Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
iScience ; 27(5): 109669, 2024 May 17.
Article En | MEDLINE | ID: mdl-38646164

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.

2.
bioRxiv ; 2024 Mar 16.
Article En | MEDLINE | ID: mdl-37873122

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.

3.
Elife ; 102021 10 22.
Article En | MEDLINE | ID: mdl-34677124

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.


Antigens/metabolism , Goblet Cells/metabolism , Transcytosis , Transport Vesicles/physiology , Animals , Mice
4.
Immunohorizons ; 5(6): 512-522, 2021 06 28.
Article En | MEDLINE | ID: mdl-34183380

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.


Cytokine Release Syndrome/immunology , Host-Pathogen Interactions/immunology , Sepsis/immunology , Adult , Age Factors , Animals , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/microbiology , Cytokine Release Syndrome/mortality , Disease Models, Animal , Disease Progression , Humans , Infant, Newborn , Sepsis/genetics , Sepsis/microbiology , Sepsis/mortality , Severity of Illness Index
5.
Clin Exp Allergy ; 51(4): 518-526, 2021 04.
Article En | MEDLINE | ID: mdl-33403739

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.


Antigens/immunology , Food Hypersensitivity/immunology , Gastrointestinal Microbiome/immunology , Intestines/immunology , Milk, Human/immunology , Administration, Oral , Allergens/immunology , Anti-Bacterial Agents , Breast Feeding , Dendritic Cells/immunology , Goblet Cells/immunology , Humans , Infant , Infant, Newborn , Th2 Cells/immunology
6.
Cell Rep Med ; 1(7)2020 10 20.
Article En | MEDLINE | ID: mdl-33196055

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.


Diarrhea/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Immunity, Mucosal , Immunoglobulin A/biosynthesis , Irritable Bowel Syndrome/immunology , Stress, Psychological/immunology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Bacteria/immunology , Bacterial Translocation , Diarrhea/microbiology , Diarrhea/pathology , Dysbiosis/microbiology , Dysbiosis/pathology , Feces/microbiology , Female , Humans , Immobilization/psychology , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/pathology , Male , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics , Stress, Psychological/microbiology , Stress, Psychological/pathology , Symbiosis
7.
JCI Insight ; 5(15)2020 08 06.
Article En | MEDLINE | ID: mdl-32759496

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.


Allergens/immunology , Colon/immunology , Food Hypersensitivity/prevention & control , Immune Tolerance/immunology , Milk/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , Animals, Newborn , Antigen-Presenting Cells/immunology , Female , Food Hypersensitivity/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mothers , Ovalbumin/immunology , Weaning
8.
Nutrients ; 12(4)2020 Apr 10.
Article En | MEDLINE | ID: mdl-32290170

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.


Breast Feeding , Dysbiosis/microbiology , Gastrointestinal Microbiome , Infant, Newborn , Milk, Human/physiology , Neonatal Sepsis/microbiology , Neonatal Sepsis/prevention & control , Anti-Bacterial Agents/adverse effects , Contraindications, Drug , Dysbiosis/complications , Female , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/pathogenicity , Humans , Infant , Male , Neonatal Sepsis/etiology , Risk
9.
Proc Natl Acad Sci U S A ; 117(14): 7941-7949, 2020 04 07.
Article En | MEDLINE | ID: mdl-32179676

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.


Bacterial Translocation/immunology , ErbB Receptors/metabolism , Escherichia coli Infections/immunology , Escherichia coli/immunology , Gastrointestinal Microbiome/immunology , Milk, Human/immunology , Neonatal Sepsis/immunology , Animals , Animals, Newborn , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Breast Feeding , Colon/metabolism , Colon/microbiology , Disease Models, Animal , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Feces/chemistry , Feces/microbiology , Female , Humans , Infant, Newborn , Infant, Premature/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice , Mice, Transgenic , Milk, Human/metabolism , Neonatal Sepsis/metabolism , Neonatal Sepsis/microbiology , Signal Transduction/immunology , Time Factors
10.
Lab Anim (NY) ; 49(3): 79-88, 2020 03.
Article En | MEDLINE | ID: mdl-32042160

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.


Antigens/metabolism , Colon/immunology , Dendritic Cells/immunology , Goblet Cells/immunology , Immunologic Surveillance , Intestine, Small/immunology , Animals , Antigen Presentation/drug effects , Antigens/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dextrans/administration & dosage , Fluorescent Dyes/administration & dosage , Goblet Cells/drug effects , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Microbiota/immunology , Ovalbumin/administration & dosage , Research Design
11.
Front Immunol ; 11: 603059, 2020.
Article En | MEDLINE | ID: mdl-33613522

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.


Cell Communication , Colon/immunology , Cytokines/blood , Hypersensitivity, Delayed/immunology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Administration, Oral , Adoptive Transfer , Age Factors , Animals , Animals, Genetically Modified , Antigens/administration & dosage , Antigens/immunology , Colon/metabolism , Disease Models, Animal , Hypersensitivity, Delayed/blood , Hypersensitivity, Delayed/genetics , Immune Tolerance , Immunoglobulin E/blood , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Ovalbumin , Phenotype , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Th2 Cells/metabolism , Weaning
12.
Mucosal Immunol ; 13(2): 271-282, 2020 03.
Article En | MEDLINE | ID: mdl-31819172

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.


Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Goblet Cells/immunology , Macrophages/immunology , Mucous Membrane/immunology , T-Lymphocytes, Regulatory/immunology , Administration, Oral , Animals , Antigen Presentation , Antigens/immunology , Cells, Cultured , GTPase-Activating Proteins/metabolism , Immune Tolerance , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
J Allergy Clin Immunol ; 144(4): 1058-1073.e3, 2019 10.
Article En | MEDLINE | ID: mdl-31175877

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.


Allergens/immunology , Anaphylaxis/immunology , Food Hypersensitivity/immunology , Interleukin-13/immunology , Intestinal Mucosa/immunology , Allergens/metabolism , Anaphylaxis/metabolism , Animals , Food Hypersensitivity/metabolism , Humans , Immunoglobulin E/immunology , Interleukin-13/metabolism , Intestinal Mucosa/metabolism , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID
14.
Inflamm Bowel Dis ; 25(1): 180-193, 2019 01 01.
Article En | MEDLINE | ID: mdl-29982468

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.


Anastomosis, Surgical/adverse effects , Colectomy/adverse effects , Colon/surgery , Crohn Disease/surgery , Ileum/surgery , Postoperative Complications/diagnosis , Transcriptome/drug effects , Adult , Antibodies, Monoclonal/therapeutic use , Cohort Studies , Combined Modality Therapy , Crohn Disease/drug therapy , Crohn Disease/pathology , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Male , Postoperative Complications/etiology , Postoperative Complications/genetics , Prognosis , Recurrence , Severity of Illness Index , Tumor Necrosis Factor-alpha/therapeutic use
15.
Birth Defects Res ; 110(20): 1494-1503, 2018 12 01.
Article En | MEDLINE | ID: mdl-30576093

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.


Gastrointestinal Microbiome/physiology , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Female , Homeostasis , Humans , Immune System/physiology , Mothers , Pregnancy
16.
Mucosal Immunol ; 11(6): 1551-1557, 2018 11.
Article En | MEDLINE | ID: mdl-29867079

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.


Goblet Cells/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , Transcytosis , Animals , Antigen Presentation , Antimicrobial Cationic Peptides/metabolism , Homeostasis , Humans , Immunomodulation
17.
Mucosal Immunol ; 11(4): 1103-1113, 2018 07.
Article En | MEDLINE | ID: mdl-29445136

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.


Dendritic Cells/immunology , Goblet Cells/immunology , Mucous Membrane/pathology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation , Antigens/immunology , Cell Proliferation , Dietary Proteins/immunology , Disease Transmission, Infectious , Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Salmonella typhimurium/pathogenicity
18.
Sci Immunol ; 2(18)2017 12 15.
Article En | MEDLINE | ID: mdl-29246946

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.


Antigens, Bacterial/immunology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Immune Tolerance/immunology , Animals , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Weaning
19.
Immunology ; 152(4): 613-627, 2017 12.
Article En | MEDLINE | ID: mdl-28746740

The intestinal lamina propria (LP) contains antigen-presenting cells with features of dendritic cells and macrophages, collectively referred to as mononuclear phagocytes (MNPs). Association of MNPs with the epithelium is thought to play an important role in multiple facets of intestinal immunity including imprinting MNPs with the ability to induce IgA production, inducing the expression of gut homing molecules on T cells, facilitating the capture of luminal antigens and microbes, and subsequent immune responses in the mesenteric lymph node (MLN). However, the factors promoting this process in the steady state are largely unknown, and in vivo models to test and confirm the importance of LP-MNP association with the epithelium for these outcomes are unexplored. Evaluation of epithelial expression of chemoattractants in mice where MNP-epithelial associations were impaired suggested CCL20 as a candidate promoting epithelial association. Expression of CCR6, the only known receptor for CCL20, was required for MNPs to associate with the epithelium. LP-MNPs from CCR6-/- mice did not display defects in acquiring antigen and stimulating T-cell responses in ex vivo assays or in responses to antigen administered systemically. However, LP-MNPs from CCR6-deficient mice were impaired at acquiring luminal and epithelial antigens, inducing IgA production in B cells, inducing immune responses in the MLN, and capturing and trafficking luminal commensal bacteria to the MLN. These findings identify a crucial role for CCR6 in promoting LP-MNPs to associate with the intestinal epithelium in the steady state to perform multiple functions promoting gut immune homeostasis.


Dendritic Cells/immunology , Genomic Imprinting/immunology , Immunologic Surveillance , Intestinal Mucosa/immunology , Macrophages/immunology , Receptors, CCR6/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Chemokine CCL20/genetics , Chemokine CCL20/immunology , Dendritic Cells/cytology , Humans , Macrophages/cytology , Mice , Mice, Knockout , Receptors, CCR6/genetics , T-Lymphocytes/cytology , T-Lymphocytes/immunology
20.
Sci Immunol ; 2(13)2017 Jul 21.
Article En | MEDLINE | ID: mdl-28733471

Specific gut commensal bacteria improve host health by eliciting mutualistic regulatory T (Treg) cell responses. However, the bacteria that induce effector T (Teff) cells during inflammation are unclear. We addressed this by analyzing bacterial-reactive T cell receptor (TCR) transgenic cells and TCR repertoires in a murine colitis model. Unexpectedly, we found that mucosal-associated Helicobacter species triggered both Treg cell responses during homeostasis and Teff cell responses during colitis, as suggested by an increased overlap between the Teff/Treg TCR repertoires with colitis. Four of six Treg TCRs tested recognized mucosal-associated Helicobacter species in vitro and in vivo. By contrast, the marked expansion of luminal Bacteroides species seen during colitis did not trigger a commensurate Teff cell response. Unlike other Treg cell-inducing bacteria, Helicobacter species are known pathobionts and cause disease in immunodeficient mice. Thus, our study suggests a model in which mucosal bacteria elicit context-dependent Treg or Teff cell responses to facilitate intestinal tolerance or inflammation.

...