Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Sci Immunol ; 7(75): eabk2541, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-36054336

RESUMO

Interactions between the mammalian host and commensal microbiota are enforced through a range of immune responses that confer metabolic benefits and promote tissue health and homeostasis. Immunoglobulin A (IgA) responses directly determine the composition of commensal species that colonize the intestinal tract but require substantial metabolic resources to fuel antibody production by tissue-resident plasma cells. Here, we demonstrate that IgA responses are subject to diurnal regulation over the course of a circadian day. Specifically, the magnitude of IgA secretion, as well as the transcriptome of intestinal IgA+ plasma cells, was found to exhibit rhythmicity. Oscillatory IgA responses were found to be entrained by time of feeding and were also found to be in part coordinated by the plasma cell-intrinsic circadian clock via deletion of the master clock gene Arntl. Moreover, reciprocal interactions between the host and microbiota dictated oscillatory dynamics among the commensal microbial community and its associated transcriptional and metabolic activity in an IgA-dependent manner. Together, our findings suggest that circadian networks comprising intestinal IgA, diet, and the microbiota converge to align circadian biology in the intestinal tract and to ensure host-microbial mutualism.


Assuntos
Microbiota , Simbiose , Animais , Imunoglobulina A Secretora , Intestinos , Mamíferos , Periodicidade
2.
Immunology ; 166(1): 138-152, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35199335

RESUMO

Intestinal macrophages play a vital role in the maintenance of gut homeostasis through signals derived from the microbiota. We previously demonstrated that microbial-derived metabolites can shape the metabolic functions of macrophages. Here, we show that antibiotic-induced disruption of the intestinal microbiota dramatically alters both the local metabolite environment and the metabolic functions of macrophages in the colon. Broad-spectrum antibiotic administration in mice increased the expression of the large neutral amino acid transporter LAT1 and accordingly, amino acid uptake. Subsequently, antibiotic administration enhanced the metabolic functions of colonic macrophages, increasing phosphorylation of components of mammalian/mechanistic target of rapamycin signalling pathways, with increased expression of genes involved in glycolysis and oxidative phosphorylation (OXPHOS), increased mitochondrial function, increased rate of extracellular acidification (ECAR; measure of glycolysis) and increased rate of oxygen consumption (OCR; measure of OXPHOS). Small bowel macrophages were less metabolically active than their colonic counterparts, with macrophage metabolism in the small intestine being independent of the microbiota. Finally, we reveal tissue-resident Tim4+  CD4+ macrophages exhibit enhanced fatty acid uptake alongside reduced fatty acid synthesis compared to recruited macrophages. Thus, the microbiota shapes gut macrophage metabolism in a compartment-specific manner, with important implications for monocyte recruitment and macrophage differentiation.


Assuntos
Microbioma Gastrointestinal , Macrófagos , Animais , Antibacterianos/farmacologia , Colo , Ácidos Graxos/metabolismo , Macrófagos/metabolismo , Mamíferos , Camundongos
3.
Forensic Sci Int Synerg ; 3: 100164, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34527895

RESUMO

This is the second paper on the development and implementation of a universal experimental protocol for transfer and persistence of trace evidence. Here, we present the results of five individual researchers who implemented the universal experimental protocol for the first time. Over 2500 images were collected, computationally analysed and statistically compared. The results were shown to be reliable and consistent under all conditions tested and were used to model the rate of loss of transferred particles over a 7-day timescale. The protocol was additionally extended to include a test of camera settings. The protocol was found to be useable and robust in this preliminary trial paving the way for it to be deployed more widely.

4.
Parasitology ; : 1-8, 2021 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-34075861

RESUMO

Parasitic worms are amongst the most common pathogens to infect humans and have a long-established history of inflicting disease in their hosts. There is a large body of evidence that states intestine-dwelling helminths ensure their survival by influencing the host immune response against them. In recent years, it has become apparent that the large and diverse microbial communities that exist in the gastrointestinal (GI) tract of the host and within the parasite itself have a pivotal role in worm survival and persistence. Using a variety of mouse models (including laboratory, germ-free and rewilded mice), there have been new insights into how bacteria and worms interact with each other; this includes the discovery that Trichuris is unable to hatch and/or infect their host in the absence of bacteria, and that these worms contain a Trichuris-specific gut microbiota. These interactions are determined in part by the capacity of the host, gut microbiota and worms to communicate via metabolites such as butyrate, which are microbially derived and have known immunoregulatory properties. By exploring the contribution of gut bacteria to worm infections and the intricate relationship that exists between them, an exciting and emerging field in whipworm parasitology is established.

5.
Cell Rep Med ; 1(5): 100077, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32904427

RESUMO

Supplementation with members of the early-life microbiota as "probiotics" is increasingly used in attempts to beneficially manipulate the preterm infant gut microbiota. We performed a large observational longitudinal study comprising two preterm groups: 101 infants orally supplemented with Bifidobacterium and Lactobacillus (Bif/Lacto) and 133 infants non-supplemented (control) matched by age, sex, and delivery method. 16S rRNA gene profiling on fecal samples (n = 592) showed a predominance of Bifidobacterium and a lower abundance of pathobionts in the Bif/Lacto group. Metabolomic analysis showed higher fecal acetate and lactate and a lower fecal pH in the Bif/Lacto group compared to the control group. Fecal acetate positively correlated with relative abundance of Bifidobacterium, consistent with the ability of the supplemented Bifidobacterium strain to metabolize human milk oligosaccharides into acetate. This study demonstrates that microbiota supplementation is associated with a Bifidobacterium-dominated preterm microbiota and gastrointestinal environment more closely resembling that of full-term infants.


Assuntos
Bifidobacterium/fisiologia , Microbioma Gastrointestinal/fisiologia , Recém-Nascido Prematuro/metabolismo , Recém-Nascido Prematuro/fisiologia , Lactobacillus/fisiologia , Metaboloma/fisiologia , Bifidobacterium/genética , Aleitamento Materno/métodos , Suplementos Nutricionais/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal/genética , Humanos , Lactente , Recém-Nascido , Lactobacillus/genética , Estudos Longitudinais , Leite Humano/microbiologia , Probióticos/administração & dosagem , RNA Ribossômico 16S/genética
6.
Nature ; 584(7820): 274-278, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32760003

RESUMO

Colonization by the microbiota causes a marked stimulation of B cells and induction of immunoglobulin, but mammals colonized with many taxa have highly complex and individualized immunoglobulin repertoires1,2. Here we use a simplified model of defined transient exposures to different microbial taxa in germ-free mice3 to deconstruct how the microbiota shapes the B cell pool and its functional responsiveness. We followed the development of the immunoglobulin repertoire in B cell populations, as well as single cells by deep sequencing. Microbial exposures at the intestinal mucosa generated oligoclonal responses that differed from those of germ-free mice, and from the diverse repertoire that was generated after intravenous systemic exposure to microbiota. The IgA repertoire-predominantly to cell-surface antigens-did not expand after dose escalation, whereas increased systemic exposure broadened the IgG repertoire to both microbial cytoplasmic and cell-surface antigens. These microbial exposures induced characteristic immunoglobulin heavy-chain repertoires in B cells, mainly at memory and plasma cell stages. Whereas sequential systemic exposure to different microbial taxa diversified the IgG repertoire and facilitated alternative specific responses, sequential mucosal exposure produced limited overlapping repertoires and the attrition of initial IgA binding specificities. This shows a contrast between a flexible response to systemic exposure with the need to avoid fatal sepsis, and a restricted response to mucosal exposure that reflects the generic nature of host-microbial mutualism in the mucosa.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Imunidade nas Mucosas/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Simbiose/imunologia , Administração Intravenosa , Administração Oral , Animais , Clostridiales/imunologia , Clostridiales/isolamento & purificação , Escherichia coli/imunologia , Escherichia coli/isolamento & purificação , Feminino , Vida Livre de Germes , Imunoglobulina A/química , Imunoglobulina A/imunologia , Imunoglobulina G/química , Imunoglobulina G/imunologia , Cadeias Pesadas de Imunoglobulinas/imunologia , Memória Imunológica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/citologia , Plasmócitos/imunologia , Priming de Repetição
7.
Nutrients ; 12(4)2020 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-32235410

RESUMO

BACKGROUND: Bifidobacterium represents an important early life microbiota member. Specific bifidobacterial components, exopolysaccharides (EPS), positively modulate host responses, with purified EPS also suggested to impact microbe-microbe interactions by acting as a nutrient substrate. Thus, we determined the longitudinal effects of bifidobacterial EPS on microbial communities and metabolite profiles using an infant model colon system. METHODS: Differential gene expression and growth characteristics were determined for each strain; Bifidobacterium breve UCC2003 and corresponding isogenic EPS-deletion mutant (B. breve UCC2003del). Model colon vessels were inoculated with B. breve and microbiome dynamics monitored using 16S rRNA sequencing and metabolomics (NMR). RESULTS: Transcriptomics of EPS mutant vs. B. breve UCC2003 highlighted discrete differential gene expression (e.g., eps biosynthetic cluster), though overall growth dynamics between strains were unaffected. The EPS-positive vessel had significant shifts in microbiome and metabolite profiles until study end (405 h); with increases of Tyzzerella and Faecalibacterium, and short-chain fatty acids, with further correlations between taxa and metabolites which were not observed within the EPS-negative vessel. CONCLUSIONS: These data indicate that B. breve UCC2003 EPS is potentially metabolized by infant microbiota members, leading to differential microbial metabolism and altered metabolite by-products. Overall, these findings may allow development of EPS-specific strategies to promote infant health.


Assuntos
Bifidobacterium breve/genética , Bifidobacterium breve/fisiologia , Colo/metabolismo , Colo/microbiologia , Suplementos Nutricionais , Microbioma Gastrointestinal/fisiologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Saúde do Lactente , Polissacarídeos Bacterianos/genética , Polissacarídeos Bacterianos/metabolismo , Bifidobacterium breve/crescimento & desenvolvimento , Expressão Gênica , Humanos , Lactente , Mutação , RNA Ribossômico 16S/genética
8.
ISME J ; 14(2): 635-648, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31740752

RESUMO

Diet-microbe interactions play an important role in modulating the early-life microbiota, with Bifidobacterium strains and species dominating the gut of breast-fed infants. Here, we sought to explore how infant diet drives distinct bifidobacterial community composition and dynamics within individual infant ecosystems. Genomic characterisation of 19 strains isolated from breast-fed infants revealed a diverse genomic architecture enriched in carbohydrate metabolism genes, which was distinct to each strain, but collectively formed a pangenome across infants. Presence of gene clusters implicated in digestion of human milk oligosaccharides (HMOs) varied between species, with growth studies indicating that within single infants there were differences in the ability to utilise 2'FL and LNnT HMOs between strains. Cross-feeding experiments were performed with HMO degraders and non-HMO users (using spent or 'conditioned' media and direct co-culture). Further 1H-NMR analysis identified fucose, galactose, acetate, and N-acetylglucosamine as key by-products of HMO metabolism; as demonstrated by modest growth of non-HMO users on spend media from HMO metabolism. These experiments indicate how HMO metabolism permits the sharing of resources to maximise nutrient consumption from the diet and highlights the cooperative nature of bifidobacterial strains and their role as 'foundation' species in the infant ecosystem. The intra- and inter-infant bifidobacterial community behaviour may contribute to the diversity and dominance of Bifidobacterium in early life and suggests avenues for future development of new diet and microbiota-based therapies to promote infant health.


Assuntos
Bifidobacterium , Metabolismo dos Carboidratos/genética , Leite Humano , Oligossacarídeos/genética , Bifidobacterium/genética , Bifidobacterium/isolamento & purificação , Bifidobacterium/fisiologia , Aleitamento Materno , Ecossistema , Feminino , Genes Bacterianos , Variação Genética , Genoma Bacteriano , Humanos , Lactente , Metagenoma/genética , Metagenoma/fisiologia , Interações Microbianas , Microbiota , Leite Humano/química , Oligossacarídeos/metabolismo
9.
MedEdPublish (2016) ; 8: 221, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-38089297

RESUMO

This article was migrated. The article was marked as recommended. Introduction: Formal mentoring programs are a professional development approach to help junior faculty develop an academic medicine career. This study investigated the perceptions of mentors versus mentees in formal career mentoring partnerships across multiple institutions. Methods: The authors implemented departmental mentoring programs for junior faculty at four academic medical centers. They collected post-program data from mentors and mentees in order to examine the predictors of mentoring satisfaction, mentee outcomes, and work-related variables. Results: The pattern of relationships between the variables differed for mentors versus mentees. Mentoring focus, mentor accessibility and mentee initiative predicted partnership satisfaction and mentee progress. Partnerships that used a mentoring agreement reported greater progress and satisfaction. There were some relationships between partnership outcomes and work-related outcomes. While partnership satisfaction predicted job and administrative/leadership satisfaction for mentors, it predicted positive perceptions of the department's mentoring culture and professional development opportunities for mentees. Conclusions: The study identified unique antecedents and consequences of mentoring partnership satisfaction and mentee outcomes. The varying perspectives of mentors versus mentees indicated a need to clearly communicate partnership expectations and desired outcomes. Overall, the positive impact of formal mentoring programs on partnership and work-related outcomes was supported with implications for future programs and research.

10.
Sci Transl Med ; 10(464)2018 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-30355800

RESUMO

Macrophages in the healthy intestine are highly specialized and usually respond to the gut microbiota without provoking an inflammatory response. A breakdown in this tolerance leads to inflammatory bowel disease (IBD), but the mechanisms by which intestinal macrophages normally become conditioned to promote microbial tolerance are unclear. Strong epidemiological evidence linking disruption of the gut microbiota by antibiotic use early in life to IBD indicates an important role for the gut microbiota in modulating intestinal immunity. Here, we show that antibiotic use causes intestinal macrophages to become hyperresponsive to bacterial stimulation, producing excess inflammatory cytokines. Re-exposure of antibiotic-treated mice to conventional microbiota induced a long-term, macrophage-dependent increase in inflammatory T helper 1 (TH1) responses in the colon and sustained dysbiosis. The consequences of this dysregulated macrophage activity for T cell function were demonstrated by increased susceptibility to infections requiring TH17 and TH2 responses for clearance (bacterial Citrobacter rodentium and helminth Trichuris muris infections), corresponding with increased inflammation. Short-chain fatty acids (SCFAs) were depleted during antibiotic administration; supplementation of antibiotics with the SCFA butyrate restored the characteristic hyporesponsiveness of intestinal macrophages and prevented T cell dysfunction. Butyrate altered the metabolic behavior of macrophages to increase oxidative phosphorylation and also promoted alternative macrophage activation. In summary, the gut microbiota is essential to maintain macrophage-dependent intestinal immune homeostasis, mediated by SCFA-dependent pathways. Oral antibiotics disrupt this process to promote sustained T cell-mediated dysfunction and increased susceptibility to infections, highlighting important implications of repeated broad-spectrum antibiotic use.


Assuntos
Antibacterianos/farmacologia , Homeostase/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Intestinos/citologia , Macrófagos/metabolismo , Linfócitos T/imunologia , Animais , Butiratos/farmacologia , Citocinas/metabolismo , Ácidos Graxos/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Receptores CCR2/metabolismo , Linfócitos T/efeitos dos fármacos , Células Th1/efeitos dos fármacos
11.
Immunity ; 49(3): 545-559.e5, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30193848

RESUMO

Although the mammalian microbiota is well contained within the intestine, it profoundly shapes development and metabolism of almost every host organ. We questioned the range and depth of microbial metabolite penetration into the host, and how this is modulated by intestinal immunity. Chemically identical microbial and host metabolites were distinguished by stable isotope tracing from 13C-labeled live non-replicating Escherichia coli, differentiating 12C host isotopes with high-resolution mass spectrometry. Hundreds of endogenous microbial compounds penetrated 23 host tissues and fluids after intestinal exposure: subsequent 12C host metabolome signatures included lipidemia, reduced glycolysis, and inflammation. Penetrant bacterial metabolites from the small intestine were rapidly cleared into the urine, whereas induced antibodies curtailed microbial metabolite exposure by accelerating intestinal bacterial transit into the colon where metabolite transport mechanisms are limiting. Pervasive penetration of microbial molecules can cause extensive host tissue responses: these are limited by immune and non-immune intestinal mucosal adaptations to the microbiota.


Assuntos
Anticorpos/metabolismo , Microbioma Gastrointestinal/fisiologia , Glicólise/imunologia , Hiperlipidemias/imunologia , Inflamação/imunologia , Mamíferos/imunologia , Animais , Anticorpos/imunologia , Radioisótopos de Carbono/análise , Interações Hospedeiro-Patógeno , Imunidade , Cadeias Pesadas de Imunoglobulinas/genética , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
Environ Microbiol ; 20(7): 2337-2353, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28892253

RESUMO

The gastrointestinal tract is a highly complex organ in which multiple dynamic physiological processes are tightly coordinated while interacting with a dense and extremely diverse microbial population. From establishment in early life, through to host-microbe symbiosis in adulthood, the gut microbiota plays a vital role in our development and health. The effect of the microbiota on gut development and physiology is highlighted by anatomical and functional changes in germ-free mice, affecting the gut epithelium, immune system and enteric nervous system. Microbial colonisation promotes competent innate and acquired mucosal immune systems, epithelial renewal, barrier integrity, and mucosal vascularisation and innervation. Interacting or shared signalling pathways across different physiological systems of the gut could explain how all these changes are coordinated during postnatal colonisation, or after the introduction of microbiota into germ-free models. The application of cell-based in-vitro experimental systems and mathematical modelling can shed light on the molecular and signalling pathways which regulate the development and maintenance of homeostasis in the gut and beyond.


Assuntos
Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos , Animais , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Homeostase , Humanos , Transdução de Sinais , Simbiose
13.
Immunology ; 149(3): 262-269, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27441396

RESUMO

Tertiary lymphoid follicles (TLFs) can develop in the respiratory tract in response to infections or chronic inflammation. However, their functional relevance remains unclear because they are implicated in both protective and pathological responses. In contrast to homeostatic conditions, external antigens and damage to the lung tissue may drive TLF formation in inflamed lungs, and once established, the presence of pulmonary TLFs may signal the progression of chronic lung disease. This novel concept will be discussed in light of recent work in chronic obstructive pulmonary disease and how changes in the pulmonary microbiota may drive and direct TLF formation and function. We will also discuss the cellularity of TLFs at the pulmonary mucosa, with emphasis on the potential roles of lymphoid tissue inducer cells, and B- and T-cell aggregates, and will examine the function of key chemokines and cytokines including CXCL13 and interleukin-17, in the formation and maintenance of pulmonary TLFs.


Assuntos
Linfócitos B/imunologia , Microbiota/imunologia , Pneumonia/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Mucosa Respiratória/imunologia , Linfócitos T/imunologia , Estruturas Linfoides Terciárias/imunologia , Animais , Quimiocina CXCL13/metabolismo , Doença Crônica , Humanos , Interleucina-17/metabolismo , Mucosa Respiratória/microbiologia , Estruturas Linfoides Terciárias/microbiologia
14.
Nat Commun ; 6: 8292, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26392213

RESUMO

The overall composition of the mammalian intestinal microbiota varies between individuals: within each individual there are differences along the length of the intestinal tract related to host nutrition, intestinal motility and secretions. Mucus is a highly regenerative protective lubricant glycoprotein sheet secreted by host intestinal goblet cells; the inner mucus layer is nearly sterile. Here we show that the outer mucus of the large intestine forms a unique microbial niche with distinct communities, including bacteria without specialized mucolytic capability. Bacterial species present in the mucus show differential proliferation and resource utilization compared with the same species in the intestinal lumen, with high recovery of bioavailable iron and consumption of epithelial-derived carbon sources according to their genome-encoded metabolic repertoire. Functional competition for existence in this intimate layer is likely to be a major determinant of microbiota composition and microbial molecular exchange with the host.


Assuntos
Bactérias/classificação , Bactérias/isolamento & purificação , Mucosa Intestinal/microbiologia , Animais , Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Vida Livre de Germes , Ferro/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Família Multigênica , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Transcriptoma
16.
Sci Transl Med ; 6(237): 237ra66, 2014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24848256

RESUMO

A prerequisite for establishment of mutualism between the host and the microbial community that inhabits the large intestine is the stringent mucosal compartmentalization of microorganisms. Microbe-loaded dendritic cells trafficking through lymphatics are arrested at the mesenteric lymph nodes, which constitute the firewall of the intestinal lymphatic circulation. We show in different mouse models that the liver, which receives the intestinal venous blood circulation, forms a vascular firewall that captures gut commensal bacteria entering the bloodstream during intestinal pathology. Phagocytic Kupffer cells in the liver of mice clear commensals from the systemic vasculature independently of the spleen through the liver's own arterial supply. Damage to the liver firewall in mice impairs functional clearance of commensals from blood, despite heightened innate immunity, resulting in spontaneous priming of nonmucosal immune responses through increased systemic exposure to gut commensals. Systemic immune responses consistent with increased extraintestinal commensal exposure were found in humans with liver disease (nonalcoholic steatohepatitis). The liver may act as a functional vascular firewall that clears commensals that have penetrated either intestinal or systemic vascular circuits.


Assuntos
Translocação Bacteriana , Interações Hospedeiro-Patógeno , Intestinos/irrigação sanguínea , Intestinos/microbiologia , Circulação Hepática , Hepatopatias/microbiologia , Fígado/irrigação sanguínea , Fígado/microbiologia , Adulto , Idoso , Animais , Carga Bacteriana , Modelos Animais de Doenças , Fígado Gorduroso/imunologia , Fígado Gorduroso/microbiologia , Fígado Gorduroso/fisiopatologia , Fezes/microbiologia , Feminino , Humanos , Imunidade Inata , Imunidade nas Mucosas , Intestinos/imunologia , Células de Kupffer/microbiologia , Fígado/imunologia , Fígado/patologia , Hepatopatias/imunologia , Hepatopatias/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica , Estudos Retrospectivos , Fatores de Tempo
18.
J Acad Nutr Diet ; 112(8): 1247-52, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22818732

RESUMO

When individuals with a suspected or diagnosed eating disorder adopt a vegetarian diet, health care professionals might worry that this choice could function as a socially acceptable way to legitimize food avoidance. Yet only limited research has examined vegetarianism in relation to eating disorders. Our study objectives were to compare individuals with and without an eating disorder history and individuals at different stages of eating disorder recovery on past and current vegetarianism and motivations for and age at becoming vegetarian. Participants were females seen at some point for an eating disorder (n=93) and controls who never had an eating disorder (n=67). Recruitment and data collection for this cross-sectional study occurred in 2007-2008. χ(2) analyses and analyses of variance and covariance were used to examine the research questions. Compared with controls, individuals with an eating disorder history were considerably more likely to ever have been vegetarian (52% vs 12%; P<0.001), to be currently vegetarian (24% vs 6%; P<0.01), and to be primarily motivated by weight-related reasons (42% vs 0%; P<0.05). The three recovery status groups (fully recovered, partially recovered, and active eating disorder) did not differ significantly in percentiles endorsing a history of vegetarianism or weight-related reasons as primary, but they differed significantly in current vegetarianism (33% of active cases, 13% of partially recovered, 5% of fully recovered; P<0.05). Most perceived that their vegetarianism was related to their eating disorder (68%) and emerged after its onset. Results shed light on the vegetarianism-eating disorders relation and suggest intervention considerations for clinicians (eg, investigating motives for vegetarianism).


Assuntos
Comportamento de Escolha , Dieta Vegetariana/psicologia , Transtornos da Alimentação e da Ingestão de Alimentos/psicologia , Comportamentos Relacionados com a Saúde , Motivação , Adolescente , Análise de Variância , Índice de Massa Corporal , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , Estudos Transversais , Transtornos da Alimentação e da Ingestão de Alimentos/epidemiologia , Transtornos da Alimentação e da Ingestão de Alimentos/terapia , Feminino , Humanos , Fenômenos Fisiológicos da Nutrição/fisiologia , Estado Nutricional , Adulto Jovem
19.
J Clin Invest ; 121(10): 3991-4002, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21881212

RESUMO

B cell activation factor of the TNF family (BAFF) is a potent B cell survival factor. BAFF overexpressing transgenic mice (BAFF-Tg mice) exhibit features of autoimmune disease, including B cell hyperplasia and hypergammaglobulinemia, and develop fatal nephritis with age. However, basal serum IgA levels are also elevated, suggesting that the pathology in these mice may be more complex than initially appreciated. Consistent with this, we demonstrate here that BAFF-Tg mice have mesangial deposits of IgA along with high circulating levels of polymeric IgA that is aberrantly glycosylated. Renal disease in BAFF-Tg mice was associated with IgA, because serum IgA was highly elevated in nephritic mice and BAFF-Tg mice with genetic deletion of IgA exhibited less renal pathology. The presence of commensal flora was essential for the elevated serum IgA phenotype, and, unexpectedly, commensal bacteria-reactive IgA antibodies were found in the blood. These data illustrate how excess B cell survival signaling perturbs the normal balance with the microbiota, leading to a breach in the normal mucosal-peripheral compartmentalization. Such breaches may predispose the nonmucosal system to certain immune diseases. Indeed, we found that a subset of patients with IgA nephropathy had elevated serum levels of a proliferation inducing ligand (APRIL), a cytokine related to BAFF. These parallels between BAFF-Tg mice and human IgA nephropathy may provide a new framework to explore connections between mucosal environments and renal pathology.


Assuntos
Fator Ativador de Células B/genética , Fator Ativador de Células B/imunologia , Glomerulonefrite por IGA/etiologia , Animais , Anticorpos Antinucleares/sangue , Anticorpos Antibacterianos/sangue , Fator Ativador de Células B/sangue , Proteínas de Ligação a DNA/sangue , Modelos Animais de Doenças , Feminino , Expressão Gênica , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/imunologia , Glomerulonefrite por IGA/patologia , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Rim/imunologia , Rim/patologia , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Transcrição/sangue
20.
Immunity ; 34(5): 794-806, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21596591

RESUMO

Mammals harbor a dense commensal microbiota in the colon. Regulatory T (Treg) cells are known to limit microbe-triggered intestinal inflammation and the CD4+ T cell compartment is shaped by the presence of particular microbes or bacterial compounds. It is, however, difficult to distinguish whether these effects reflect true mutualistic immune adaptation to intestinal colonization or rather idiosyncratic immune responses. To investigate truly mutualistic CD4+ T cell adaptation, we used the altered Schaedler flora (ASF). Intestinal colonization resulted in activation and de novo generation of colonic Treg cells. Failure to activate Treg cells resulted in the induction of T helper 17 (Th17) and Th1 cell responses, which was reversed by wild-type Treg cells. Efficient Treg cell induction was also required to maintain intestinal homeostasis upon dextran sulfate sodium-mediated damage in the colon. Thus, microbiota colonization-induced Treg cell responses are a fundamental intrinsic mechanism to induce and maintain host-intestinal microbial T cell mutualism.


Assuntos
Imunidade Adaptativa , Colo/imunologia , Colo/microbiologia , Linfócitos T Reguladores/imunologia , Animais , Proliferação de Células , Colo/citologia , Homeostase , Imunidade nas Mucosas , Interleucina-10/imunologia , Ativação Linfocitária , Camundongos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Linfócitos T Reguladores/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...