/microbiología , Desnutrición/virología , ARN Ribosómico 16S , Infecciones por Rotavirus/virología , Análisis de Secuencia de ARN , Porcinos , Esparcimiento de Virus , Aumento de Peso
18.
Hepatobiliary Pancreat Dis Int ; 17(4): 290-300, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30173786

RESUMEN

BACKGROUND: Patients with end-stage liver disease (ESLD) have a compromised nutritional status because of the liver crucial role in regulating metabolic homeostasis and energy balance. DATA SOURCES: A systematic review of literature based on extensive relevant articles published from 2001 to 2017 in English in PubMed database was performed by searching keywords such as liver disease, non-alcoholic liver disease, alcoholic liver disease, malnutrition, epigenetics, gut microbiota, and probiotics. RESULTS: Liver transplantation would be one eligible therapy for ESLD patients, even if, the clinical outcome is negatively influenced by malnutrition and/or infections. The malnutrition is a condition of nutrient imbalance with a high incidence in ESLD patients. An accurate evaluation of nutritional status could be fundamental for reducing complications and prolonging the survival of ESLD patients including those undergoing liver transplantation. In addition, the interaction among nutrients, diet and genes via epigenetics has emerged as a potential target to reduce the morbidity and mortality in ESLD patients. The malnutrition induces changes in gut microbiota causing dysbiosis with a probable translocation of bacteria and/or pathogen-derived factors from the intestine to the liver. Gut microbiota contribute to the progression of chronic liver diseases as well as hepatocellular carcinoma. The administration of probiotics modulating gut microbiota could improve all chronic liver diseases. CONCLUSIONS: This review provides an update on malnutrition status linked to epigenetics and the potential benefit of some probiotics on the management of ESLD patients. In support of this view and to reveal the constant and growing interest in this field, some clinical trials are reported.


Asunto(s)
Bacterias/patogenicidad , Enfermedad Hepática en Estado Terminal/microbiología , Metabolismo Energético , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Desnutrición/microbiología , Estado Nutricional , Animales , Traslocación Bacteriana , Disbiosis , Enfermedad Hepática en Estado Terminal/genética , Enfermedad Hepática en Estado Terminal/fisiopatología , Enfermedad Hepática en Estado Terminal/terapia , Metabolismo Energético/genética , Epigénesis Genética , Tracto Gastrointestinal/metabolismo , Interacción Gen-Ambiente , Interacciones Huésped-Patógeno , Humanos , Desnutrición/genética , Desnutrición/fisiopatología , Desnutrición/terapia , Estado Nutricional/genética , Probióticos/uso terapéutico , Pronóstico
19.
Microb Pathog ; 106: 127-138, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-26853753

RESUMEN

Malnutrition is the leading cause of death worldwide in children under the age of five, and is the focus of the first World Health Organization (WHO) Millennium Development Goal. Breastfeeding, food and water security are major protective factors against malnutrition and critical factors in the maturation of healthy gut microbiota, characterized by a transient bifidobacterial bloom before a global rise in anaerobes. Early depletion in gut Bifidobacterium longum, a typical maternal probiotic, known to inhibit pathogens, represents the first step in gut microbiota alteration associated with severe acute malnutrition (SAM). Later, the absence of the Healthy Mature Anaerobic Gut Microbiota (HMAGM) leads to deficient energy harvest, vitamin biosynthesis and immune protection, and is associated with diarrhea, malabsorption and systemic invasion by microbial pathogens. A therapeutic diet and infection treatment may be unable to restore bifidobacteria and HMAGM. Besides refeeding and antibiotics, future trials including non-toxic missing microbes and nutrients necessary to restore bifidobacteria and HMAGM, including prebiotics and antioxidants, are warranted in children with severe or refractory disease.


Asunto(s)
Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Desnutrición/microbiología , Animales , Antibacterianos , Antioxidantes , Bacterias/clasificación , Bacterias/crecimiento & desarrollo , Bacterias/patogenicidad , Bacterias Aerobias/crecimiento & desarrollo , Bacterias Anaerobias/crecimiento & desarrollo , Bifidobacterium/crecimiento & desarrollo , Biodiversidad , Preescolar , Diarrea , Dieta , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Humanos , Inmunoglobulina A , Infecciones , Kwashiorkor , Síndromes de Malabsorción , Desnutrición/tratamiento farmacológico , Desnutrición/metabolismo , Parásitos/clasificación , Parásitos/crecimiento & desarrollo , Filogenia , Probióticos/uso terapéutico , Factores de Riesgo , Sepsis , Desnutrición Aguda Severa , Virus/clasificación , Virus/crecimiento & desarrollo
20.
J Pediatr Gastroenterol Nutr ; 65(5): 491-495, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28489672

RESUMEN

Malnutrition contributes significantly to death and illness worldwide and especially to the deaths of children younger than 5 years. The relation between intestinal changes in malnutrition and morbidity and mortality has not been well characterized; however, recent research indicates that the functional and morphologic changes of the intestine secondary to malnutrition itself contribute significantly to these negative clinical outcomes and may be potent targets of intervention. The aim of this review was to summarize current knowledge of experimental and clinically observed changes in the intestine from malnutrition preclinical models and human studies. Limited clinical studies have shown villous blunting, intestinal inflammation, and changes in the intestinal microbiome of malnourished children. In addition to these findings, experimental data using various animal models of malnutrition have found evidence of increased intestinal permeability, upregulated intestinal inflammation, and loss of goblet cells. More mechanistic studies are urgently needed to improve our understanding of malnutrition-related intestinal dysfunction and to identify potential novel targets for intervention.


Asunto(s)
Intestinos/patología , Intestinos/fisiopatología , Desnutrición/patología , Desnutrición/fisiopatología , Animales , Microbioma Gastrointestinal , Humanos , Inflamación/microbiología , Inflamación/patología , Inflamación/fisiopatología , Intestinos/microbiología , Desnutrición/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA