Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Pediatr Res ; 77(1-2): 214-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25310761

RESUMO

Limited microbial exposure is suggested to underlie the increase of allergic diseases in affluent countries, and bacterial diversity seems to be more important than specific bacteria taxa. Prospective studies indicate that the gut microbiota composition during the first months of life influences allergy development, and support the theory that factors influencing the early maturation of the immune system might be important for subsequent allergic disease. However, recent research indicates that microbial exposure during pregnancy may be even more important for the preventative effects against allergic disease. This review gives a background of the epidemiology, immunology, and microbiology literature in this field. It focuses on possible underlying mechanisms such as immune-regulated epigenetic imprinting and bacterial translocation during pregnancy, potentially providing the offspring with a pioneer microbiome. We suggest that a possible reason for the initial exposure of bacterial molecular patterns to the fetus in utero is to prime the immune system and/or the epithelium to respond appropriately to pathogens and commensals after birth.


Assuntos
Epigênese Genética/imunologia , Trato Gastrointestinal/microbiologia , Hipersensibilidade/epidemiologia , Hipersensibilidade/imunologia , Hipersensibilidade/microbiologia , Microbiota , Efeitos Tardios da Exposição Pré-Natal/microbiologia , Feminino , Humanos , Modelos Biológicos , Gravidez
2.
FASEB J ; 24(10): 4078-88, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20519636

RESUMO

Gut commensals modulate host immune, endocrine, and metabolic functions. They also affect peripheral and central neural reflexes and function. We have previously shown that daily ingestion of Lactobacillus reuteri (LR) for 9 d inhibits the pseudoaffective cardiac response and spinal single-fiber discharge evoked by visceral distension, and decreases intestinal motility and myenteric AH cell slow afterhyperpolarization (sAHP) by inhibiting a Ca-activated K (IK(Ca)) channel. We tested whether luminal LR could acutely decrease motility in an ex vivo perfusion model of naive Balb/c jejunum. Live LR dose dependently decreased motor complex pressure wave amplitudes with 9- to 16-min onset latency and an IC(50) of 5 × 10(7) cells/ml Krebs. Heat-killed LR or another live commensal, Lactobacillus salivarius, were without effect. The IK(Ca) channel blocker TRAM-34, but neither the opener (DCEBIO) nor the hyperpolarization-activated cationic channel inhibitor ZD7288 (5 µM) (or TTX 1 µM), mimicked the LR effect on motility acutely ex vivo. We provide evidence for a rapid, strain-specific, dose-dependent action of a live Lactobacillus on small intestinal motility reflexes that recapitulates the long-term effects of LR ingestion. These observations may be useful as a first step to unraveling the pathways involved in bacteria to the nervous system communication.


Assuntos
Motilidade Gastrointestinal , Jejuno/fisiologia , Lactobacillus , Probióticos , Animais , Masculino , Camundongos , Camundongos Endogâmicos BALB C
3.
Mol Nutr Food Res ; 64(21): e2000519, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32926533

RESUMO

SCOPE: Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency and currently the leading cause of mortality in preterm infants. Recent studies show that human milk oligosaccharides (HMOs) reduce the frequency and incidence of NEC; however, the molecular mechanisms for their protection are largely unexplored. METHODS AND RESULTS: To address this gap, a genome-wide profiling of the intestinal epithelial transcriptome in response to HMOs using RNA-sequencing is performed. It is found that HMOs alter the host transcriptome in 225 unique target genes pertaining to cell proliferation and differentiation, including upregulation of stem cell differentiation marker HMGCS2. To validate these results, differentiation in Caco-2Bbe1 (Caco-2) intestinal cells is verified by Alcian Blue staining and transepithelial electrical resistance (TER) recordings. Furthermore, an in vivo model of NEC is also employed whereby neonatal pups are gavage fed HMOs. Interestingly, HMOs-fed pups show enhanced cell MUC2 differentiation and HMGCS2 expression. CONCLUSIONS: These findings demonstrate HMOs protect against NEC in part by altering the differentiation of the crypt-villus axis. In addition, this study suggests that pooled HMOs directly induce a series of biological processes, which provide mechanistic insights to how HMOs protect the host intestine.


Assuntos
Enterocolite Necrosante/patologia , Enterocolite Necrosante/prevenção & controle , Leite Humano/química , Oligossacarídeos/farmacologia , Animais , Células CACO-2 , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cães , Enterocolite Necrosante/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Hidroximetilglutaril-CoA Sintase/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Células Madin Darby de Rim Canino , Masculino , Camundongos Endogâmicos C57BL , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo
4.
Mol Nutr Food Res ; 63(3): e1800658, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30407734

RESUMO

SCOPE: Necrotizing enterocolitis (NEC) is a leading cause of morbidity and death in preterm infants, occurring more often in formula-fed than breastfed infants. Studies in both rats and humans show that human milk oligosaccharides (HMOs) lower the incidence of NEC, but the mechanism underlying such protection is currently unclear. METHODS AND RESULTS: By extracting HMOs from pooled human breastmilk, the impact of HMOs on the intestinal mucin levels in a murine model of NEC are investigated. To confirm the results, the findings are validated by exposing human intestinal epithelial cells and intestinal organoids to HMOs and evaluated for mucin expression. HMO-gavage to pups increases Muc2 levels and decreases intestinal permeability to macromolecular dextran. HMO-treated cells have increased Muc2 expression, decreased bacterial attachment and dextran permeability during challenge by enteric pathogens. To identify the mediators involved in HMO induction of mucins, it is demonstrated that HMOs directly induce the expression of chaperone proteins including protein disulfide isomerase (PDI). Suppression of PDI activity removes the protective effects of HMOs on barrier function in vitro as well as NEC protection in vivo. CONCLUSIONS: Taken together, the results provide insights to the possible mechanisms by which HMOs protect the neonatal intestine through upregulation of mucins.


Assuntos
Enterocolite Necrosante/prevenção & controle , Leite Humano/química , Mucina-2/genética , Oligossacarídeos/farmacologia , Animais , Animais Recém-Nascidos , Células CACO-2 , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Enterocolite Necrosante/metabolismo , Células Caliciformes/efeitos dos fármacos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Mucina-2/análise , Isomerases de Dissulfetos de Proteínas/fisiologia
5.
J Crohns Colitis ; 13(4): 431-441, 2019 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-30418545

RESUMO

BACKGROUND AND AIMS: For women with inflammatory bowel disease [IBD], it is not very well known how IBD or IBD treatment affects their breast milk components. We aimed to investigate whether breast milk composition differs in healthy control [HC] versus IBD mothers in terms of antibodies, cytokines, and metabolite,s to identify potential impact of IBD breast milk on neonatal immune system. METHODS: Breast milk specimens from HC [n = 17] and IBD [n = 31 for Crohn's disease [CD]; and n = 41 for ulcerative colitis [UC]; were collected at 3 and 6 months postpartum [PP3] and [PP6], respectively. Faecal samples were also collected. Cytokines and immunoglobulins [IgA/IgG/IgE] were analysed by multiplex Meso Scale Discovery [MSD] and commercial kits. Moreover, breast milk metabolites were analysed by 1H nuclear magnetic resonance [NMR]. RESULTS: We found that breast milk from IBD mothers showed significantly lower levels of IgA, sugar metabolite [lactose], and 2-aminobutyrate. In contrast, we observed that breast milk from mothers with IBD had increased levels of pro-inflammatory cytokines and higher energy metabolites [lactate and succinate] than milk from healthy mothers. In addition, we noticed that the type of treatment [5-aminosalicylic acid versus biologics] influenced the milk cytokines and metabolites profile. CONCLUSIONS: The reduction in immunoprotective components of IBD breast milk such as sIgA and lactose theoretically may modulate the potential protective effects of breastfeeding. On the other hand, presence of higher levels of pro-inflammatory cytokines, lactate, and succinate may predispose the offspring to an inflammatory condition or impact on the gut microbiome. Better understanding of the role of succinate in infants and its potential effects on microbiome or mucosal immunity merits further investigations.


Assuntos
Colite Ulcerativa/metabolismo , Doença de Crohn/metabolismo , Citocinas/metabolismo , Imunoglobulina A/metabolismo , Metaboloma , Leite Humano/metabolismo , Aminobutiratos/metabolismo , Anti-Inflamatórios não Esteroides/uso terapêutico , Produtos Biológicos/uso terapêutico , Estudos de Casos e Controles , Colite Ulcerativa/tratamento farmacológico , Doença de Crohn/tratamento farmacológico , Citocinas/efeitos dos fármacos , Fezes/química , Feminino , Voluntários Saudáveis , Humanos , Lactente , Recém-Nascido , Ácido Láctico/metabolismo , Lactose/metabolismo , Complexo Antígeno L1 Leucocitário/análise , Mesalamina/uso terapêutico , Metaboloma/efeitos dos fármacos , Período Pós-Parto , Ácido Succínico/metabolismo , Fatores de Tempo
6.
Sci Rep ; 7: 46616, 2017 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-28492284

RESUMO

Maternal separation (MS) in neonates can lead to intestinal injury. MS in neonatal mice disrupts mucosal morphology, induces colonic inflammation and increases trans-cellular permeability. Several studies indicate that intestinal epithelial stem cells are capable of initiating gut repair in a variety of injury models but have not been reported in MS. The pathophysiology of MS-induced gut injury and subsequent repair remains unclear, but communication between the brain and gut contribute to MS-induced colonic injury. Corticotropin-releasing hormone (CRH) is one of the mediators involved in the brain-gut axis response to MS-induced damage. We investigated the roles of the CRH receptors, CRHR1 and CRHR2, in MS-induced intestinal injury and subsequent repair. To distinguish their specific roles in mucosal injury, we selectively blocked CRHR1 and CRHR2 with pharmacological antagonists. Our results show that in response to MS, CRHR1 mediates gut injury by promoting intestinal inflammation, increasing gut permeability, altering intestinal morphology, and modulating the intestinal microbiota. In contrast, CRHR2 activates intestinal stem cells and is important for gut repair. Thus, selectively blocking CRHR1 and promoting CRHR2 activity could prevent the development of intestinal injuries and enhance repair in the neonatal period when there is increased risk of intestinal injury such as necrotizing enterocolitis.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Mucosa Intestinal , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Animais , Animais Recém-Nascidos , Colo/lesões , Colo/metabolismo , Colo/patologia , Mucosa Intestinal/lesões , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/biossíntese
7.
Adv Nutr ; 7(5): 928-37, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27633108

RESUMO

Necrotizing enterocolitis (NEC) is a devastating intestinal disease in preterm infants characterized by barrier disruption, intestinal microbial dysbiosis, and persistent inflammation of the colon, which results in high mortality rates. Current strategies used to manage this disease are not sufficient, although the use of human breast milk reduces the risk of NEC. Mother's milk is regarded as a fundamental nutritional source for neonates, but pasteurization of donor breast milk affects the composition of bioactive compounds. Current research is evaluating the benefits and potential pitfalls of adding probiotics and prebiotics to pasteurized milk so as to improve the functionality of the milk and thereby reduce the burden of illness caused by NEC. Probiotics (live micro-organisms that confer health to the host) and prebiotics (nondigestible oligosaccharides that stimulate the growth of healthy bacteria) are functional foods known to mediate immune responses and modulate microbial populations in the gut. Clinical research shows strain- and compound-specific responses when probiotics or prebiotics are administered in conjunction with donor breast milk for the prevention of NEC. Despite ongoing controversy surrounding optimal treatment strategies, randomized controlled studies are now investigating the use of synbiotics to reduce the incidence and severity of NEC. Synbiotics, a combination of probiotics and prebiotics, have been proposed to enhance beneficial health effects in the intestinal tract more than either agent administered alone. This review considers the implications of using probiotic-, prebiotic-, and synbiotic-supplemented breast milk as a strategy to prevent NEC and issues that could be encountered with the preparations.


Assuntos
Enterocolite Necrosante/prevenção & controle , Leite Humano , Prebióticos , Probióticos , Simbióticos , Bactérias , Humanos , Leite Humano/química , Leite Humano/microbiologia , Neonatologia , Oligossacarídeos
8.
Nat Commun ; 5: 3326, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24561718

RESUMO

The segmentation motor activity of the gut that facilitates absorption of nutrients was first described in the late 19th century, but the fundamental mechanisms underlying it remain poorly understood. The dominant theory suggests alternate excitation and inhibition from the enteric nervous system. Here we demonstrate that typical segmentation can occur after total nerve blockade. The segmentation motor pattern emerges when the amplitude of the dominant pacemaker, the slow wave generated by interstitial cells of Cajal associated with the myenteric plexus (ICC-MP), is modulated by the phase of induced lower frequency rhythmic transient depolarizations, generated by ICC associated with the deep muscular plexus (ICC-DMP), resulting in a waxing and waning of the amplitude of the slow wave and a rhythmic checkered pattern of segmentation motor activity. Phase-amplitude modulation of the slow waves points to an underlying system of coupled nonlinear oscillators originating in the networks of ICC.


Assuntos
Intestinos/fisiologia , Atividade Motora/fisiologia , Animais , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/fisiologia , Feminino , Células Intersticiais de Cajal/metabolismo , Células Intersticiais de Cajal/fisiologia , Mucosa Intestinal/metabolismo , Camundongos , Plexo Mientérico/metabolismo , Plexo Mientérico/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA