Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Eur J Nutr ; 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38662018

RESUMEN

PURPOSE: Impaired gut barrier function is associated with systemic inflammation and many chronic diseases. Undigested dietary proteins are fermented in the colon by the gut microbiota which produces nitrogenous metabolites shown to reduce barrier function in vitro. With growing evidence of sex-based differences in gut microbiotas, we determined whether there were sex by dietary protein interactions which could differentially impact barrier function via microbiota modification. METHODS: Fermentation systems were inoculated with faeces from healthy males (n = 5) and females (n = 5) and supplemented with 0.9 g of non-hydrolysed proteins sourced from whey, fish, milk, soya, egg, pea, or mycoprotein. Microbial populations were quantified using fluorescence in situ hybridisation with flow cytometry. Metabolite concentrations were analysed using gas chromatography, solid phase microextraction coupled with gas chromatography-mass spectrometry and ELISA. RESULTS: Increased protein availability resulted in increased proteolytic Bacteroides spp (p < 0.01) and Clostridium coccoides (p < 0.01), along with increased phenol (p < 0.01), p-cresol (p < 0.01), indole (p = 0.018) and ammonia (p < 0.01), varying by protein type. Counts of Clostridium cluster IX (p = 0.03) and concentration of p-cresol (p = 0.025) increased in males, while females produced more ammonia (p = 0.02), irrespective of protein type. Further, we observed significant sex-protein interactions affecting bacterial populations and metabolites (p < 0.005). CONCLUSIONS: Our findings suggest that protein fermentation by the gut microbiota in vitro is influenced by both protein source and the donor's sex. Should these results be confirmed through human studies, they could have major implications for developing dietary recommendations tailored by sex to prevent chronic illnesses.

2.
Front Nutr ; 9: 927754, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36267902

RESUMEN

Iron deficiency is the most prevalent human micronutrient deficiency, disrupting the physiological development of millions of infants and children. Oral iron supplementation is used to address iron-deficiency anemia and reduce associated stunting but can promote infection risk since restriction of iron availability serves as an innate immune mechanism against invading pathogens. Raised iron availability is associated with an increase in enteric pathogens, especially Enterobacteriaceae species, accompanied by reductions in beneficial bacteria such as Bifidobacteria and lactobacilli and may skew the pattern of gut microbiota development. Since the gut microbiota is the primary driver of immune development, deviations from normal patterns of bacterial succession in early life can have long-term implications for immune functionality. There is a paucity of knowledge regarding how both iron deficiency and luminal iron availability affect gut microbiota development, or the subsequent impact on immunity, which are likely to be contributors to the increased risk of infection. Piglets are naturally iron deficient. This is largely due to their low iron endowments at birth (primarily due to large litter sizes), and their rapid growth combined with the low iron levels in sow milk. Thus, piglets consistently become iron deficient within days of birth which rapidly progresses to anemia in the absence of iron supplementation. Moreover, like humans, pigs are omnivorous and share many characteristics of human gut physiology, microbiota and immunity. In addition, their precocial nature permits early maternal separation, individual housing, and tight control of nutritional intake. Here, we highlight the advantages of piglets as valuable and highly relevant models for human infants in promoting understanding of how early iron status impacts physiological development. We also indicate how piglets offer potential to unravel the complexities of microbiota-immune responses during iron deficiency and in response to iron supplementation, and the link between these and increased risk of infectious disease.

3.
Med ; 2(2): 164-179.e12, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33665641

RESUMEN

BACKGROUND: How specific nutrients influence adaptive immunity is of broad interest. Iron deficiency is the most common micronutrient deficiency worldwide and imparts a significant burden of global disease; however, its effects on immunity remain unclear. METHODS: We used a hepcidin mimetic and several genetic models to examine the effect of low iron availability on T cells in vitro and on immune responses to vaccines and viral infection in mice. We examined humoral immunity in human patients with raised hepcidin and low serum iron caused by mutant TMPRSS6. We tested the effect of iron supplementation on vaccination-induced humoral immunity in piglets, a natural model of iron deficiency. FINDINGS: We show that low serum iron (hypoferremia), caused by increased hepcidin, severely impairs effector and memory responses to immunizations. The intensified metabolism of activated lymphocytes requires the support of enhanced iron acquisition, which is facilitated by IRP1/2 and TFRC. Accordingly, providing extra iron improved the response to vaccination in hypoferremic mice and piglets, while conversely, hypoferremic humans with chronically increased hepcidin have reduced concentrations of antibodies specific for certain pathogens. Imposing hypoferremia blunted the T cell, B cell, and neutralizing antibody responses to influenza virus infection in mice, allowing the virus to persist and exacerbating lung inflammation and morbidity. CONCLUSIONS: Hypoferremia, a well-conserved physiological innate response to infection, can counteract the development of adaptive immunity. This nutrient trade-off is relevant for understanding and improving immune responses to infections and vaccines in the globally common contexts of iron deficiency and inflammatory disorders. FUNDING: Medical Research Council, UK.


Asunto(s)
Deficiencias de Hierro , Trastornos del Metabolismo del Hierro , Animales , Hepcidinas/genética , Humanos , Inmunidad Humoral , Hierro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Porcinos , Vacunación
4.
Nutrients ; 12(12)2020 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-33327501

RESUMEN

Ferrous iron supplementation has been reported to adversely alter the gut microbiota in infants. To date, the impact of iron on the adult microbiota is limited, particularly at low supplementary concentrations. The aim of this research was to explore the impact of low-level iron supplementation on the gut microbiota of healthy and Irritable Bowel Syndrome (IBS) volunteers. Anaerobic, pH-controlled in vitro batch cultures were inoculated with faeces from healthy or IBS donors along with iron (ferrous sulphate, nanoparticulate iron and pea ferritin (50 µmol-1 iron)). The microbiota were explored by fluorescence in situ hybridisation coupled with flow cytometry. Furthermore, metabolite production was assessed by gas chromatography. IBS volunteers had different starting microbial profiles to healthy controls. The sources of iron did not negatively impact the microbial population, with results of pea ferritin supplementation being similar to nanoparticulate iron, whilst ferrous sulphate led to enhanced Bacteroides spp. The metabolite data suggested no shift to potentially negative proteolysis. The results indicate that low doses of iron from the three sources were not detrimental to the gut microbiota. This is the first time that pea ferritin fermentation has been tested and indicates that low dose supplementation of iron is unlikely to be detrimental to the gut microbiota.


Asunto(s)
Suplementos Dietéticos , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Hierro/farmacología , Síndrome del Colon Irritable/microbiología , Técnicas de Cultivo Celular por Lotes , Técnicas de Cultivo de Célula , Fermentación , Compuestos Ferrosos/farmacología , Humanos , Nanopartículas , Proteínas de Guisantes/farmacología , Proteolisis/efectos de los fármacos
5.
Jt Comm J Qual Patient Saf ; 46(12): 682-690, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32994132

RESUMEN

BACKGROUND: Most antibiotics are prescribed in outpatient settings, including urgent care clinics (UCCs); however, few UCCs have described implementing antibiotic stewardship. This study describes interventions to reduce total antibiotic and azithromycin use in a UCC network. METHODS: The researchers conducted a prospective performance improvement project in five UCCs in Delaware, with > 40 providers and > 75,000 visits annually. In April 2017 all providers received in-person education on guideline-recommended management of common infections. The UCC lead physician performed chart audits and provided group and individual feedback. Individual antibiotic utilization rates were provided beginning in February 2018, and chart audits ceased in May 2018. Patient education included posters in waiting and exam rooms, discharge materials, and external website revisions. The researchers used control charts to analyze trends in prescribing over time, and calculated rate ratios (RRs) between pre-/early, mid- and postintervention periods. RESULTS: Compared to the pre-/early intervention study period (54.7 prescriptions per 100 visits), total antibiotic use declined to 40.2 (RR, 0.74; 95% confidence interval [CI] = 0.72-0.75) in the mid-intervention period and to 35.0 (RR, 0.42; 95% CI = 0.40-0.44) in the postintervention period. Azithromycin use declined from 8.5 prescriptions/100 visits to 3.5 (RR 0.64; 95% CI = 0.63-0.65) and 1.9 (RR 0.22; 95% CI = 0.21-0.24), respectively. The control charts indicated decreasing mean antibiotic prescribing rates as well as decreased variability. CONCLUSION: A multifaceted and iterative approach significantly reduced prescribing of all antibiotics, including azithromycin, regardless of diagnosis. Although the approach was initially resource-intensive, sending prescribing data directly to providers automated the process without an observed rebound in prescribing.


Asunto(s)
Programas de Optimización del Uso de los Antimicrobianos , Instituciones de Atención Ambulatoria , Antibacterianos/uso terapéutico , Humanos , Pacientes Ambulatorios , Estudios Prospectivos
7.
Front Immunol ; 10: 2705, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921096

RESUMEN

Although sex disparity in immunological function and susceptibility to various inflammatory and infectious disease is recognized in adults, far less is known about the situation in young infants during immune development. We have used an outbred piglet model to explore potential early sex disparity underlying both mucosal immune development and systemic responses to novel antigen. Despite similarities in intestinal barrier function and therefore, presumably, antigen exposure, females had less CD172+ (Sirp-α) antigen presenting cells and expression of MHCIIDR at 28 days old compared to males, along with greater regulatory T-cell numbers. This suggests that, during infancy, females may have greater potential for local immune regulation than their male counterparts. However, females also presented with significantly greater systemic antibody responses to injected ovalbumin and dietary soya. Females also synthesized significantly more IgA in mesenteric lymph nodes, whereas males synthesized more in caecal mucosa, suggesting that plasma cells were retained within the MLN in females, but increased numbers of plasma cells circulated through to the mucosal tissue in males. Significant effects of inulin and Bifidobacterium lactis NCC2818 on the developing immune system were also sex-dependent. Our results may start to explain inconsistencies in outcomes of trials of functional foods in infants, as distinction between males and females is seldom made. Since later functionality of the immune system is highly dependent on appropriate development during infancy, stratifying nutritional interventions by sex may present a novel means of optimizing treatments and preventative strategies to reduce the risk of the development of immunological disorders in later life.


Asunto(s)
Dieta , Mucosa Intestinal/inmunología , Caracteres Sexuales , Animales , Animales Recién Nacidos , Inulina/inmunología , Inulina/farmacología , Probióticos , Porcinos
8.
Front Immunol ; 9: 1061, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29868021

RESUMEN

Epidemiological studies have demonstrated that exposure to farm environments during childhood can be linked to reductions in the incidence of immune disorders, but generating an appropriate model is difficult. 108 half-sibling piglets were born on either extensive (outdoor) or intensive (indoor) farms: at 1 day old, a subset of piglets from each litter were transferred to a high-hygiene isolator facility to create differences in rearing environment either during birth/first day or during the subsequent 56 days of life. Interactions between CD14, CD16, MHCIIDR, and capillary endothelium were assessed using four-color quantitative fluorescence immunohistology. Effects of birth and rearing environment on the antigen-presenting microenvironment of the proximal and distal jejunum (professional and stromal) were apparent at 5, 28, and 56 days after birth However, effects on CD4+CD25+Foxp3+ regulatory T-cells (Tregs) in the intestinal mucosa were apparent around weaning at 28 days but had disappeared by 56 days. These Tregs were reduced in the isolator piglets compared to their farm-reared siblings, but this effect was less marked in piglets born on the extensive farm and required administration of antibiotics. Our results suggest that there may be at least two windows of opportunity in which different farm environments were influencing immune development: one during the perinatal period (up to the first day of life), and one during later infancy. Furthermore, the differences on Tregs suggest that the effects of early life influences may be particularly critical around weaning.


Asunto(s)
Adaptación Fisiológica , Antibacterianos/farmacología , Exposición a Riesgos Ambientales , Granjas , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Animales , Animales Recién Nacidos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Biomarcadores , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunidad Mucosa/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Porcinos , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Destete
9.
Sci Rep ; 7(1): 5310, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28706260

RESUMEN

Inflammatory and metabolic diseases can originate during early-life and have been correlated with shifts in intestinal microbial ecology. Here we demonstrate that minor environmental fluctuations during the early neonatal period had sustained effects on the developing porcine microbiota and host-microbe interface. These inter-replicate effects appear to originate during the first day of life, and are likely to reflect very early microbiota acquisition from the environment. We statistically link early systemic inflammation with later local increases in inflammatory cytokine (IL-17) production, which could have important enteric health implications. Immunity, intestinal barrier function, host metabolism and host-microbiota co-metabolism were further modified by Bifidobacterium lactis NCC2818 supplementation, although composition of the in situ microbiota remained unchanged. Finally, our robust model identified novel, strong correlations between urinary metabolites (eg malonate, phenylacetylglycine, alanine) and mucosal immunoglobulin (IgM) and cytokine (IL-10, IL-4) production, thus providing the possibility of the development of urinary 'dipstick' tests to assess non-accessible mucosal immune development and identify early precursors (biomarkers) of disease. These results have important implications for infants exposed to neonatal factors including caesarean delivery, antibiotic therapy and delayed discharge from hospital environments, which may predispose to the development of inflammatory and metabolic diseases in later life.


Asunto(s)
Bifidobacterium animalis/crecimiento & desarrollo , Exposición a Riesgos Ambientales , Microbioma Gastrointestinal , Probióticos/administración & dosificación , Animales , Animales Recién Nacidos , Intervención Educativa Precoz , Inmunidad Mucosa , Enfermedades Metabólicas/prevención & control , Porcinos , Enfermedades de los Porcinos/prevención & control , Síndrome de Respuesta Inflamatoria Sistémica/prevención & control
11.
Pract Midwife ; 19(6): 33-5, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27451491

RESUMEN

This article explores the differences in perceptions of safety between midwives and women. The authors use their experience of providing midwife-led care in a local setting to discuss the impact that language and women's perceptions of safety have on the acceptance of midwife-led care.


Asunto(s)
Enfermeras Obstetrices , Seguridad del Paciente , Embarazo/psicología , Femenino , Humanos
12.
ISME J ; 10(1): 145-57, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26066712

RESUMEN

The postnatal environment, including factors such as weaning and acquisition of the gut microbiota, has been causally linked to the development of later immunological diseases such as allergy and autoimmunity, and has also been associated with a predisposition to metabolic disorders. We show that the very early-life environment influences the development of both the gut microbiota and host metabolic phenotype in a porcine model of human infants. Farm piglets were nursed by their mothers for 1 day, before removal to highly controlled, individual isolators where they received formula milk until weaning at 21 days. The experiment was repeated, to create two batches, which differed only in minor environmental fluctuations during the first day. At day 1 after birth, metabolic profiling of serum by (1)H nuclear magnetic resonance spectroscopy demonstrated significant, systemic, inter-batch variation which persisted until weaning. However, the urinary metabolic profiles demonstrated that significant inter-batch effects on 3-hydroxyisovalerate, trimethylamine-N-oxide and mannitol persisted beyond weaning to at least 35 days. Batch effects were linked to significant differences in the composition of colonic microbiota at 35 days, determined by 16 S pyrosequencing. Different weaning diets modulated both the microbiota and metabolic phenotype independently of the persistent batch effects. We demonstrate that the environment during the first day of life influences development of the microbiota and metabolic phenotype and thus should be taken into account when interrogating experimental outcomes. In addition, we suggest that intervention at this early time could provide 'metabolic rescue' for at-risk infants who have undergone aberrant patterns of initial intestinal colonisation.


Asunto(s)
Microbioma Gastrointestinal , Intestinos/microbiología , Porcinos/microbiología , Animales , Colon/crecimiento & desarrollo , Colon/metabolismo , Colon/microbiología , Femenino , Humanos , Mucosa Intestinal/metabolismo , Intestinos/crecimiento & desarrollo , Fenotipo , Porcinos/crecimiento & desarrollo , Porcinos/fisiología , Destete
13.
Midwives ; 19(Winter): 32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-30726608
14.
Br J Nutr ; 110(7): 1243-52, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23473077

RESUMEN

Weaning is associated with a major shift in the microbial community of the intestine, and this instability may make it more acquiescent than the adult microbiota to long-term changes. Modulation achieved through dietary interventions may have potentially beneficial effects on the developing immune system, which is driven primarily by the microbiota. The specific aim of the present study was to determine whether immune development could be modified by dietary supplementation with the human probiotic Bifidobacterium lactis NCC2818 in a tractable model of weaning in infants. Piglets were reared by their mothers before being weaned onto a solid diet supplemented with B. lactis NCC2818, while sibling controls did not receive supplementation. Probiotic supplementation resulted in a reduction in IgA (P<0·0005) and IgM (P<0·009) production by mucosal tissues but had no effect on IgG production (P>0·05). Probiotic-supplemented pigs had more mast cells than unsupplemented littermates (P<0·0001), although numbers in both groups were low. In addition, the supplemented piglets made stronger serum IgG responses to fed and injected antigens (P<0·05). The present findings are consistent with B. lactis NCC2818 reducing intestinal permeability induced by weaning, and suggest that the piglet is a valuable intermediate between rodent models and human infants. The results also strongly suggest that measures of the effect of probiotic supplementation on the immune system need to be interpreted carefully as proxy measures of health benefit. However, they are useful in developing an understanding of the mechanism of action of probiotic strains, an important factor in predicting favourable health outcomes of nutritional intervention.


Asunto(s)
Bifidobacterium , Sistema Inmunológico/crecimiento & desarrollo , Inmunoglobulinas/metabolismo , Mucosa Intestinal/metabolismo , Tejido Linfoide/metabolismo , Probióticos , Destete , Animales , Animales Recién Nacidos , Antígenos , Modelos Animales de Enfermedad , Sistema Inmunológico/microbiología , Inmunoglobulina A/biosíntesis , Inmunoglobulina G/metabolismo , Inmunoglobulina M/biosíntesis , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Tejido Linfoide/inmunología , Mastocitos/metabolismo , Permeabilidad , Valores de Referencia , Porcinos
15.
Gut ; 62(6): 842-51, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22661492

RESUMEN

BACKGROUND: The process of weaning causes a major shift in intestinal microbiota and is a critical period for developing appropriate immune responses in young mammals. OBJECTIVE: To use a new systems approach to provide an overview of host metabolism and the developing immune system in response to nutritional intervention around the weaning period. DESIGN: Piglets (n=14) were weaned onto either an egg-based or soya-based diet at 3 weeks until 7 weeks, when all piglets were switched onto a fish-based diet. Half the animals on each weaning diet received Bifidobacterium lactis NCC2818 supplementation from weaning onwards. Immunoglobulin production from immunologically relevant intestinal sites was quantified and the urinary (1)H NMR metabolic profile was obtained from each animal at post mortem (11 weeks). RESULTS: Different weaning diets induced divergent and sustained shifts in the metabolic phenotype, which resulted in the alteration of urinary gut microbial co-metabolites, even after 4 weeks of dietary standardisation. B lactis NCC2818 supplementation affected the systemic metabolism of the different weaning diet groups over and above the effects of diet. Additionally, production of gut mucosa-associated IgA and IgM was found to depend upon the weaning diet and on B lactis NCC2818 supplementation. CONCLUSION: The correlation of urinary (1)H NMR metabolic profile with mucosal immunoglobulin production was demonstrated, thus confirming the value of this multi-platform approach in uncovering non-invasive biomarkers of immunity. This has clear potential for translation into human healthcare with the development of urine testing as a means of assessing mucosal immune status. This might lead to early diagnosis of intestinal dysbiosis and with subsequent intervention, arrest disease development. This system enhances our overall understanding of pathologies under supra-organismal control.


Asunto(s)
Bifidobacterium , Dieta , Mucosa Intestinal/inmunología , Metaboloma , Probióticos/administración & dosificación , Destete , Fenómenos Fisiológicos Nutricionales de los Animales/inmunología , Animales , Huevos , Inmunoglobulina A/sangre , Inmunoglobulina M/sangre , Mucosa Intestinal/efectos de los fármacos , Intestinos/microbiología , Espectroscopía de Resonancia Magnética , Fenotipo , Glycine max , Porcinos
16.
Vet Immunol Immunopathol ; 152(1-2): 13-9, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23078904

RESUMEN

Studying the pathogenesis of an infectious disease like colibacillosis requires an understanding of the responses of target hosts to the organism both as a pathogen and as a commensal. The mucosal immune system constitutes the primary line of defence against luminal micro-organisms. The immunoglobulin-superfamily-based adaptive immune system evolved in the earliest jawed vertebrates, and the adaptive and innate immune system of humans, mice, pigs and ruminants co-evolved in common ancestors for approximately 300 million years. The divergence occurred only 100 mya and, as a consequence, most of the fundamental immunological mechanisms are very similar. However, since pressure on the immune system comes from rapidly evolving pathogens, immune systems must also evolve rapidly to maintain the ability of the host to survive and reproduce. As a consequence, there are a number of areas of detail where mammalian immune systems have diverged markedly from each other, such that results obtained in one species are not always immediately transferable to another. Thus, animal models of specific diseases need to be selected carefully, and the results interpreted with caution. Selection is made simpler where specific host species like cattle and pigs can be both target species and reservoirs for human disease, as in infections with Escherichia coli.


Asunto(s)
Evolución Biológica , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/inmunología , Escherichia coli/genética , Escherichia coli/inmunología , Inmunidad Innata/genética , Inmunidad Mucosa/genética , Animales , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/microbiología , Interacciones Huésped-Patógeno , Humanos , Ratones , Rumiantes , Porcinos
17.
Autoimmun Rev ; 12(6): 643-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23201916

RESUMEN

Multicellularity evolved well before 600 million years ago, and all multicellular animals have evolved since then with the need to protect against pathogens. There is no reason to expect their immune systems to be any less sophisticated than ours. The vertebrate system, based on rearranging immunoglobulin-superfamily domains, appears to have evolved partly as a result of chance insertion of RAG genes by horizontal transfer. Remarkably sophisticated systems for expansion of immunological repertoire have evolved in parallel in many groups of organisms. Vaccination of invertebrates against commercially important pathogens has been empirically successful, and suggests that the definition of an adaptive and innate immune system should no longer depend on the presence of memory and specificity, since these terms are hard to define in themselves. The evolution of randomly-created immunological repertoire also carries with it the potential for generating autoreactive specificities and consequent autoimmune damage. While invertebrates may use systems analogous to ours to control autoreactive specificities, they may have evolved alternative mechanisms which operate either at the level of individuals-within-populations rather than cells-within-individuals, by linking self-reactive specificities to regulatory pathways and non-self-reactive to effector pathways.


Asunto(s)
Evolución Biológica , Sistema Inmunológico , Inmunidad Adaptativa , Animales , Humanos
18.
Vet Immunol Immunopathol ; 149(3-4): 216-24, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22868203

RESUMEN

Maximising the ability of piglets to survive exposure to pathogens is essential to reduce early piglet mortality, an important factor in efficient commercial pig production. Mortality rates can be influenced by many factors, including early colonization by microbial commensals. Here we describe the development of an intestinal microbiota, the Bristol microbiota, for use in gnotobiotic pigs and its influence on synthesis of systemic immunoglobulins. Such a microbiota will be of value in studies of the consequences of early microbial colonization on development of the intestinal immune system and subsequent susceptibility to disease. Gnotobiotic pig studies lack a well-established intestinal microbiota. The use of the Altered Schaedler Flora (ASF), a murine intestinal microbiota, to colonize the intestines of Caesarean-derived, gnotobiotic pigs prior to gut closure, resulted in unreliable colonization with most (but not all) strains of the ASF. Subsequently, a novel, simpler porcine microbiota was developed. The novel microbiota reliably colonized the length of the intestinal tract when administered to gnotobiotic piglets. No health problems were observed, and the novel microbiota induced a systemic increase in serum immunoglobulins, in particular IgA and IgM. The Bristol microbiota will be of value for highly controlled, reproducible experiments of the consequences of early microbial colonization on susceptibility to disease in neonatal piglets, and as a biomedical model for the impact of microbial colonization on development of the intestinal mucosa and immune system in neonates.


Asunto(s)
Tracto Gastrointestinal/microbiología , Vida Libre de Gérmenes/inmunología , Metagenoma/inmunología , Porcinos/microbiología , Animales , Animales Recién Nacidos , ADN Bacteriano/química , ADN Bacteriano/genética , Ensayo de Inmunoadsorción Enzimática/veterinaria , Citometría de Flujo/veterinaria , Tracto Gastrointestinal/inmunología , Inmunoglobulinas/sangre , Reacción en Cadena de la Polimerasa/veterinaria , ARN Ribosómico 16S/química , ARN Ribosómico 16S/genética , Porcinos/inmunología
19.
Pediatr Allergy Immunol ; 23(3): 265-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22300455

RESUMEN

BACKGROUND: In mammals, early-life environmental variations appear to affect microbial colonization and therefore competent immune development, and exposure to farm environments in infants has been inversely correlated with allergy development. Modelling these effects using manipulation of neonatal rodents is difficult due to their dependency on the mother, but the relatively independent piglet is increasingly identified as a valuable translational model for humans. This study was designed to correlate immune regulation in piglets with early-life environment. METHODS: Piglets were nursed by their mother on a commercial farm, while isolator-reared siblings were formula fed. Fluorescence immunohistology was used to quantify T-reg and effector T-cell populations in the intestinal lamina propria and the systemic response to food proteins was quantified by capture ELISA. RESULTS: There was more CD4(+) and CD4(+) CD25(+) effector T-cell staining in the intestinal mucosa of the isolator-reared piglets compared with their farm-reared counterparts. In contrast, these isolator-reared piglets had a significantly reduced CD4(+) CD25(+) Foxp3(+) regulatory T-cell population compared to farm-reared littermates, resulting in a significantly higher T-reg-to-effector ratio in the farm animals. Consistent with these findings, isolator-reared piglets had an increased serum IgG anti-soya response to novel dietary soya protein relative to farm-reared piglets. CONCLUSION: Here, we provide the first direct evidence, derived from intervention, that components of the early-life environment present on farms profoundly affects both local development of regulatory components of the mucosal immune system and immune responses to food proteins at weaning. We propose that neonatal piglets provide a tractable model which allows maternal and treatment effects to be statistically separated.


Asunto(s)
Agricultura , Animales Recién Nacidos/inmunología , Exposición a Riesgos Ambientales , Modelos Animales , Porcinos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Femenino , Humanos , Hipersensibilidad/inmunología , Sistema Inmunológico , Inmunidad Mucosa , Destete
20.
PLoS One ; 6(12): e28284, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22164261

RESUMEN

BACKGROUND: Early gut colonization events are purported to have a major impact on the incidence of infectious, inflammatory and autoimmune diseases in later life. Hence, factors which influence this process may have important implications for both human and animal health. Previously, we demonstrated strong influences of early-life environment on gut microbiota composition in adult pigs. Here, we sought to further investigate the impact of limiting microbial exposure during early life on the development of the pig gut microbiota. METHODOLOGY/PRINCIPAL FINDINGS: Outdoor- and indoor-reared animals, exposed to the microbiota in their natural rearing environment for the first two days of life, were transferred to an isolator facility and adult gut microbial diversity was analyzed by 16S rRNA gene sequencing. From a total of 2,196 high-quality 16S rRNA gene sequences, 440 phylotypes were identified in the outdoor group and 431 phylotypes in the indoor group. The majority of clones were assigned to the four phyla Firmicutes (67.5% of all sequences), Proteobacteria (17.7%), Bacteroidetes (13.5%) and to a lesser extent, Actinobacteria (0.1%). Although the initial maternal and environmental microbial inoculum of isolator-reared animals was identical to that of their naturally-reared littermates, the microbial succession and stabilization events reported previously in naturally-reared outdoor animals did not occur. In contrast, the gut microbiota of isolator-reared animals remained highly diverse containing a large number of distinct phylotypes. CONCLUSIONS/SIGNIFICANCE: The results documented here indicate that establishment and development of the normal gut microbiota requires continuous microbial exposure during the early stages of life and this process is compromised under conditions of excessive hygiene.


Asunto(s)
Tracto Gastrointestinal/microbiología , Metagenoma/fisiología , Animales , Biodiversidad , Cruzamientos Genéticos , Biblioteca de Genes , Higiene , Mucosa Intestinal/microbiología , Filogenia , ARN Ribosómico 16S/genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...