Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Gut Microbes ; 16(1): 2379862, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39042143

RESUMEN

Streptococcus agalactiae, also known as Group B Streptococcus (GBS), is a predominant pathogen of neonatal sepsis, commonly associated with early-onset neonatal sepsis. GBS has also been associated with cases of late-onset sepsis potentially originating from the intestine. Previous findings have shown GBS can colonize the infant intestinal tract as part of the neonatal microbiota. To better understand GBS colonization dynamics in the neonatal intestine, we collected stool and milk samples from prematurely born neonates for identification of potential pathogens in the neonatal intestinal microbiota. GBS was present in approximately 10% of the cohort, and this colonization was not associated with maternal GBS status, delivery route, or gestational weight. Interestingly, we observed the relative abundance of GBS in the infant stool negatively correlated with maternal IgA concentration in matched maternal milk samples. Using a preclinical murine model of GBS infection, we report that both vertical transmission and direct oral introduction resulted in intestinal colonization of GBS; however, translocation beyond the intestine was limited. Finally, vaccination of dams prior to breeding induced strong immunoglobulin responses, including IgA responses, which were associated with reduced mortality and GBS intestinal colonization. Taken together, we show that maternal IgA may contribute to infant immunity by limiting the colonization of GBS in the intestine.


Asunto(s)
Traslocación Bacteriana , Inmunoglobulina A , Infecciones Estreptocócicas , Streptococcus agalactiae , Streptococcus agalactiae/crecimiento & desarrollo , Streptococcus agalactiae/inmunología , Animales , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/prevención & control , Infecciones Estreptocócicas/inmunología , Femenino , Recién Nacido , Humanos , Ratones , Transmisión Vertical de Enfermedad Infecciosa , Heces/microbiología , Intestinos/microbiología , Intestinos/inmunología , Leche Humana/microbiología , Microbioma Gastrointestinal , Embarazo , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Masculino
2.
Immunol Rev ; 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39001685

RESUMEN

Oral tolerance promotes the suppression of immune responses to innocuous antigen and is primarily mediated by regulatory T cell (Tregs). The development of oral tolerance begins in early life during a "window of tolerance," which occurs around weaning and is mediated by components in breastmilk. Herein, we review the factors dictating this window and how Tregs are uniquely educated in early life. In early life, the translocation of luminal antigen for Treg induction is primarily dictated by goblet cell-associated antigen passages (GAPs). GAPs in the colon are negatively regulated by maternally-derived epidermal growth factor and the microbiota, restricting GAP formation to the "periweaning" period (postnatal day 11-21 in mice, 4-6 months in humans). The induction of solid food also promotes the diversification of the bacteria such that bacterially-derived metabolites known to promote Tregs-short-chain fatty acids, tryptophan metabolites, and bile acids-peak during the periweaning phase. Further, breastmilk immunoglobulins-IgA and IgG-regulate both microbial diversity and the interaction of microbes with the epithelium, further controlling which antigens are presented to T cells. Overall, these elements work in conjunction to induce a long-lived population of Tregs, around weaning, that are crucial for maintaining homeostasis in adults.

3.
iScience ; 27(5): 109669, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38646164

RESUMEN

Neonates born prematurely are vulnerable to life-threatening conditions such as bacterial sepsis. Streptococcus agalactiae (GBS) and Escherichia coli are frequent causative pathogens of neonatal sepsis, however, it remains unclear if these pathogens induce differential immune responses. We find that γδ T cells rapidly respond to single-organism GBS and E. coli bloodstream infections in neonatal mice. Furthermore, GBS and E. coli induce distinct cytokine production from IFN-γ and IL-17 producing γδ T cells, respectively. We also find that IL-17 production during E. coli infection is driven by γδTCR signaling, whereas IFN-γ production during GBS infection occurs independently of γδTCR signaling. The divergent effector responses of γδ T cells during GBS and E. coli infections impart distinctive neuroinflammatory phenotypes on the neonatal brain. Thus, the neonatal adaptive immune system differentially responds to distinct bacterial stimuli, resulting in unique neuroinflammatory phenotypes.

4.
bioRxiv ; 2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-37873122

RESUMEN

Neonates born prematurely are highly vulnerable to life-threatening conditions such as bacterial sepsis. Streptococcus agalactiae, also known as group B Streptococcus (GBS) and Escherichia coli are frequent causative pathogens of neonatal sepsis, however, it remains unclear if distinct sepsis pathogens induce differential adaptive immune responses. In the present study, we find that γδ T cells in neonatal mice rapidly respond to single-organism GBS and E. coli bloodstream infections and that these pathogens induce distinct activation and cytokine production from IFN-γ and IL-17 producing γδ T cells, respectively. We also report differential reliance on γδTCR signaling to elicit effector cytokine responses during neonatal sepsis, with IL-17 production during E. coli infection being driven by γδTCR signaling, and IFN-γ production during GBS infection occurring independently of γδTCR signaling. Furthermore, we report that the divergent effector responses of γδ T cells during GBS and E. coli infections impart distinctive neuroinflammatory phenotypes on the neonatal brain. The present study reveals that the neonatal adaptive immune system differentially responds to distinct bacterial stimuli, resulting in unique neuroinflammatory phenotypes.

5.
Elife ; 102021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34677124

RESUMEN

Intestinal goblet cells maintain the protective epithelial barrier through mucus secretion and yet sample lumenal substances for immune processing through formation of goblet cell associated antigen passages (GAPs). The cellular biology of GAPs and how these divergent processes are balanced and regulated by goblet cells remains unknown. Using high-resolution light and electron microscopy, we found that in mice, GAPs were formed by an acetylcholine (ACh)-dependent endocytic event remarkable for delivery of fluid-phase cargo retrograde into the trans-golgi network and across the cell by transcytosis - in addition to the expected transport of fluid-phase cargo by endosomes to multi-vesicular bodies and lysosomes. While ACh also induced goblet cells to secrete mucins, ACh-induced GAP formation and mucin secretion were functionally independent and mediated by different receptors and signaling pathways, enabling goblet cells to differentially regulate these processes to accommodate the dynamically changing demands of the mucosal environment for barrier maintenance and sampling of lumenal substances.


Cells in the gut need to be protected against the many harmful microbes which inhabit this environment. Yet the immune system also needs to 'keep an eye' on intestinal contents to maintain tolerance to innocuous substances, such as those from the diet. The 'goblet cells' that are part of the gut lining do both: they create a mucus barrier that stops germs from invading the body, but they also can pass on molecules from the intestine to immune cells deep in the tissue to promote tolerance. This is achieved through a 'GAP' mechanism. A chemical messenger called acetylcholine can trigger both mucus release and the GAP process in goblet cells. Gustafsson et al. investigated how the cells could take on these two seemingly opposing roles in response to the same signal. A fluorescent molecule was introduced into the intestines of mice, and monitored as it pass through the goblet cells. This revealed how the GAP process took place: the cells were able to capture molecules from the intestines, wrap them in internal sack-like vesicles and then transport them across the entire cell. To explore the role of acetylcholine, Gustafsson et al. blocked the receptors that detect the messenger at the surface of goblet cells. Different receptors and therefore different cascades of molecular events were found to control mucus secretion and GAP formation; this explains how the two processes can be performed in parallel and independently from each other. Understanding how cells relay molecules to the immune system is relevant to other tissues in contact with the environment, such as the eyes, the airways, or the inside of the genital and urinary tracts. Understanding, and then ultimately harnessing this mechanism could help design of new ways to deliver drugs to the immune system and alter immune outcomes.


Asunto(s)
Antígenos/metabolismo , Células Caliciformes/metabolismo , Transcitosis , Vesículas Transportadoras/fisiología , Animales , Ratones
6.
Immunohorizons ; 5(6): 512-522, 2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34183380

RESUMEN

Sepsis can result from a variety of pathogens, originating from a range of sources. A vast range of presenting symptoms is included in the catch-all term of "bacteremia," making diagnosis and prognosis particularly troublesome. One underexplored factor contributing to disparate outcomes is the age of the patient. Neonatal sepsis in very-low-birth-weight infants can result in vastly different immunological outcomes unique from sepsis in adults. It is also becoming increasingly clear, both from preclinical experimental models and clinical observations, that the age and history of previous microbial exposures can significantly influence the course of infection from sepsis and cytokine storms to immunopathology. In this study, we will explore key differences between neonatal and adult sepsis, experimental models used to study sepsis, and how responses to the surrounding microbial universe shape development of the immune system and impact, positively or negatively, the course of disease.


Asunto(s)
Síndrome de Liberación de Citoquinas/inmunología , Interacciones Huésped-Patógeno/inmunología , Sepsis/inmunología , Adulto , Factores de Edad , Animales , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/microbiología , Síndrome de Liberación de Citoquinas/mortalidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Recién Nacido , Sepsis/genética , Sepsis/microbiología , Sepsis/mortalidad , Índice de Severidad de la Enfermedad
7.
Clin Exp Allergy ; 51(4): 518-526, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33403739

RESUMEN

The increasing incidence of food allergy remains a significant public health concern. Food allergy is partially due to a lack, or loss of tolerance to food allergens. Clinical outcomes surrounding early life practices, such as breastfeeding, antibiotic use and food allergen exposure, indicate the first year of life in children represents a unique time for shaping the immune system to reduce allergic outcomes. Animal models have identified distinctive aspects of when and where dietary antigens are delivered within the intestinal tract to promote oral tolerance prior to weaning. Additionally, animal models have identified contributions from maternal proteins from breast milk and bacterial products from the gut microbiota in regulating dietary antigen exposure and promoting oral tolerance, thus connecting decades of clinical observations on the benefits of breastfeeding, early food allergen introduction and antibiotic avoidance in the first year of life in reducing allergic outcomes. Here, we discuss how exposure to gut luminal antigens, including food allergens, is regulated in early life to generate protective tolerance and the implications of this process for preventing and treating food allergies.


Asunto(s)
Antígenos/inmunología , Hipersensibilidad a los Alimentos/inmunología , Microbioma Gastrointestinal/inmunología , Intestinos/inmunología , Leche Humana/inmunología , Administración Oral , Alérgenos/inmunología , Antibacterianos , Lactancia Materna , Células Dendríticas/inmunología , Células Caliciformes/inmunología , Humanos , Lactante , Recién Nacido , Células Th2/inmunología
8.
Cell Rep Med ; 1(7)2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33196055

RESUMEN

Stress is a known trigger for flares of inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS); however, this process is not well understood. Here, we find that restraint stress in mice leads to signs of diarrhea, fecal dysbiosis, and a barrier defect via the opening of goblet-cell associated passages. Notably, stress increases host immunity to gut bacteria as assessed by immunoglobulin A (IgA)-bound gut bacteria. Stress-induced microbial changes are necessary and sufficient to elicit these effects. Moreover, similar to mice, many diarrhea-predominant IBS (IBS-D) patients from two cohorts display increased antibacterial immunity as assessed by IgA-bound fecal bacteria. This antibacterial IgA response in IBS-D correlates with somatic symptom severity and was distinct from healthy controls or IBD patients. These findings suggest that stress may play an important role in patients with IgA-associated IBS-D by disrupting the intestinal microbial community that alters gastrointestinal function and host immunity to commensal bacteria.


Asunto(s)
Diarrea/inmunología , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Inmunoglobulina A/biosíntesis , Síndrome del Colon Irritable/inmunología , Estrés Psicológico/inmunología , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/crecimiento & desarrollo , Bacterias/inmunología , Traslocación Bacteriana , Diarrea/microbiología , Diarrea/patología , Disbiosis/microbiología , Disbiosis/patología , Heces/microbiología , Femenino , Humanos , Inmovilización/psicología , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/patología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , Estrés Psicológico/microbiología , Estrés Psicológico/patología , Simbiosis
9.
JCI Insight ; 5(15)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32759496

RESUMEN

Allergic disorders, characterized by Th2 immune responses to environmental substances, are increasingly common in children in Western societies. Multiple studies indicate that breastfeeding, early complementary introduction of food allergens, and antibiotic avoidance in the first year of life reduces allergic outcomes in at-risk children. Why the benefit of these practices is restricted to early life is largely unknown. We identified a preweaning interval during which dietary antigens are assimilated by the colonic immune system. This interval is under maternal control via temporal changes in breast milk, coincides with an influx of naive T cells into the colon, and is followed by the development of a long-lived population of colonic peripherally derived Tregs (pTregs) that can be specific for dietary antigens encountered during this interval. Desynchronization of mothers and offspring produced durable deficits in these pTregs, impaired tolerance to dietary antigens introduced during and after this preweaning interval, and resulted in spontaneous Th2 responses. These effects could be rescued by pTregs from the periweaning colon or by Tregs generated in vitro using periweaning colonic antigen-presenting cells. These findings demonstrate that mothers and their offspring are synchronized for the development of a balanced immune system.


Asunto(s)
Alérgenos/inmunología , Colon/inmunología , Hipersensibilidad a los Alimentos/prevención & control , Tolerancia Inmunológica/inmunología , Leche/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Animales , Animales Recién Nacidos , Células Presentadoras de Antígenos/inmunología , Femenino , Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos C57BL , Madres , Ovalbúmina/inmunología , Destete
10.
Nutrients ; 12(4)2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32290170

RESUMEN

Late-onset sepsis (LOS) and other systemic bloodstream infections are notable causes of neonatal mortality, particularly in prematurely born very low birth weight infants. Breastfeeding in early life has numerous health benefits, impacting the health of the newborn in both the short-term and in the long-term. Though the known benefits of an exclusive mother's own milk diet in early life have been well recognized and described, it is less understood how breastfed infants enjoy a potential reduction in risk of LOS and other systemic infections. Here we review how gut residing pathogens within the intestinal microbiota of infants can cause a subset of sepsis cases and the components of breastmilk that may prevent the dissemination of pathogens from the intestine.


Asunto(s)
Lactancia Materna , Disbiosis/microbiología , Microbioma Gastrointestinal , Recién Nacido , Leche Humana/fisiología , Sepsis Neonatal/microbiología , Sepsis Neonatal/prevención & control , Antibacterianos/efectos adversos , Contraindicaciones de los Medicamentos , Disbiosis/complicaciones , Femenino , Bacterias Gramnegativas/patogenicidad , Bacterias Grampositivas/patogenicidad , Humanos , Lactante , Masculino , Sepsis Neonatal/etiología , Riesgo
11.
Proc Natl Acad Sci U S A ; 117(14): 7941-7949, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32179676

RESUMEN

Late-onset sepsis (LOS) is a highly consequential complication of preterm birth and is defined by a positive blood culture obtained after 72 h of age. The causative bacteria can be found in patients' intestinal tracts days before dissemination, and cohort studies suggest reduced LOS risk in breastfed preterm infants through unknown mechanisms. Reduced concentrations of epidermal growth factor (EGF) of maternal origin within the intestinal tract of mice correlated to the translocation of a gut-resident human pathogen Escherichia coli, which spreads systemically and caused a rapid, fatal disease in pups. Translocation of Escherichia coli was associated with the formation of colonic goblet cell-associated antigen passages (GAPs), which translocate enteric bacteria across the intestinal epithelium. Thus, maternally derived EGF, and potentially other EGFR ligands, prevents dissemination of a gut-resident pathogen by inhibiting goblet cell-mediated bacterial translocation. Through manipulation of maternally derived EGF and alteration of the earliest gut defenses, we have developed an animal model of pathogen dissemination which recapitulates gut-origin neonatal LOS.


Asunto(s)
Traslocación Bacteriana/inmunología , Receptores ErbB/metabolismo , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Microbioma Gastrointestinal/inmunología , Leche Humana/inmunología , Sepsis Neonatal/inmunología , Animales , Animales Recién Nacidos , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Lactancia Materna , Colon/metabolismo , Colon/microbiología , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Escherichia coli/aislamiento & purificación , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Heces/química , Heces/microbiología , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Transgénicos , Leche Humana/metabolismo , Sepsis Neonatal/metabolismo , Sepsis Neonatal/microbiología , Transducción de Señal/inmunología , Factores de Tiempo
12.
Lab Anim (NY) ; 49(3): 79-88, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32042160

RESUMEN

The intestinal immune system samples luminal contents to induce adaptive immune responses that include tolerance in the steady state and protective immunity during infection. How luminal substances are delivered to the immune system has not been fully investigated. Goblet cells have an important role in this process by delivering luminal substances to the immune system through the formation of goblet cell-associated antigen passages (GAPs). Soluble antigens in the intestinal lumen are transported across the epithelium transcellularly through GAPs and delivered to dendritic cells for presentation to T cells and induction of immune responses. GAPs can be identified and quantified by using the ability of GAP-forming goblet cells to take up fluorescently labeled dextran. Here, we describe a method to visualize GAPs and other cells that have the capacity to take up luminal substances by intraluminal injection of fluorescent dextran in mice under anesthesia, tissue sectioning for slide preparation and imaging with fluorescence microscopy. In contrast to in vivo two-photon imaging previously used to identify GAPs, this technique is not limited by anatomical constraints and can be used to visualize GAP formation throughout the length of the intestine. In addition, this method can be combined with common immunohistochemistry protocols to visualize other cell types. This approach can be used to compare GAP formation following different treatments or changes to the luminal environment and to uncover how sampling of luminal substances is altered in pathophysiological conditions. This protocol requires 8 working hours over 2-3 d to be completed.


Asunto(s)
Antígenos/metabolismo , Colon/inmunología , Células Dendríticas/inmunología , Células Caliciformes/inmunología , Vigilancia Inmunológica , Intestino Delgado/inmunología , Animales , Presentación de Antígeno/efectos de los fármacos , Antígenos/inmunología , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Dextranos/administración & dosificación , Colorantes Fluorescentes/administración & dosificación , Células Caliciformes/efectos de los fármacos , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL , Microbiota/inmunología , Ovalbúmina/administración & dosificación , Proyectos de Investigación
13.
Front Immunol ; 11: 603059, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613522

RESUMEN

Atopic disorders including allergic rhinitis, asthma, food allergy, and dermatitis, are increasingly prevalent in Western societies. These disorders are largely characterized by T helper type 2 (Th2) immune responses to environmental triggers, particularly inhaled and dietary allergens. Exposure to such stimuli during early childhood reduces the frequency of allergies in at-risk children. These allergic responses can be restrained by regulatory T cells (Tregs), particularly Tregs arising in the gut. The unique attributes of how early life exposure to diet and microbes shape the intestinal Treg population is a topic of significant interest. While imprinting during early life promotes the development of a balanced immune system and protects against immunopathology, it remains unclear if Tregs that develop in early life continue to restrain systemic inflammatory responses throughout adulthood. Here, an inducible deletion strategy was used to label Tregs at specified time points with a targeted mechanism to be deleted later. Deletion of the Tregs labeled peri-weaning at day of life 24, but not before weaning at day of life 14, resulted in increased circulating IgE and IL-13, and abrogated induction of tolerance towards new antigens. Thus, Tregs developing peri-weaning, but not before day of life 14 are continually required to restrain allergic responses into adulthood.


Asunto(s)
Comunicación Celular , Colon/inmunología , Citocinas/sangre , Hipersensibilidad Tardía/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Administración Oral , Traslado Adoptivo , Factores de Edad , Animales , Animales Modificados Genéticamente , Antígenos/administración & dosificación , Antígenos/inmunología , Colon/metabolismo , Modelos Animales de Enfermedad , Hipersensibilidad Tardía/sangre , Hipersensibilidad Tardía/genética , Tolerancia Inmunológica , Inmunoglobulina E/sangre , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Ovalbúmina , Fenotipo , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Células Th2/metabolismo , Destete
14.
Mucosal Immunol ; 13(2): 271-282, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31819172

RESUMEN

Tolerance to innocuous antigens from the diet and the commensal microbiota is a fundamental process essential to health. Why tolerance is efficiently induced to substances arising from the hostile environment of the gut lumen is incompletely understood but may be related to how these antigens are encountered by the immune system. We observed that goblet cell associated antigen passages (GAPs), but not other pathways of luminal antigen capture, correlated with the acquisition of luminal substances by lamina propria (LP) antigen presenting cells (APCs) and with the sites of tolerance induction to luminal antigens. Strikingly this role extended beyond antigen delivery. The GAP function of goblet cells facilitated maintenance of pre-existing LP T regulatory cells (Tregs), imprinting LP-dendritic cells with tolerogenic properties, and facilitating LP macrophages to produce the immunomodulatory cytokine IL-10. Moreover, tolerance to dietary antigen was impaired in the absence of GAPs. Thus, by delivering luminal antigens, maintaining pre-existing LP Tregs, and imprinting tolerogenic properties on LP-APCs GAPs support tolerance to substances encountered in the hostile environment of the gut lumen.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Dendríticas/inmunología , Células Caliciformes/inmunología , Macrófagos/inmunología , Membrana Mucosa/inmunología , Linfocitos T Reguladores/inmunología , Administración Oral , Animales , Presentación de Antígeno , Antígenos/inmunología , Células Cultivadas , Proteínas Activadoras de GTPasa/metabolismo , Tolerancia Inmunológica , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
15.
J Allergy Clin Immunol ; 144(4): 1058-1073.e3, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31175877

RESUMEN

BACKGROUND: Food-induced anaphylaxis (FIA) is an IgE-dependent immune response that can affect multiple organs and lead to life-threatening complications. The processes by which food allergens cross the mucosal surface and are delivered to the subepithelial immune compartment to promote the clinical manifestations associated with food-triggered anaphylaxis are largely unexplored. OBJECTIVE: We sought to define the processes involved in the translocation of food allergens across the mucosal epithelial surface to the subepithelial immune compartment in FIA. METHODS: Two-photon confocal and immunofluorescence microscopy was used to visualize and trace food allergen passage in a murine model of FIA. A human colon cancer cell line, RNA silencing, and pharmacologic approaches were used to identify the molecular regulation of intestinal epithelial allergen uptake and translocation. Human intestinal organoid transplants were used to demonstrate the conservation of these molecular processes in human tissues. RESULTS: Food allergens are sampled by using small intestine (SI) epithelial secretory cells (termed secretory antigen passages [SAPs]) that are localized to the SI villous and crypt region. SAPs channel food allergens to lamina propria mucosal mast cells through an IL-13-CD38-cyclic adenosine diphosphate ribose (cADPR)-dependent process. Blockade of IL-13-induced CD38/cADPR-dependent SAP antigen passaging in mice inhibited induction of clinical manifestations of FIA. IL-13-CD38-cADPR-dependent SAP sampling of food allergens was conserved in human intestinal organoids. CONCLUSION: We identify that SAPs are a mechanism by which food allergens are channeled across the SI epithelium mediated by the IL-13/CD38/cADPR pathway, regulate the onset of FIA reactions, and are conserved in human intestine.


Asunto(s)
Alérgenos/inmunología , Anafilaxia/inmunología , Hipersensibilidad a los Alimentos/inmunología , Interleucina-13/inmunología , Mucosa Intestinal/inmunología , Alérgenos/metabolismo , Anafilaxia/metabolismo , Animales , Hipersensibilidad a los Alimentos/metabolismo , Humanos , Inmunoglobulina E/inmunología , Interleucina-13/metabolismo , Mucosa Intestinal/metabolismo , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID
16.
Inflamm Bowel Dis ; 25(1): 180-193, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29982468

RESUMEN

Background: Assessing risk of Crohn's disease (CD) recurrence following ileocolic resection (ICR) is necessary to optimize medical management and prevent long-term complications. This study aimed to identify noninvasive markers that could predict postoperative disease activity. Methods: Inclusion criteria were a diagnosis of CD, first ICR, interval colonoscopy, and whole transcriptome array meeting quality control standards. Demographic and clinical data were obtained from the electronic medical record. RNA extraction and human transcriptome microarray were performed on noninflamed ileal margins from operative specimens. Clinical data and random forest were analyzed in R. Principal components analysis, hierarchical clustering, and pathway enrichment were performed in Partek. Results: Sixty-five patients completed the study, and 5 were excluded from analysis due to extreme variability on whole transcriptome analysis. Unsupervised hierarchical clustering revealed that patients with an i0 Rutgeerts score generally segregated from all others. In anti-TNF-naïve patients, unsupervised hierarchical clustering revealed complete segregation of patients with an i0 score. Reduced escalation in therapy and continued mucosal remission, consistent with indolent disease, were seen in the 4 years following surgery. Random forest identified 30 transcripts differentiating i0 patients from the other groups. Pathway enrichment highlighted toll-like receptor, NOD-like receptor, and TNF signaling. This transcriptome signature did not identify i0 anti-TNF-exposed patients. However, anti-TNF-exposed patients with indolent postoperative courses were found to have a transcriptome signature distinct from those with aggressive disease. Conclusions: Anti-TNF-naïve and -exposed patients have unique expression profiles at the time of surgery, which may offer predictive value in assessing the risk of nonrecurrence. 10.1093/ibd/izy228_video1izy228.video15804852517001.


Asunto(s)
Anastomosis Quirúrgica/efectos adversos , Colectomía/efectos adversos , Colon/cirugía , Enfermedad de Crohn/cirugía , Íleon/cirugía , Complicaciones Posoperatorias/diagnóstico , Transcriptoma/efectos de los fármacos , Adulto , Anticuerpos Monoclonales/uso terapéutico , Estudios de Cohortes , Terapia Combinada , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/patología , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Humanos , Masculino , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/genética , Pronóstico , Recurrencia , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/uso terapéutico
17.
Birth Defects Res ; 110(20): 1494-1503, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30576093

RESUMEN

The gut microbiome and the immune system codevelop around the time of birth, well after genetic information has been passed from the parents to the offspring. Each of these "organ systems" displays plasticity. The immune system can mount highly specific adaptive responses to newly encountered antigens, and the gut microbiota is affected by changes in the environment. Despite this plasticity, there is a growing appreciation that these organ systems, once established, are remarkably stable. In health, the immune system rapidly mounts responses to infections, and once cleared, resolves inflammatory responses to return to homeostasis. However, a skewed immune system, such as seen in allergy, does not easily return to homeostasis. Allergic responses are often seen to multiple antigens. Likewise, a dysbiotic gut microbiota is seen in multiple diseases. Attempts to reset the gut microbiota as a therapy for disease have met with varied success. Therefore, how these codeveloping "organ systems" become established is a central question relevant to our overall health. Recent observations suggest that maternal factors encountered both in utero and after birth can directly or indirectly impact the development of the offspring's gut microbiome and immune system. Here, we discuss how these nongenetic maternal influences can have long-term effects on the progeny's health.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Femenino , Homeostasis , Humanos , Sistema Inmunológico/fisiología , Madres , Embarazo
18.
Mucosal Immunol ; 11(6): 1551-1557, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29867079

RESUMEN

Goblet cells (GCs) are specialized epithelial cells that line multiple mucosal surfaces and have a well-appreciated role in barrier maintenance through the secretion of mucus. Moreover, GCs secrete anti-microbial proteins, chemokines, and cytokines demonstrating functions in innate immunity beyond barrier maintenance. Recently it was appreciated that GCs can form goblet cell-associated antigen passages (GAPs) and deliver luminal substances to underlying lamina propria (LP) antigen-presenting cells (APCs) in a manner capable of inducing adaptive immune responses. GCs at other mucosal surfaces share characteristics with the GAP forming intestinal GCs, suggesting that GAP formation may not be restricted to the gut, and that GCs may perform this gatekeeper function at other mucosal surfaces. Here we review observations of how GCs contribute to immunity at mucosal surfaces through barrier maintenance, the delivery of luminal substances to APCs, interactions with APCs, and secretion of factors modulating immune responses.


Asunto(s)
Células Caliciformes/inmunología , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Transcitosis , Animales , Presentación de Antígeno , Péptidos Catiónicos Antimicrobianos/metabolismo , Homeostasis , Humanos , Inmunomodulación
19.
Mucosal Immunol ; 11(4): 1103-1113, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29445136

RESUMEN

Dietary antigen acquisition by lamina propria (LP) dendritic cells (DCs) is crucial to induce oral tolerance and maintain homeostasis. However, encountering innocuous antigens during infection can lead to inflammatory responses, suggesting processes may limit steady-state luminal antigen capture during infection. We observed that goblet cell (GC) associated antigen passages (GAPs), a steady-state pathway delivering luminal antigens to LP-DCs, are inhibited during Salmonella infection. GAP inhibition was mediated by IL-1ß. Infection abrogated luminal antigen delivery and antigen-specific T cell proliferation in the mesenteric lymph node (MLN). Antigen-specific T cell proliferation to dietary antigen was restored by overriding GAP suppression; however, this did not restore regulatory T cell induction, but induced inflammatory T cell responses. Salmonella translocation to the MLN required GCs and correlated with GAPs. Genetic manipulations overriding GAP suppression, or antibiotics inducing colonic GAPs, but not antibiotics that do not, increased dissemination and worsened outcomes independent of luminal pathogen burden. Thus, steady-state sampling pathways are suppressed during infection to prevent responses to dietary antigens, limit pathogen entry, and lessen the disease. Moreover, antibiotics may worsen Salmonella infection by means beyond blunting gut microbiota colonization resistance, providing new insight into how precedent antibiotic use aggravates enteric infection.


Asunto(s)
Células Dendríticas/inmunología , Células Caliciformes/inmunología , Membrana Mucosa/patología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T/inmunología , Animales , Presentación de Antígeno , Antígenos/inmunología , Proliferación Celular , Proteínas en la Dieta/inmunología , Transmisión de Enfermedad Infecciosa , Microbioma Gastrointestinal/inmunología , Interacciones Huésped-Patógeno , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Salmonella typhimurium/patogenicidad
20.
Sci Immunol ; 2(18)2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29246946

RESUMEN

We have a mutually beneficial relationship with the trillions of microorganisms inhabiting our gastrointestinal tract. However, maintaining this relationship requires recognizing these organisms as affable and restraining inflammatory responses to these organisms when encountered in hostile settings. How and when the immune system develops tolerance to our gut microbial members is not well understood. We identify a specific preweaning interval in which gut microbial antigens are encountered by the immune system to induce antigen-specific tolerance to gut bacteria. For some bacterial taxa, physiologic encounters with the immune system are restricted to this interval, despite abundance of these taxa in the gut lumen at later times outside this interval. Antigen-specific tolerance to gut bacteria induced during this preweaning interval is stable and maintained even if these taxa are encountered later in life in an inflammatory setting. However, inhibiting microbial antigen encounter during this interval or extending these encounters beyond the normal interval results in a failure to induce tolerance and robust antigen-specific effector responses to gut bacteria upon reencounter in an inflammatory setting. Thus, we have identified a defined preweaning interval critical for developing tolerance to gut bacteria and maintaining the mutually beneficial relationship with our gut microbiota.


Asunto(s)
Antígenos Bacterianos/inmunología , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Tolerancia Inmunológica/inmunología , Animales , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Destete
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA