Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nutr Cancer ; 74(5): 1701-1711, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34353196

RESUMO

Compelling animal studies report increased intestinal permeability, inflammation, and colorectal carcinogenesis with exposure to certain emulsifiers commonly added to processed foods, but human data are lacking. Highly processed food consumption is also associated with obesity and higher risk of chronic diseases. We cross-sectionally examined the association of emulsifier and highly processed food consumption estimated from six 24-h dietary recalls among 588 U.S. men and women over one year, with biomarkers of intestinal permeability and inflammation measured from two fasting blood samples collected six months apart. In multivariable-adjusted generalized linear models, greater emulsifier intake (g/d) was not associated with antibodies to flagellin (P-trend = 0.88), lipopolysaccharide (LPS) (P-trend = 0.56), or the combined total thereof (P-trend = 0.65) but was positively associated with an inflammatory biomarker, glycoprotein acetyls (GlycA) (P-trend = 0.02). Highly processed food intake (% kcal/d) was associated with higher anti-LPS antibodies (P-trend = 0.001) and total anti-flagellin and anti-LPS antibodies (P-trend = 0.005) but not with other biomarkers, whereas processed food intake expressed as % g/d was associated with higher GlycA (P-trend = 0.02). Our findings suggest that, broadly, highly processed food consumption may be associated with intestinal permeability biomarkers, and both emulsifier and highly processed food intakes may be associated with inflammation. Additional studies are warranted to further evaluate these relationships.Supplemental data for this article is available online at https://doi.org/10.1080/01635581.2021.1957947.


Assuntos
Dieta , Neoplasias , Animais , Biomarcadores , Ingestão de Alimentos , Ingestão de Energia , Fast Foods , Feminino , Humanos , Inflamação , Permeabilidade
2.
Cancer Prev Res (Phila) ; 14(3): 393-402, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33229339

RESUMO

Gut barrier dysfunction promotes chronic inflammation, contributing to several gastrointestinal diseases, including colorectal cancer. Preliminary evidence suggests that vitamin D and calcium could prevent colorectal carcinogenesis, in part, by influencing gut barrier function. However, relevant human data are scarce. We tested the effects of supplemental calcium (1,200 mg/day) and/or vitamin D3 (1,000 IU/day) on circulating concentrations of biomarkers of gut permeability (anti-flagellin and anti-lipopolysaccharide IgA and IgG, measured via ELISA) from baseline to 1 and 3 or 5 years postbaseline among 175 patients with colorectal adenoma in a randomized, double-blinded, placebo-controlled clinical trial. We also assessed factors associated with baseline concentrations of these biomarkers. We found no appreciable effects of supplemental vitamin D3 and/or calcium on individual or aggregate biomarkers of gut permeability. At baseline, a combined permeability score (the summed concentrations of all four biomarkers) was 14% lower among women (P = 0.01) and 10% higher among those who consumed >1 serving per day of red or processed meats relative to those who consumed none (P trend = 0.03). The permeability score was estimated to be 49% higher among participants with a body mass index (BMI) > 35 kg/m2 relative to those with a BMI < 22.5 kg/m2 (P trend = 0.17). Our results suggest that daily supplemental vitamin D3 and/or calcium may not modify circulating concentrations of gut permeability biomarkers within 1 or 3-5 years, but support continued investigation of modifiable factors, such as diet and excess adiposity, that could affect gut permeability. PREVENTION RELEVANCE: Calcium and vitamin D may be involved in regulating and maintaining the integrity of the intestinal mucosal barrier, the dysfunction of which results in exposure of the host to luminal bacteria, endotoxins, and antigens leading to potentially cancer-promoting endotoxemia and chronic colon inflammation. While our results suggest that daily supplementation with these chemopreventive agents does not modify circulating concentrations of gut permeability biomarkers, they support continued investigation of other potential modifiable factors, such as diet and excess adiposity, that could alter gut barrier function, to inform the development of treatable biomarkers of risk for colorectal neoplasms and effective colon cancer preventive strategies.


Assuntos
Adenoma/tratamento farmacológico , Biomarcadores Tumorais/sangue , Cálcio da Dieta/administração & dosagem , Neoplasias do Colo/tratamento farmacológico , Suplementos Nutricionais , Trato Gastrointestinal/efeitos dos fármacos , Vitamina D/administração & dosagem , Adenoma/metabolismo , Adenoma/patologia , Idoso , Cálcio da Dieta/sangue , Estudos de Casos e Controles , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Método Duplo-Cego , Feminino , Seguimentos , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Permeabilidade , Prognóstico , Vitamina D/sangue , Vitaminas/administração & dosagem , Vitaminas/sangue
3.
J Nutr ; 150(8): 2175-2182, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32455424

RESUMO

BACKGROUND: Environmental enteric dysfunction (EED), characterized by altered intestinal permeability/inflammation, microbial translocation, and systemic inflammation (SI), may be a significant contributor to micronutrient deficiencies and poor growth in infants from low-resource settings. OBJECTIVE: We examined associations among EED, SI, growth, and iron status at 6 mo of age. METHODS: We performed a cross-sectional analysis of 6-mo-old infants (n = 548) enrolled in a Ugandan birth-cohort study (NCT04233944). EED was assessed via serum concentrations of anti-flagellin and anti- LPS immunoglobulins (Igs); SI was assessed via serum concentrations of ɑ1-acid glycoprotein (AGP) and C-reactive protein (CRP); iron status was assessed via serum concentrations of hemoglobin (Hb), soluble transferrin receptor (sTfR), and ferritin. Associations were assessed using adjusted linear regression analysis. RESULTS: At 6 mo, ∼35% of infants were stunted [length-for-age z score (LAZ) < -2] and ∼53% were anemic [hemoglobin (Hb) <11.0 g/dL]. Nearly half (∼46%) had elevated AGP (>1 g/L) and ∼30% had elevated CRP (>5 mg/L). EED and SI biomarkers were significantly correlated (r = 0.142-0.193, P < 0.001 for all). In adjusted linear regression models, which included adjustments for SI, higher anti-flagellin IgA, anti-LPS IgA, and anti-LPS IgG concentrations were each significantly associated with lower LAZ [ß (95% CI): -0.21 (-0.41, 0.00), -0.23 (-0.44, -0.03), and -0.33 (-0.58, -0.09)]. Furthermore, higher anti-flagellin IgA, anti-flagellin IgG, and anti-LPS IgA concentrations were significantly associated with lower Hb [ß (95% CI): -0.24 (-0.45, -0.02), -0.58 (-1.13, 0.00), and -0.26 (-0.51, 0.00)] and higher anti-flagellin IgG and anti-LPS IgG concentrations were significantly associated with higher sTfR [ß (95% CI): 2.31 (0.34, 4.28) and 3.13 (0.75, 5.51)]. CONCLUSIONS: EED is associated with both low LAZ and iron status in 6-mo-old infants. Further research on the mechanisms by which EED affects growth and micronutrient status is warranted.


Assuntos
Anemia Ferropriva/epidemiologia , Anemia Ferropriva/etiologia , Desenvolvimento Infantil , Enteropatias/microbiologia , Enteropatias/patologia , População Rural , Adulto , Estudos de Coortes , Feminino , Microbioma Gastrointestinal , Humanos , Lactente , Inflamação , Enteropatias/epidemiologia , Masculino , Uganda/epidemiologia , Adulto Jovem
4.
Cell Mol Gastroenterol Hepatol ; 9(2): 313-333, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31593782

RESUMO

BACKGROUND & AIMS: Consumption of a low-fiber, high-fat, Western-style diet (WSD) induces adiposity and adipose inflammation characterized by increases in the M1:M2 macrophage ratio and proinflammatory cytokine expression, both of which contribute to WSD-induced metabolic syndrome. WSD-induced adipose inflammation might result from endoplasmic reticulum stress in lipid-overloaded adipocytes and/or dissemination of gut bacterial products, resulting in activation of innate immune signaling. Hence, we aimed to investigate the role of the gut microbiota, and its detection by innate immune signaling pathways, in WSD-induced adipose inflammation. METHODS: Mice were fed grain-based chow or a WSD for 8 weeks, assessed metabolically, and intestinal and adipose tissue were analyzed by flow cytometry and quantitative reverse transcription polymerase chain reaction. Microbiota was ablated via antibiotics and use of gnotobiotic mice that completely lacked microbiota (germ-free mice) or had a low-complexity microbiota (altered Schaedler flora). Innate immune signaling was ablated by genetic deletion of Toll-like receptor signaling adaptor myeloid differentiation primary response 88. RESULTS: Ablation of microbiota via antibiotic, germ-free, or altered Schaedler flora approaches did not significantly impact WSD-induced adiposity, yet dramatically reduced WSD-induced adipose inflammation as assessed by macrophage populations and cytokine expression. Microbiota ablation also prevented colonic neutrophil and CD103- dendritic cell infiltration. Such reduced indices of inflammation correlated with protection against WSD-induced dysglycemia, hypercholesterolemia, and liver dysfunction. Genetic deletion of myeloid differentiation primary response 88 also prevented WSD-induced adipose inflammation. CONCLUSIONS: These results indicate that adipose inflammation, and some aspects of metabolic syndrome, are not purely a consequence of diet-induced adiposity per se but, rather, may require disturbance of intestine-microbiota interactions and subsequent activation of innate immunity.


Assuntos
Tecido Adiposo/imunologia , Adiposidade/imunologia , Dieta Ocidental/efeitos adversos , Microbioma Gastrointestinal/imunologia , Síndrome Metabólica/imunologia , Adipócitos/imunologia , Adipócitos/metabolismo , Tecido Adiposo/patologia , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/imunologia , Transplante de Microbiota Fecal , Fezes/microbiologia , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Macrófagos/imunologia , Masculino , Síndrome Metabólica/microbiologia , Camundongos , Transdução de Sinais
5.
Nat Commun ; 10(1): 5650, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31827095

RESUMO

Alterations in gut microbiota composition are associated with metabolic syndrome and chronic inflammatory diseases such as inflammatory bowel disease. One feature of inflammation-associated gut microbiotas is enrichment of motile bacteria, which can facilitate microbiota encroachment into the mucosa and activate pro-inflammatory gene expression. Here, we set out to investigate whether elicitation of mucosal anti-flagellin antibodies by direct administration of purified flagellin might serve as a general vaccine against subsequent development of chronic gut inflammation. We show, in mice, that repeated injection of flagellin elicits increases in fecal anti-flagellin IgA and alterations in microbiota composition, reduces fecal flagellin concentration, prevents microbiota encroachment, protects against IL-10 deficiency-induced colitis, and ameliorates diet-induced obesity. Flagellin's impact on the microbiota is B-lymphocyte dependent and, in humans, obese subjects exhibit increased levels of fecal flagellin and reduced levels of fecal flagellin-specific IgA, relative to normal weight subjects. Thus, administration of flagellin, and perhaps other pathobiont antigens, may confer some protection against chronic inflammatory diseases.


Assuntos
Imunidade Adaptativa , Vacinas Bacterianas/imunologia , Colite/prevenção & controle , Flagelina/imunologia , Microbioma Gastrointestinal , Animais , Anticorpos Antibacterianos/imunologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/genética , Colite/genética , Colite/imunologia , Colite/microbiologia , Fezes/microbiologia , Flagelina/administração & dosagem , Flagelina/genética , Humanos , Imunoglobulina A/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/imunologia , Obesidade/microbiologia , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia
6.
Am J Physiol Endocrinol Metab ; 317(6): E1121-E1130, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31573841

RESUMO

Metformin beneficially impacts several aspects of metabolic syndrome including dysglycemia, obesity, and liver dysfunction, thus making it a widely used frontline treatment for early-stage type 2 diabetes, which is associated with these disorders. Several mechanisms of action for metformin have been proposed, including that it acts as an anti-inflammatory agent, possibly as a result of its impact on intestinal microbiota. In accord with this possibility, we observed herein that, in mice with diet-induced metabolic syndrome, metformin impacts the gut microbiota by preventing its encroachment upon the host, a feature of metabolic syndrome in mice and humans. However, the ability of metformin to beneficially impact metabolic syndrome in mice was not markedly altered by reduction or elimination of gut microbiota, achieved by the use of antibiotics or germfree mice. Although reducing or eliminating microbiota by itself suppressed diet-induced dysglycemia, other features of metabolic syndrome including obesity, hepatic steatosis, and low-grade inflammation remained suppressed by metformin in the presence or absence of gut microbiota. These results support a role for anti-inflammatory activity of metformin, irrespective of gut microbiota, in driving some of the beneficial impacts of this drug on metabolic syndrome.


Assuntos
Microbioma Gastrointestinal/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Inflamação/metabolismo , Fígado/efeitos dos fármacos , Síndrome Metabólica/metabolismo , Metformina/farmacologia , Ampicilina/farmacologia , Animais , Antibacterianos/farmacologia , Dieta Hiperlipídica , Fígado Gorduroso/metabolismo , Fígado Gorduroso/microbiologia , Microbioma Gastrointestinal/fisiologia , Vida Livre de Germes , Hiperglicemia/metabolismo , Hiperglicemia/microbiologia , Inflamação/microbiologia , Fígado/metabolismo , Síndrome Metabólica/microbiologia , Camundongos , Neomicina/farmacologia , Obesidade/metabolismo , Obesidade/microbiologia
7.
Mol Cell Proteomics ; 18(9): 1864-1879, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31262998

RESUMO

Consumption of refined high-fat, low-fiber diets promotes development of obesity and its associated consequences. Although genetics play an important role in dictating susceptibility to such obesogenic diets, mice with nearly uniform genetics exhibit marked heterogeneity in their extent of obesity in response to such diets. This suggests non-genetic determinants play a role in diet-induced obesity. Hence, we sought to identify parameters that predict, and/or correlate with, development of obesity in response to an obesogenic diet. We assayed behavior, metabolic parameters, inflammatory markers/cytokines, microbiota composition, and the fecal metaproteome, in a cohort of mice (n = 50) prior to, and the 8 weeks following, administration of an obesogenic high-fat low-fiber diet. Neither behavioral testing nor quantitation of inflammatory markers broadly predicted severity of diet-induced obesity. Although, the small subset of mice that exhibited basal elevations in serum IL-6 (n = 5) were among the more obese mice in the cohort. While fecal microbiota composition changed markedly in response to the obesogenic diet, it lacked the ability to predict which mice were relative prone or resistant to obesity. In contrast, fecal metaproteome analysis revealed functional and taxonomic differences among the proteins associated with proneness to obesity. Targeted interrogation of microbiota composition data successfully validated the taxonomic differences seen in the metaproteome. Although future work will be needed to determine the breadth of applicability of these associations to other cohorts of animals and humans, this study nonetheless highlights the potential power of gut microbial proteins to predict and perhaps impact development of obesity.


Assuntos
Fezes/microbiologia , Microbioma Gastrointestinal/fisiologia , Obesidade/etiologia , Proteoma/metabolismo , Animais , Composição Corporal , Dieta Hiperlipídica/efeitos adversos , Dieta Ocidental/efeitos adversos , Feminino , Flagelina/metabolismo , Microbioma Gastrointestinal/genética , Imunoglobulina A/sangue , Mediadores da Inflamação/metabolismo , Lipocalina-2/metabolismo , Síndrome Metabólica/etiologia , Síndrome Metabólica/microbiologia , Camundongos Endogâmicos C57BL , Obesidade/microbiologia , Proteoma/análise , RNA Ribossômico 16S
8.
J Pediatr ; 210: 34-40.e1, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30952509

RESUMO

OBJECTIVE: To examine whether daily zinc and/or multivitamin supplementation reduce biomarkers of environmental enteric dysfunction (EED), systemic inflammation, or markers of growth in a sample of infants from Dar es Salaam, Tanzania. STUDY DESIGN: Subgroup analysis of infants participating in a randomized, double-blind, placebo-controlled trial received daily oral supplementation of zinc, multivitamins, zinc + multivitamins, or placebo for 18 months starting at 6 weeks of age. EED (anti-flagellin and anti-lipopolysaccharide immunoglobulins), systemic inflammation (C-reactive protein and alpha-1-acid glycoprotein), and growth biomarkers (insulin-like growth factor-1 and insulin-like growth factor binding protein-3) were measured via enzyme-linked immunosorbent assay in a subsample of 590 infants at 6 weeks and 6 months of age. EED biomarkers also were measured in 162 infants at 12 months of age. RESULTS: With the exception of anti-lipopolysaccharide IgG concentrations, which were significantly greater in infants who received multivitamins compared with those who did not (1.41 ± 0.61 vs 1.26 ± 0.65, P = .006), and insulin-like growth factor binding protein-3 concentrations, which were significantly lower in children who received zinc compared with those who did not (981.13 ± 297.59 vs 1019.10 ± 333.01, P = .03), at 6 months of age, we did not observe any significant treatment effects of zinc or multivitamins on EED, systemic inflammation, or growth biomarkers. CONCLUSIONS: Neither zinc nor multivitamin supplementation ameliorated markers of EED or systemic inflammation during infancy. Other interventions should be prioritized for future trials. TRIAL REGISTRATION: Clinicaltrials.gov: NCT00421668.


Assuntos
Suplementos Nutricionais , Inflamação/sangue , Inflamação/tratamento farmacológico , Enteropatias/sangue , Enteropatias/tratamento farmacológico , Intestino Delgado , Vitaminas/uso terapêutico , Zinco/uso terapêutico , Biomarcadores/sangue , Método Duplo-Cego , Feminino , Humanos , Lactente , Inflamação/complicações , Enteropatias/complicações , Masculino , Tanzânia , Resultado do Tratamento
9.
Am J Clin Nutr ; 108(4): 889-896, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30247538

RESUMO

Background: Adverse birth outcomes, including preterm birth and stunting at birth, have long-term health implications. The relation between adverse birth outcomes and chronic, asymptomatic gastrointestinal inflammation (environmental enteric dysfunction-EED) is poorly understood. Objective: We aimed to examine the relation between maternal EED and adverse birth outcomes in a sample of pregnant Ugandan women and their newborn infants. Design: We conducted a prospective cohort study in Mukono, Uganda. A total of 258 pregnant women were enrolled at their first prenatal visit (∼18 weeks of gestation). EED was measured by urinary lactulose:mannitol (L:M) ratio and serum concentrations of antibodies to the bacterial components flagellin and LPS. Covariates were obtained from survey data collected at 2 time points. Associations were assessed through the use of unadjusted and adjusted simple linear regression models. Results: Complete birth outcome data were recorded for 220 infants within 48 h of delivery. Mean ± SD gestational age was 39.7 ± 2.1 wk, and 7% were born preterm. Mean ± SD length and length-for-age z score (LAZ) at birth were 48.1 ± 3.2 cm and -0.44 ± 1.07, respectively. L:M ratio was not associated with any birth outcome. In adjusted models, higher concentrations of natural log-transformed anti-flagellin immunoglobin G (IgG) and anti-LPS IgG were significantly associated with shorter length of gestation (ß: -0.89 wk; 95% CI: -1.77, -0.01 wk, and ß: -1.01 wk; 95% CI: -1.87, -0.17 wk, respectively) and with reduced length (ß: -0.80 cm; 95% CI: -1.55, -0.05 cm, and ß: -0.79 cm; 95% CI: -1.54, -0.04 cm, respectively) and LAZ at birth (ß -0.44 z score; 95% CI: -0.83, -0.05, and ß: -0.40 z score; 95% CI: -0.79, -0.01, respectively). Conclusion: Maternal anti-flagellin and anti-LPS IgG concentrations in pregnancy, but not L:M ratio, were associated with shorter gestation and reduced infant length at birth. Further research on the relation between maternal EED and birth outcomes is warranted.


Assuntos
Estatura , Enterite/fisiopatologia , Desenvolvimento Fetal , Idade Gestacional , Inflamação/complicações , Complicações na Gravidez/fisiopatologia , Nascimento Prematuro/etiologia , Adulto , Anticorpos/sangue , Enterite/sangue , Enterite/complicações , Feminino , Flagelina , Transtornos do Crescimento/etiologia , Humanos , Imunoglobulina G/sangue , Recém-Nascido , Intestino Delgado/patologia , Intestino Delgado/fisiopatologia , Lactulose/urina , Lipopolissacarídeos , Manitol/urina , Gravidez , Complicações na Gravidez/patologia , Estudos Prospectivos , Uganda , Adulto Jovem
10.
J Pediatr Gastroenterol Nutr ; 64(1): 104-108, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27347720

RESUMO

OBJECTIVE: Diarrheal diseases are a leading cause of morbidity and mortality worldwide, but the etiology of diarrhea and its relation to nutritional outcomes in resource-limited settings is poorly defined. We sought to determine the etiology of community-acquired diarrhea in Tanzanian infants and to assess the association with anthropometrics and novel intestinal biomarkers. METHODS: A convenience sample of infants in a trial of zinc and/or multivitamin supplementation in Tanzania was selected. Subjects were enrolled at age 6 weeks and studied for 18 months. Stool samples were obtained from children with acute diarrhea. A novel, polymerase chain reaction-based TaqMan array was used to screen stool for 15 enteropathogens. A subset of subjects had serum gastrointestinal biomarkers measured. RESULTS: One hundred twenty-three subjects with diarrhea were enrolled. The mean ± SD age at stool sample collection was 12.4 ±â€Š3.9 months. Thirty-five enteropathogens were identified in 34 (27.6%) subjects: 11 rotavirus, 9 Cryptosporidium spp, 7 Shigella spp, 3 Campylobacter jejuni/coli, 3 heat stable-enterotoxigenic Escherichia coli, and 2 enteropathogenic E coli. Subjects with any identified enteropathogen had significantly lower weight-for-length z scores (-0.55 ±â€Š1.10 vs 0.03 ±â€Š1.30, P = 0.03) at the final clinic visit than those without an identified pathogen. Fifty of the 123 subjects (40.7%) had serum analyzed for antibodies to lipopolysaccharide (LPS) and flagellin. Subjects with any identified enteropathogen had lower immunoglobulin (IgA) antibodies to LPS (0.75 ±â€Š0.27 vs 1.13 ±â€Š0.77, P = 0.01) and flagellin (0.52 ±â€Š0.16 vs 0.73 ±â€Š0.47, P = 0.02) than those without an identified pathogen. CONCLUSIONS: This quantitative polymerase chain reaction method may allow identification of enteropathogens that place children at higher risk for suboptimal growth. IgA anti-LPS and flagellin antibodies hold promise as emerging intestinal biomarkers.


Assuntos
Diarreia/etiologia , Flagelina/imunologia , Microbioma Gastrointestinal , Transtornos do Crescimento/etiologia , Imunoglobulina A/sangue , Intestinos , Lipopolissacarídeos/imunologia , Biomarcadores/sangue , Peso Corporal , Campylobacter/crescimento & desenvolvimento , Cryptosporidium/crescimento & desenvolvimento , Diarreia/microbiologia , Diarreia/parasitologia , Diarreia/virologia , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Fezes/microbiologia , Fezes/parasitologia , Fezes/virologia , Feminino , Transtornos do Crescimento/microbiologia , Transtornos do Crescimento/parasitologia , Transtornos do Crescimento/virologia , Humanos , Lactente , Infecções/complicações , Enteropatias/complicações , Intestinos/microbiologia , Intestinos/parasitologia , Intestinos/virologia , Masculino , Estado Nutricional , Reação em Cadeia da Polimerase , Rotavirus/crescimento & desenvolvimento , Shigella/crescimento & desenvolvimento , Tanzânia
11.
BMC Med ; 14: 27, 2016 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-26867587

RESUMO

The intestinal microbiota is a large and diverse microbial community that inhabits the intestine, containing about 100 trillion bacteria of 500-1000 distinct species that, collectively, provide benefits to the host. The human gut microbiota composition is determined by a myriad of factors, among them genetic and environmental, including diet and medication. The microbiota contributes to nutrient absorption and maturation of the immune system. As reciprocity, the host immune system plays a central role in shaping the composition and localization of the intestinal microbiota. Secretory immunoglobulins A (sIgAs), component of the adaptive immune system, are important player in the protection of epithelium, and are known to have an important impact on the regulation of microbiota composition. A recent study published in Immunity by Fransen and colleagues aimed to mechanistically decipher the interrelationship between sIgA and microbiota diversity/composition. This commentary will discuss these important new findings, as well as how future therapies can ultimately benefit from such discovery.


Assuntos
Microbioma Gastrointestinal/imunologia , Sistema Imunitário/fisiologia , Animais , Biodiversidade , Humanos , Deficiência de IgA/imunologia , Deficiência de IgA/microbiologia , Imunoglobulina A/fisiologia , Intestinos/imunologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...