Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Gastroenterology ; 166(1): 44-58, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37734419

RESUMEN

The gut microbiota plays a significant role in the pathogenesis of both forms of inflammatory bowel disease (IBD), namely, Crohn's disease (CD) and ulcerative colitis (UC). Although evidence suggests dysbiosis and loss of beneficial microbial species can exacerbate IBD, many new studies have identified microbes with pathogenic qualities, termed "pathobionts," within the intestines of patients with IBD. The concept of pathobionts initiating or driving the chronicity of IBD has largely focused on the putative aggravating role that adherent invasive Escherichia coli may play in CD. However, recent studies have identified additional bacterial and fungal pathobionts in patients with CD and UC. This review will highlight the characteristics of these pathobionts and their implications for IBD treatment. Beyond exploring the origins of pathobionts, we discuss those associated with specific clinical features and the potential mechanisms involved, such as creeping fat (Clostridium innocuum) and impaired wound healing (Debaryomyces hansenii) in patients with CD as well as the increased fecal proteolytic activity (Bacteroides vulgatus) seen as a biomarker for UC severity. Finally, we examine the potential impact of pathobionts on current IBD therapies, and several new approaches to target pathobionts currently in the early stages of development. Despite recognizing that pathobionts likely contribute to the pathogenesis of IBD, more work is needed to define their modes of action. Determining whether causal relationships exist between pathobionts and specific disease characteristics could pave the way for improved care for patients, particularly for those not responding to current IBD therapies.


Asunto(s)
Colitis Ulcerosa , Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Humanos , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Inflamatorias del Intestino/microbiología , Colitis Ulcerosa/tratamiento farmacológico , Enfermedad de Crohn/tratamiento farmacológico , Intestinos , Heces
2.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34625492

RESUMEN

Group 3 innate lymphoid cells (ILC3s) control the formation of intestinal lymphoid tissues and play key roles in intestinal defense. They express neuropeptide vasoactive intestinal peptide (VIP) receptor 2 (VPAC2), through which VIP modulates their function, but whether VIP exerts other effects on ILC3 remains unclear. We show that VIP promotes ILC3 recruitment to the intestine through VPAC1 independent of the microbiota or adaptive immunity. VIP is also required for postnatal formation of lymphoid tissues as well as the maintenance of local populations of retinoic acid (RA)-producing dendritic cells, with RA up-regulating gut-homing receptor CCR9 expression by ILC3s. Correspondingly, mice deficient in VIP or VPAC1 suffer a paucity of intestinal ILC3s along with impaired production of the cytokine IL-22, rendering them highly susceptible to the enteric pathogen Citrobacter rodentium This heightened susceptibility to C. rodentium infection was ameliorated by RA supplementation, adoptive transfer of ILC3s, or by recombinant IL-22. Thus, VIP regulates the recruitment of intestinal ILC3s and formation of postnatal intestinal lymphoid tissues, offering protection against enteric pathogens.


Asunto(s)
Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Linfocitos/inmunología , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Animales , Células Dendríticas/inmunología , Microbioma Gastrointestinal/inmunología , Interleucinas/análisis , Tejido Linfoide/citología , Tejido Linfoide/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores CCR/biosíntesis , Receptores de Tipo II del Péptido Intestinal Vasoactivo/genética , Tretinoina/metabolismo , Péptido Intestinal Vasoactivo/genética , Interleucina-22
3.
PLoS Pathog ; 17(8): e1009719, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34352037

RESUMEN

Reducing food intake is a common host response to infection, yet it remains unclear whether fasting is detrimental or beneficial to an infected host. Despite the gastrointestinal tract being the primary site of nutrient uptake and a common route for infection, studies have yet to examine how fasting alters the host's response to an enteric infection. To test this, mice were fasted before and during oral infection with the invasive bacterium Salmonella enterica serovar Typhimurium. Fasting dramatically interrupted infection and subsequent gastroenteritis by suppressing Salmonella's SPI-1 virulence program, preventing invasion of the gut epithelium. Virulence suppression depended on the gut microbiota, as Salmonella's invasion of the epithelium proceeded in fasting gnotobiotic mice. Despite Salmonella's restored virulence within the intestines of gnotobiotic mice, fasting downregulated pro-inflammatory signaling, greatly reducing intestinal pathology. Our study highlights how food intake controls the complex relationship between host, pathogen and gut microbiota during an enteric infection.


Asunto(s)
Bacterias/crecimiento & desarrollo , Ayuno , Gastroenteritis/prevención & control , Inflamación/prevención & control , Intestinos/inmunología , FN-kappa B/antagonistas & inhibidores , Salmonelosis Animal/inmunología , Salmonella typhimurium/fisiología , Animales , Bacterias/inmunología , Bacterias/metabolismo , Femenino , Gastroenteritis/inmunología , Gastroenteritis/microbiología , Microbioma Gastrointestinal , Inflamación/inmunología , Inflamación/microbiología , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , Salmonelosis Animal/complicaciones , Salmonelosis Animal/microbiología , Salmonelosis Animal/patología
4.
Cell Microbiol ; 21(7): e13026, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30893495

RESUMEN

Mammalian cells express an array of toll-like receptors to detect and respond to microbial pathogens, including enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC). These clinically important attaching and effacing (A/E) pathogens infect the apical surface of intestinal epithelial cells, causing inflammation as well as severe diarrheal disease. Because EPEC and EHEC are human-specific, the related murine pathogen Citrobacter rodentium has been widely used to define how hosts defend against A/E pathogens. This study explored the role of TLR9, a receptor that recognises unmethylated CpG dinucleotides present in bacterial DNA, in promoting host defence against C. rodentium. Infected Tlr9-/- mice suffered exaggerated intestinal damage and carried significantly higher (10-100 fold) pathogen burdens in their intestinal tissues as compared with wild type (WT) mice. C. rodentium infection also induced increased antimicrobial responses, as well as hyperactivation of NF-κB signalling in the intestines of Tlr9-/- mice. These changes were associated with accelerated depletion of the intestinal microbiota in Tlr9-/- mice as compared with WT mice. Notably, antibiotic-based depletion of the gut microbiota in WT mice prior to infection increased their susceptibility to the levels seen in Tlr9-/- mice. Our results therefore indicate that TLR9 signalling suppresses intestinal antimicrobial responses, thereby promoting microbiota-mediated colonisation resistance against C. rodentium infection.


Asunto(s)
Citrobacter rodentium/genética , Infecciones por Enterobacteriaceae/genética , Microbioma Gastrointestinal/genética , Receptor Toll-Like 9/genética , Animales , Antibacterianos/farmacología , Antiinfecciosos/farmacología , Citrobacter rodentium/patogenicidad , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Escherichia coli Enterohemorrágica/genética , Escherichia coli Enterohemorrágica/patogenicidad , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/patogenicidad , Interacciones Huésped-Patógeno/efectos de los fármacos , Ratones , FN-kappa B/genética
5.
Dysphagia ; 35(5): 773-779, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31773332

RESUMEN

Eosinophilic esophagitis (EoE) is a chronic condition that requires repeated endoscopies/biopsies to track the disease and treatment response. This invasive procedure involves risk to the patient and has significant costs. We studied whether the detection of specific proteins (cytokines and eosinophil degranulation products) from oral swabs could serve as a minimally invasive test for EoE. Swabs of the oral cavity (buccal and oropharyngeal) were obtained prior to endoscopy/biopsies in patients with EoE, possible EoE, and non-EoE patients in addition to obtaining additional esophageal biopsy tissue. ELISAs measuring the levels of cytokines IL-5, IL-8, IL-13, and eosinophil degranulation products including major basic protein (MBP), eosinophil derived neurotoxin (EDN), and eosinophil peroxidase (EPO) were performed on the samples. Comparisons were made to peak esophageal eosinophil counts. Tolerability of the swabs was evaluated. 43 patients, 4-17 years old, participated in the study. Swabs were well tolerated and all showed measurable protein. 26 patients had EoE [14 active (> 15 eosinophils/high power field), 12 non-active], 17 patients did not have EoE. Results obtained from oral swabs showed poor correlation with those from esophageal tissue. Only measurement of eosinophil degranulation products EDN and EPO from esophageal tissues showed strong correlations with eosinophil counts. In this study, the levels of cytokines and eosinophil degranulation products detected from oral swabs did not correlate with esophageal eosinophilia, and their detection would be insufficient to displace endoscopy/biopsies.


Asunto(s)
Esofagitis Eosinofílica , Eosinófilos , Adolescente , Biomarcadores , Niño , Preescolar , Neurotoxina Derivada del Eosinófilo , Esofagitis Eosinofílica/diagnóstico , Humanos
6.
PLoS Pathog ; 11(8): e1005108, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26285214

RESUMEN

Enterohemorrhagic Escherichia coli and related food and waterborne pathogens pose significant threats to human health. These attaching/effacing microbes infect the apical surface of intestinal epithelial cells (IEC), causing severe diarrheal disease. Colonizing the intestinal luminal surface helps segregate these microbes from most host inflammatory responses. Based on studies using Citrobacter rodentium, a related mouse pathogen, we speculate that hosts rely on immune-mediated changes in IEC, including goblet cells to defend against these pathogens. These changes include a CD4+ T cell-dependent increase in IEC proliferation to replace infected IEC, as well as altered production of the goblet cell-derived mucin Muc2. Another goblet cell mediator, REsistin-Like Molecule (RELM)-ß is strongly induced within goblet cells during C. rodentium infection, and was detected in the stool as well as serum. Despite its dramatic induction, RELM-ß's role in host defense is unclear. Thus, wildtype and RELM-ß gene deficient mice (Retnlb-/-) were orally infected with C. rodentium. While their C. rodentium burdens were only modestly elevated, infected Retnlb-/- mice suffered increased mortality and mucosal ulceration due to deep pathogen penetration of colonic crypts. Immunostaining for Ki67 and BrDU revealed Retnlb-/- mice were significantly impaired in infection-induced IEC hyper-proliferation. Interestingly, exposure to RELM-ß did not directly increase IEC proliferation, rather RELM-ß acted as a CD4+ T cell chemoattractant. Correspondingly, Retnlb-/- mice showed impaired CD4+ T cell recruitment to their infected colons, along with reduced production of interleukin (IL)-22, a multifunctional cytokine that directly increased IEC proliferation. Enema delivery of RELM-ß to Retnlb-/- mice restored CD4+ T cell recruitment, concurrently increasing IL-22 levels and IEC proliferation, while reducing mucosal pathology. These findings demonstrate that RELM-ß and goblet cells play an unexpected, yet critical role in recruiting CD4+ T cells to the colon to protect against an enteric pathogen, in part via the induction of increased IEC proliferation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Colitis/inmunología , Células Caliciformes/inmunología , Hormonas Ectópicas/inmunología , Mucosa Intestinal/inmunología , Animales , Separación Celular , Citrobacter rodentium , Colitis/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Caliciformes/metabolismo , Hormonas Ectópicas/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa
7.
Infect Immun ; 84(12): 3608-3617, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27736776

RESUMEN

At present, there is no vaccine for enterotoxigenic Escherichia coli (ETEC), an important cause of diarrheal illness. Nevertheless, recent microbial pathogenesis studies have identified a number of molecules produced by ETEC that contribute to its virulence and are novel antigenic targets to complement canonical vaccine approaches. EtpA is a secreted two-partner adhesin that is conserved within the ETEC pathovar. EtpA interacts with the tips of ETEC flagella to promote bacterial adhesion, toxin delivery, and intestinal colonization by forming molecular bridges between the bacteria and the epithelial surface. However, the nature of EtpA interactions with the intestinal epithelium remains poorly defined. Here, we demonstrate that EtpA interacts with glycans presented by transmembrane and secreted intestinal mucins at epithelial surfaces to facilitate pathogen-host interactions that culminate in toxin delivery. Moreover, we found that a major effector molecule of ETEC, the heat-labile enterotoxin (LT), may enhance these interactions by stimulating the production of the gel-forming mucin MUC2. Our studies suggest, however, that EtpA participates in complex and dynamic interactions between ETEC and the gastrointestinal mucosae in which host glycoproteins promote bacterial attachment while simultaneously limiting the epithelial engagement required for effective toxin delivery. Collectively, these data provide additional insight into the intricate nature of ETEC interactions with the intestinal epithelium that have potential implications for rational approaches to vaccine design.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Escherichia coli Enterotoxigénica/fisiología , Mucosa Intestinal/microbiología , Mucinas/metabolismo , Acetilgalactosamina/metabolismo , Adhesinas Bacterianas/genética , Animales , Células CACO-2 , Escherichia coli Enterotoxigénica/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Células HT29 , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucina 2/genética , Mucina 2/metabolismo , Interferencia de ARN
8.
Infect Immun ; 83(8): 3257-67, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26034212

RESUMEN

Attaching and effacing pathogens, including enterohemorrhagic Escherichia coli in humans and Citrobacter rodentium in mice, raise serious public health concerns. Here we demonstrate that interleukin-1 receptor (IL-1R) signaling is indispensable for protection against C. rodentium infection in mice. Four days after infection with C. rodentium, there were significantly fewer neutrophils (CD11b+ Ly6C+ Ly6G+) in the colons of IL-1R−/− mice than in wild-type mice. Levels of mRNA and protein of KC/CXCL1 were also significantly reduced in colon homogenates of infected IL-1R−/− mice relative to wild-type mice. Of note, infiltrated CD11b+ Ly6C+ Ly6G+ neutrophils were the main source of IL-22 secretion after C. rodentium infection. Interestingly, intestinal stromal cells isolated from IL-1R−/− mice secreted lower levels of KC/CXCL1 than stromal cells from wild-type mice during C. rodentium infection. Similar effects were found when mouse intestinal stromal cells and human nasal polyp stromal cells were treated with IL-1R antagonists (i.e., anakinra) in vitro. These results suggest that IL-1 signaling plays a pivotal role in activating mucosal stromal cells to secrete KC/CXCL1, which is essential for infiltration of IL-22-secreting neutrophils upon bacterial infection.


Asunto(s)
Quimiocina CXCL1/metabolismo , Citrobacter rodentium/fisiología , Infecciones por Enterobacteriaceae/metabolismo , Interleucina-1/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Neutrófilos/metabolismo , Células del Estroma/metabolismo , Animales , Células Cultivadas , Quimiocina CXCL1/genética , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/microbiología , Femenino , Humanos , Interleucina-1/genética , Interleucinas/genética , Intestinos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Transducción de Señal , Interleucina-22
9.
J Infect Dis ; 207(1): 50-60, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23100560

RESUMEN

Interleukin (IL)-1 is a well-known cytokine for the initiation of innate immunity in bacterial infection. However, the underlying mechanism of IL-1 on the respiratory infection is not fully elucidated. We studied how IL-1 contributes to the host defense against Streptococcus pneumoniae. IL-1R(-/-) mice showed high mortality, local cytokine storm, and substantial infiltrates in the lower respiratory tract after intratracheal challenge with S. pneumoniae. The IL-1-deficient condition did not suppress the propagation of bacteria in the lung, although the recruitment and the bacteria-killing ability of neutrophils (CD11b(+)Ly6C(+)Ly6G(+)) were not defective compared with wild-type mice. Unexpectedly, we found that the transcription of fibrinogen alpha and gamma genes were highly activated in the lungs of wild-type mice after the infection, whereas no significant changes were found in IL-1R(-/-) mice. Of note, synthesis of fibrinogen was dependent on the IL-1-IL-6-Stat3 cascade. Treatment with recombinant fibrinogen improved survival and bacterial propagation in the IL-1R(-/-) mice and blockade of the coagulation increased the susceptibility of wild-type mice to pneumococcal pneumonia. Our findings suggest that IL-1 signaling leads to the synthesis of fibrinogen in the lung after pneumococcus infection and is followed by coagulation, which contributes to the control of bacterial infection in the pulmonary tract.


Asunto(s)
Coagulación Sanguínea/inmunología , Interleucina-1/inmunología , Pulmón/inmunología , Neumonía Neumocócica/inmunología , Streptococcus pneumoniae/inmunología , Animales , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Fibrinógeno/metabolismo , Inmunidad Innata , Pulmón/metabolismo , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Mutación , Neutrófilos/inmunología , Neumonía Neumocócica/tratamiento farmacológico , Neumonía Neumocócica/microbiología , Neumonía Neumocócica/mortalidad , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Proteínas Recombinantes , Transducción de Señal , Organismos Libres de Patógenos Específicos , Streptococcus pneumoniae/efectos de los fármacos , Análisis de Supervivencia , Trombina/metabolismo
10.
Eur J Immunol ; 42(3): 618-28, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22105301

RESUMEN

The role of TLR signaling in linking the innate and adaptive immune systems has been a controversial issue that remains to be solved. Here, we determined whether MyD88-dependent TLR signals are required for the generation of B-cell responses during chronic Salmonella infection. Oral administration of recombinant attenuated Salmonella enterica serovar Typhimurium vaccine (RASV) strain in MyD88(-/-) mice resulted in chronic infection. Infection was accompanied by enlarged germinal centers and hypergammaglobulinemia with anti-double-stranded DNA (dsDNA)-specific Ab in sera, and the deposition of immune complexes in the kidneys, suggesting onset of autoimmunity. CD4(+) T cells expressing PD-1, CXCR5, ICOS, and IL-21 were dramatically increased in chronically infected mice, indicating the expansion of follicular helper T (Tfh)-like cells. Of note, the depletion of CD4(+) T cells completely blocked the generation of polyclonal IgG Ab in sera after oral RASV challenge. Inflammatory myeloid cells expressing CD11b and Gr-1 accumulated in high numbers in the spleen of MyD88(-/-) mice. Interestingly, the blockade of PD-1 or ICOS significantly reduced the hypergammaglobulinemia and dsDNA-specific autoantibody production. Overall, these results suggest that Tfh-like cells in chronic bacterial infection trigger autoimmune hypergammaglobulinemia in a PD-1- and ICOS-dependent manner.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos B/inmunología , Hipergammaglobulinemia/microbiología , Factor 88 de Diferenciación Mieloide/deficiencia , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/inmunología , Hipergammaglobulinemia/inmunología , Inmunoglobulina G/sangre , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Interleucinas/economía , Interleucinas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/inmunología , Receptor de Muerte Celular Programada 1 , ARN/química , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR5/genética , Receptores CXCR5/inmunología , Infecciones por Salmonella/microbiología , Organismos Libres de Patógenos Específicos
11.
Gastroenterology ; 138(4): 1468-78, 1478.e1-6, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19914251

RESUMEN

BACKGROUND & AIMS: Retinoic acid (RA) is a crucial factor for maintaining homeostasis in the gut, including lymphocyte homing, immunoglobulin (Ig) A production, and T regulatory cells (Treg) and T helper cell 17 (T(H)17) generation. Until now, most attention has focused on the function of dendritic cells (DCs) to initiate adaptive immunity including T and B lymphocytes through RA. To investigate the effects of RA on DCs of gut-associated lymphoid tissue (GALT), we analyzed the phenotype and function of DC subsets from GALT of vitamin A-deficient (VAD) mice. METHOD: VAD mice were prepared by feeding them a VAD diet over 12 weeks from gestational days 10-14. RESULTS: Here, we report that tremendous increase of langerin(+) DCs occurred in the mesenteric lymph nodes (MLNs) and gut lamina propria of VAD mice dependent on CCR7 signaling. Langerin(+) DCs have phenotypes more similar to those of bone marrow-derived dermal langerin(+) DCs than epidermal Langerhans cells. Moreover, RA receptor antagonists enhance the differentiation of langerin(+) DCs from mouse and human precursors of bone marrow and peripheral blood. Langerin(+) DCs were highly differentiated but less inflammatory than langerin(-) DCs of MLNs of VAD mice. Moreover, tolerance to orally delivered antigen was completely abrogated by depletion of langerin(+) DCs in the VAD mice. CONCLUSIONS: These results suggest that generation of langerin(+) DCs in the GALT is tightly regulated by RA and that the microenvironment of tissues determines the phenotype of DCs.


Asunto(s)
Antígenos de Superficie/fisiología , Células Dendríticas/inmunología , Intestinos/inmunología , Lectinas Tipo C/fisiología , Tejido Linfoide/inmunología , Lectinas de Unión a Manosa/fisiología , Tretinoina/fisiología , Aldehído Oxidorreductasas/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Tolerancia Inmunológica , Células de Langerhans/citología , Células de Langerhans/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores CCR7/fisiología , Deficiencia de Vitamina A/inmunología
12.
J Immunol ; 182(4): 2305-12, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19201885

RESUMEN

Salmonella organisms are Gram negative and facultative anaerobic bacteria that cause typhoid fever in humans. In this study, we evaluated LPS-specific adaptive immunity in innate immune-deficient mice after oral administration of attenuated Salmonella enterica serovar Typhimurium (S. Typhimurium) strains. Of interest, identical levels of LPS-specific IgG and IgA Abs were elicited in the systemic (i.e., serum and spleen) and mucosal (i.e., fecal extract and small intestine) compartments of wild-type, TLR4(-/-), and MyD88(-/-) mice following oral vaccination with recombinant attenuated S. Typhimurium (RASV). Depletion of CD4(+) T cells during RASV vaccination completely abrogated the generation of LPS-specific Abs in MyD88(-/-) mice. In addition, mRNA expression levels of a B cell-activating factor of the TNF family were significantly increased in the spleens of MyD88(-/-) mice after oral administration, implying that T cell-independent B cell switching might be also enhanced in the MyD88 signal-deficient condition. Of most interest, orally vaccinated MyD88(-/-) mice that possessed high levels of LPS-specific IgG and IgA, which had a neutralizing effect against Salmonella, died earlier than nonvaccinated wild-type mice following lethal oral challenge with virulent Salmonella species. These results suggest that innate immunity mediated by MyD88 signal is dispensable for induction of LPS-specific Ab responses following oral administration of attenuated Salmonella strains but indispensable for efficient protection.


Asunto(s)
Linfocitos B/inmunología , Inmunidad Innata/inmunología , Lipopolisacáridos/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Infecciones por Salmonella/inmunología , Vacunas contra la Salmonella/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Linfocitos T CD4-Positivos/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
13.
J Mol Med (Berl) ; 98(12): 1781-1794, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33128578

RESUMEN

Autotaxin (ATX) is a secreted enzyme that hydrolyzes lysophosphatidylcholine (LPC) to lysophosphatidic acid (LPA) and choline. ATX has been implicated in multiple chronic inflammatory diseases, but little is known about its role in the development of inflammatory bowel disease (IBD). Here, we investigated how ATX contributed to intestinal inflammation during colitis. We found that ATX expression levels were upregulated in the intestines of ulcerative colitis (UC) patients in acute state as well as in the intestines of dextran sulfate sodium (DSS)-induced colitis mice, which is likely due to increased infiltration of inflammatory cells including macrophages. Intriguingly, the inhibition of ATX activity led to reduced production of inflammatory cytokines, as well as attenuated colitis. These findings suggest that ATX may display strong pro-inflammatory properties. Supporting this, treatment with recombinant mouse ATX (rmATX) increased the production of inflammatory cytokines and enzymes in mouse macrophage cell line RAW264.7 and bone marrow-derived macrophages (BMDM), whereas silencing ATX by siRNA reduced LPS-stimulated production of pro-inflammatory factors. Notably, we found that the levels of LPA2 (an LPA receptor) were dramatically upregulated in rmATX-treated RAW264.7 cells and DSS-treated mice. Gene silencing of lpa2 in RAW264.7 cells by siRNA led to reduced production of inflammatory cytokines. Moreover, adenovirus-mediated delivery of lpa2 short hairpin RNA into DSS-treated mice ameliorated colitis. Collectively, our research suggests that ATX may exacerbate DSS-induced colitis by activating LPA2 receptor in macrophages and represent a promising target for the treatment of IBD. KEY MESSAGES: Increased ATX expression and secretion in colitic colons are likely due to increased infiltration of inflammatory cells including macrophages. Recombinant ATX promotes, but ATX silencing inhibits, the production of inflammatory cytokines in LPS-stimulated RAW264.7 cells and BMDM. •LPA2 mediates the pro-inflammatory effects of ATX on macrophages. Inhibition of ATX and downregulation of LPA2 ameliorate DSS-induced colitis.


Asunto(s)
Colitis/etiología , Colitis/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Receptores del Ácido Lisofosfatídico/agonistas , Animales , Colitis/patología , Citocinas/metabolismo , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Mediadores de Inflamación/metabolismo , Ratones , Hidrolasas Diéster Fosfóricas/genética , Células RAW 264.7
14.
Gut Microbes ; 12(1): 1847976, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33258388

RESUMEN

Ulcerative colitis (UC) is a chronic inflammatory condition linked to intestinal microbial dysbiosis, including the expansion of E. coli strains related to extra-intestinal pathogenic E. coli. These "pathobionts" exhibit pathogenic properties, but their potential to promote UC is unclear due to the lack of relevant animal models. Here, we established a mouse model using a representative UC pathobiont strain (p19A), and mice lacking single immunoglobulin and toll-interleukin 1 receptor domain (SIGIRR), a deficiency increasing susceptibility to gut infections. Strain p19A was found to adhere to the cecal mucosa of Sigirr -/- mice, causing modest inflammation. Moreover, it dramatically worsened dextran sodium sulfate-induced colitis. This potentiation was attenuated using a p19A strain lacking α-hemolysin genes, or when we targeted pathobiont adherence using a p19A strain lacking the adhesin FimH, or following treatment with FimH antagonists. Thus, UC pathobionts adhere to the intestinal mucosa, and worsen the course of colitis in susceptible hosts.


Asunto(s)
Colitis Ulcerosa/genética , Colitis Ulcerosa/microbiología , Escherichia coli/crecimiento & desarrollo , Microbioma Gastrointestinal , Adhesinas de Escherichia coli/genética , Adhesinas de Escherichia coli/metabolismo , Animales , Colitis Ulcerosa/inmunología , Modelos Animales de Enfermedad , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología
15.
mBio ; 10(6)2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31848276

RESUMEN

The Gram-negative marine bacterium Vibrio parahaemolyticus is a common cause of infectious gastroenteritis due to the ingestion of contaminated seafood. Most virulent V. parahaemolyticus strains encode two type III secretion systems (T3SS1 and T3SS2); however, the roles they and their translocated effectors play in causing intestinal disease remain unclear. While studies have identified T3SS1 effectors as responsible for killing epithelial cells in culture, the T3SS2 effectors caused massive epithelial cell disruption in a rabbit ileal loop model. Additional models are thus needed to clarify the pathogen-host interactions that drive V. parahaemolyticus-associated gastroenteritis. Germfree mice were infected with a pathogenic clinical isolate of V. parahaemolyticus, RIMD2210633 (RIMD). The pathogen was found to adhere to as well as invade the cecal mucosa, accompanied by severe inflammation and dramatic mucosal damage, including widespread sloughing of infected epithelial cells. Mice infected with a V. parahaemolyticus strain lacking the T3SS1 (POR2) also developed severe pathology, similar to that seen with RIMD. In contrast, the ΔT3SS2 strain (POR3) appeared unable to invade the intestinal mucosa or cause any mucosal pathology. Confirming a role for TS332 effectors, a strain expressing the T3SS2 but lacking VopC (POR2ΔvopC), a T3SS2 effector implicated in epithelial cell invasion in culture, was strongly attenuated in invading the intestinal mucosa and in causing gastroenteritis, although infection with this mutant resulted in more pathology than the ΔT3SS2 strain. We thus present an experimental system that enables further characterization of T3SS effectors as well as the corresponding host inflammatory response involved in the gastroenteritis caused by invasive V. parahaemolyticusIMPORTANCEVibrio parahaemolyticus causes severe gastroenteritis following consumption of contaminated seafood. Global warming has allowed this pathogen to spread worldwide, contributing to recent outbreaks. Clinical isolates are known to harbor an array of virulence factors, including T3SS1 and T3SS2; however, the precise role these systems play in intestinal disease remains unclear. There is an urgent need to improve our understanding of how V. parahaemolyticus infects hosts and causes disease. We present a novel mouse model for this facultative intracellular pathogen and observe that the T3SS2 is essential to pathogenicity. Moreover, we show that the T3SS2 effector VopC, previously shown to be a Rac and Cdc42 deamidase that facilitates bacterial uptake by nonphagocytic cells, also plays a key role in the ability of V. parahaemolyticus to invade the intestinal mucosa and cause gastroenteritis. This experimental model thus provides a valuable tool for future elucidation of virulence mechanisms used by this facultative intracellular pathogen during in vivo infection.


Asunto(s)
Gastroenteritis/microbiología , Sistemas de Secreción Tipo III , Vibriosis/microbiología , Vibrio parahaemolyticus/fisiología , Factores de Virulencia/genética , Animales , Antibacterianos/farmacología , Muerte Celular , Proliferación Celular , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Gastroenteritis/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Ratones , Estreptomicina/farmacología , Vibrio parahaemolyticus/efectos de los fármacos , Virulencia
16.
Sci Rep ; 9(1): 8397, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31182790

RESUMEN

Inhibitor of kappa B (IκB)-ζ transcription is rapidly induced by stimulation with TLR ligands and IL-1. Despite high IκBζ expression in inflammation sites, the association of IκBζ with host defence via systemic immune responses against bacterial infection remains unclear. Oral immunisation with a recombinant attenuated Salmonella vaccine (RASV) strain did not protect IκBζ-deficient mice against a lethal Salmonella challenge. IκBζ-deficient mice failed to produce Salmonella LPS-specific IgG, especially IgG2a, although inflammatory cytokine production and immune cell infiltration into the liver increased after oral RASV administration. Moreover, IκBζ-deficient mice exhibited enhanced splenic germinal centre reactions followed by increased total IgG production, despite IκBζ-deficient B cells having an intrinsic antibody class switching defect. IκBζ-deficient CD4+ T cells poorly differentiated into Th1 cells. IFN-γ production by CD4+ T cells from IκBζ-deficient mice immunised with RASV significantly decreased after restimulation with heat-killed RASV in vitro, suggesting that IκBζ-deficient mice failed to mount protective immune responses against Salmonella infection because of insufficient Th1 and IgG production. Therefore, IκBζ is crucial in protecting against Salmonella infection by inducing Th1 differentiation followed by IgG production.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular , Inmunidad , Inmunoglobulina G/biosíntesis , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/prevención & control , Células TH1/inmunología , Administración Oral , Animales , Enfermedad Crónica , Centro Germinal/metabolismo , Inmunización , Inflamación/patología , Interferón gamma/metabolismo , Lipopolisacáridos , Ratones Endogámicos C57BL , Infecciones por Salmonella/parasitología , Vacunas contra la Salmonella/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Vacunas Atenuadas/inmunología , Virulencia
17.
Artículo en Inglés | MEDLINE | ID: mdl-30410551

RESUMEN

BACKGROUND: Ginseng is a perennial herb used in traditional Chinese medicine, which has become increasingly popular world-wide due to its proposed medicinal effects. There are two major species of ginseng, Panax ginseng (Korean or Asian ginseng), and Panax quinquefolius (American ginseng). Although cases of allergy due to Korean ginseng have been reported in adults, there are no reported cases of allergy to American ginseng, and no reported cases of ginseng allergy in pediatric patients. CASE PRESENTATION: We present two unique cases of pediatric patients with suspected allergic reactions to American ginseng. The first patient is a 6-year-old girl who presented to the emergency department in anaphylaxis (urticaria and respiratory symptoms) minutes after inhaling powdered American ginseng. There was evidence of sensitization to American ginseng on skin prick testing (SPT) (13 × 12 mm wheal) and evidence of allergy to American ginseng on basophil activation testing, with a dose-dependent increase in expression of CD63 on basophils in response to American ginseng extract. The second patient is a 3-year-old boy who presented with recurrent allergic conjunctivitis upon exposure to aerosolized powdered ginseng, with evidence of sensitization to American ginseng on SPT (13 × 7 mm wheal), but with no evidence of IgE-mediated allergic reaction during oral challenge with American ginseng powder. CONCLUSIONS: These cases highlight two different allergic responses to American ginseng in pediatric patients. To our knowledge, these are the first reported cases of allergy to American ginseng, in addition to the first reported cases of allergy to ginseng in pediatric patients.

18.
Front Immunol ; 9: 2211, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319652

RESUMEN

Background: Current ulcerative colitis (UC) treatments are focused on symptom management primarily via immune suppression. Despite the current arsenal of immunosuppressant treatments, the majority of patients with UC still experience disease progression. Importantly, aggressive long-term inhibition of immune function comes with consequent risk, such as serious infections and malignancy. There is thus a recognized need for new, safe and effective treatment strategies for people living with UC that work upstream of managing the symptoms of the disease. The objective of this study was to evaluate a microbial-based treatment, QBECO, that functions to productively activate rather than suppress mucosal immune function as a novel approach to treat UC. Methods: Two established models of experimental colitis, namely chemically-induced DSS colitis and the spontaneous colitis that develops in Muc2 deficient mice, were used to assess whether QBECO treatment could ameliorate gastrointestinal disease. A small exploratory 16-week QBECO open-label trial was subsequently conducted to test the safety and tolerability of this approach and also to determine whether similar improvements in clinical disease and histopathology could be demonstrated in patients with moderate-to-severe UC. Results: QBECO treatment successfully reduced inflammation and promoted mucosal and histological healing in both experimental models and in UC patients. The preclinical models of colitis showed that QBECO ameliorated mucosal pathology, in part by reducing inflammatory cell infiltration, primarily that induced by neutrophils and inflammatory T cells. The most rapid and noticeable change observed in QBECO treated UC patients was a marked reduction in rectal bleeding. Conclusion: Collectively, this work demonstrates for the first time that strategically activating immune function rather than suppressing it, not only does not worsen colitis induced-damage, but may lead to an objective reduction in UC disease pathology.


Asunto(s)
Colitis Ulcerosa/terapia , Escherichia coli/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos , Mucosa Intestinal/metabolismo , Adulto , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/genética , Colitis Ulcerosa/inmunología , Colon/inmunología , Colon/metabolismo , Colon/microbiología , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Femenino , Humanos , Inyecciones Subcutáneas , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucina 2/genética , Resultado del Tratamiento , Adulto Joven
19.
Cell Host Microbe ; 16(2): 249-256, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25121752

RESUMEN

Inflammasome-mediated host defenses have been extensively studied in innate immune cells. Whether inflammasomes function for innate defense in intestinal epithelial cells, which represent the first line of defense against enteric pathogens, remains unknown. We observed enhanced Salmonella enterica serovar Typhimurium colonization in the intestinal epithelium of caspase-11-deficient mice, but not at systemic sites. In polarized epithelial monolayers, siRNA-mediated depletion of caspase-4, a human ortholog of caspase-11, also led to increased bacterial colonization. Decreased rates of pyroptotic cell death, a host defense mechanism that extrudes S. Typhimurium-infected cells from the polarized epithelium, accounted for increased pathogen burdens. The caspase-4 inflammasome also governs activation of the proinflammatory cytokine, interleukin (IL)-18, in response to intracellular (S. Typhimurium) and extracellular (enteropathogenic Escherichia coli) enteric pathogens, via intracellular LPS sensing. Therefore, an epithelial cell-intrinsic noncanonical inflammasome plays a critical role in antimicrobial defense at the intestinal mucosal surface.


Asunto(s)
Caspasas Iniciadoras/metabolismo , Caspasas/metabolismo , Infecciones por Escherichia coli/enzimología , Inflamasomas/fisiología , Infecciones por Salmonella/enzimología , Animales , Línea Celular Tumoral , Escherichia coli Enteropatógena/inmunología , Activación Enzimática , Infecciones por Escherichia coli/inmunología , Gastroenteritis/enzimología , Gastroenteritis/microbiología , Humanos , Interleucina-18/metabolismo , Mucosa Intestinal/enzimología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Salmonella/inmunología , Salmonella enterica/inmunología
20.
Vaccine ; 30(48): 6816-23, 2012 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-23000127

RESUMEN

Influenza infection followed by pneumococcal infection can cause severe pneumonia and this secondary pneumococcal pneumonia is the most common cause of influenza-associated death. Therefore, vaccination against Streptococcus pneumoniae is highly desirable for reducing the disease burden caused by seasonal epidemic and pandemic influenza. In this study, mice were vaccinated orally with a recombinant attenuated Salmonella vaccine (RASV) strain that delivers pneumococcal surface protein A (PspA) in order to examine protective efficacy against secondary pneumococcal pneumonia. A single dose of oral RASV resulted in attenuated pulmonary inflammation and effective protection against secondary pneumococcal pneumonia. Additionally, oral RASV induced long-term protection against secondary pneumococcal pneumonia. Treatment with an antiviral drug (i.e., oseltamivir) after treatment with RASV further prevented pulmonary inflammation after secondary pneumococcal infection. These results imply that oral RASV can protect mice from secondary pneumococcal infection after influenza infection and that vaccination against respiratory bacterial pathogens is a promising approach for dampening the impact of annual epidemic and pandemic influenza outbreaks.


Asunto(s)
Proteínas Bacterianas/inmunología , Vacunas Neumococicas/inmunología , Neumonía Neumocócica/prevención & control , Administración Oral , Animales , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/complicaciones , Vacunas Neumococicas/administración & dosificación , Neumonía Neumocócica/inmunología , Vacunas contra la Salmonella/administración & dosificación , Vacunas contra la Salmonella/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA