Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
3.
Nature ; 603(7902): 672-678, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35296857

RESUMEN

The fungal microbiota (mycobiota) is an integral part of the complex multikingdom microbial community colonizing the mammalian gastrointestinal tract and has an important role in immune regulation1-6. Although aberrant changes in the mycobiota have been linked to several diseases, including inflammatory bowel disease3-9, it is currently unknown whether fungal species captured by deep sequencing represent living organisms and whether specific fungi have functional consequences for disease development in affected individuals. Here we developed a translational platform for the functional analysis of the mycobiome at the fungal-strain- and patient-specific level. Combining high-resolution mycobiota sequencing, fungal culturomics and genomics, a CRISPR-Cas9-based fungal strain editing system, in vitro functional immunoreactivity assays and in vivo models, this platform enables the examination of host-fungal crosstalk in the human gut. We discovered a rich genetic diversity of opportunistic Candida albicans strains that dominate the colonic mucosa of patients with inflammatory bowel disease. Among these human-gut-derived isolates, strains with high immune-cell-damaging capacity (HD strains) reflect the disease features of individual patients with ulcerative colitis and aggravated intestinal inflammation in vivo through IL-1ß-dependent mechanisms. Niche-specific inflammatory immunity and interleukin-17A-producing T helper cell (TH17 cell) antifungal responses by HD strains in the gut were dependent on the C. albicans-secreted peptide toxin candidalysin during the transition from a benign commensal to a pathobiont state. These findings reveal the strain-specific nature of host-fungal interactions in the human gut and highlight new diagnostic and therapeutic targets for diseases of inflammatory origin.


Asunto(s)
Hongos , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Microbiota , Micobioma , Animales , Sistemas CRISPR-Cas , Candida albicans , Hongos/genética , Hongos/patogenicidad , Variación Genética , Humanos , Inmunidad , Inflamación , Mamíferos
4.
Cell ; 185(5): 831-846.e14, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35176228

RESUMEN

Fungal communities (the mycobiota) are an integral part of the gut microbiota, and the disruption of their integrity contributes to local and gut-distal pathologies. Yet, the mechanisms by which intestinal fungi promote homeostasis remain unclear. We characterized the mycobiota biogeography along the gastrointestinal tract and identified a subset of fungi associated with the intestinal mucosa of mice and humans. Mucosa-associated fungi (MAF) reinforced intestinal epithelial function and protected mice against intestinal injury and bacterial infection. Notably, intestinal colonization with a defined consortium of MAF promoted social behavior in mice. The gut-local effects on barrier function were dependent on IL-22 production by CD4+ T helper cells, whereas the effects on social behavior were mediated through IL-17R-dependent signaling in neurons. Thus, the spatial organization of the gut mycobiota is associated with host-protective immunity and epithelial barrier function and might be a driver of the neuroimmune modulation of mouse behavior through complementary Type 17 immune mechanisms.


Asunto(s)
Microbioma Gastrointestinal , Micobioma , Receptores de Interleucina-17/metabolismo , Conducta Social , Animales , Hongos , Inmunidad Mucosa , Mucosa Intestinal , Ratones , Membrana Mucosa
5.
Nat Microbiol ; 6(12): 1493-1504, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34811531

RESUMEN

Secretory immunoglobulin A (sIgA) plays an important role in gut barrier protection by shaping the resident microbiota community, restricting the growth of bacterial pathogens and enhancing host protective immunity via immunological exclusion. Here, we found that a portion of the microbiota-driven sIgA response is induced by and directed towards intestinal fungi. Analysis of the human gut mycobiota bound by sIgA revealed a preference for hyphae, a fungal morphotype associated with virulence. Candida albicans was a potent inducer of IgA class-switch recombination among plasma cells, via an interaction dependent on intestinal phagocytes and hyphal programming. Characterization of sIgA affinity and polyreactivity showed that hyphae-associated virulence factors were bound by these antibodies and that sIgA influenced C. albicans morphotypes in the murine gut. Furthermore, an increase in granular hyphal morphologies in patients with Crohn's disease compared with healthy controls correlated with a decrease in antifungal sIgA antibody titre with affinity to two hyphae-associated virulence factors. Thus, in addition to its importance in gut bacterial regulation, sIgA targets the uniquely fungal phenomenon of hyphal formation. Our findings indicate that antifungal sIgA produced in the gut can play a role in regulating intestinal fungal commensalism by coating fungal morphotypes linked to virulence, thereby providing a protective mechanism that might be dysregulated in patients with Crohn's disease.


Asunto(s)
Enfermedad de Crohn/microbiología , Hongos/fisiología , Microbioma Gastrointestinal , Inmunoglobulina A Secretora/inmunología , Simbiosis , Animales , Candida albicans/genética , Candida albicans/fisiología , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Femenino , Hongos/genética , Interacciones Huésped-Patógeno , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitos/inmunología , Fagocitos/microbiología
6.
Cell ; 184(4): 1017-1031.e14, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33548172

RESUMEN

Antibodies mediate natural and vaccine-induced immunity against viral and bacterial pathogens, whereas fungi represent a widespread kingdom of pathogenic species for which neither vaccine nor neutralizing antibody therapies are clinically available. Here, using a multi-kingdom antibody profiling (multiKAP) approach, we explore the human antibody repertoires against gut commensal fungi (mycobiota). We identify species preferentially targeted by systemic antibodies in humans, with Candida albicans being the major inducer of antifungal immunoglobulin G (IgG). Fungal colonization of the gut induces germinal center (GC)-dependent B cell expansion in extraintestinal lymphoid tissues and generates systemic antibodies that confer protection against disseminated C. albicans or C. auris infection. Antifungal IgG production depends on the innate immunity regulator CARD9 and CARD9+CX3CR1+ macrophages. In individuals with invasive candidiasis, loss-of-function mutations in CARD9 are associated with impaired antifungal IgG responses. These results reveal an important role of gut commensal fungi in shaping the human antibody repertoire through CARD9-dependent induction of host-protective antifungal IgG.


Asunto(s)
Anticuerpos Antifúngicos/inmunología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Inmunidad , Inmunoglobulina G/inmunología , Micobioma/inmunología , Animales , Linfocitos B/inmunología , Candida albicans/inmunología , Candidiasis/inmunología , Candidiasis/microbiología , Heces/microbiología , Centro Germinal/inmunología , Humanos , Ratones Endogámicos C57BL , Fagocitos/metabolismo , Polimorfismo de Nucleótido Simple/genética , Unión Proteica , Transducción de Señal
7.
Annu Rev Nutr ; 40: 323-343, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32680437

RESUMEN

The human gastrointestinal tract is home to a vibrant, diverse ecosystem of prokaryotic and eukaryotic microorganisms. The gut fungi (mycobiota) have recently risen to prominence due to their ability to modulate host immunity. Colonization of the gut occurs through a combination of vertical transmission from the maternal mycobiota and environmental and dietary exposure. Data from human and animal studies demonstrate that nutrition strongly affects the mycobiota composition and that changes in the fungal communities can aggravate metabolic diseases. The mechanisms pertaining to the mycobiota's influence on host health, pathology, and resident gastrointestinal communities through intrakingdom, transkingdom, and immune cross talk are beginning to come into focus, setting the stage for a new chapter in microbiota-host interactions. Herein, we examine the inception, maturation, and dietary modulation of gastrointestinal and nutritional fungal communities and inspect their impact on metabolic diseases in humans.


Asunto(s)
Hongos/fisiología , Microbioma Gastrointestinal/fisiología , Estado Nutricional , Interacciones Microbiota-Huesped , Humanos
8.
Cell Host Microbe ; 27(5): 823-829.e3, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32298656

RESUMEN

Fecal microbiota transplantation (FMT) targeting gut microbiota has recently been successfully applied to ulcerative colitis. However, only a subset of patients responds to FMT, and there is a pressing need for biomarkers of responsiveness. Fungi (the mycobiota) represent a highly immunologically reactive component of the gut microbiota. We analyzed samples from a large randomized controlled trial of FMT for ulcerative colitis (UC). High Candida abundance pre-FMT was associated with a clinical response, whereas decreased Candida abundance post-FMT was indicative of ameliorated disease severity. High pre-FMT Candida was associated with increased bacterial diversity post-FMT, and the presence of genera was linked to FMT responsiveness. Although we detected elevated anti-Candida antibodies in placebo recipients, this increase was abrogated in FMT recipients. Our data suggest that FMT might reduce Candida to contain pro-inflammatory immunity during intestinal disease and highlight the utility of mycobiota-focused approaches to identify FMT responders prior to therapy initiation.


Asunto(s)
Colitis Ulcerosa/terapia , Trasplante de Microbiota Fecal/métodos , Heces/microbiología , Hongos , Adulto , Anticuerpos Antifúngicos/sangre , Bacterias/genética , Candida , Colitis Ulcerosa/microbiología , Femenino , Hongos/genética , Microbioma Gastrointestinal/fisiología , Humanos , Masculino , ARN Ribosómico 16S/genética , Resultado del Tratamiento
9.
J Exp Med ; 216(12): 2689-2700, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31601676

RESUMEN

Host-microbiota interactions are critical in regulating mammalian health and disease. In addition to bacteria, parasites, and viruses, beneficial communities of fungi (the mycobiome) are important modulators of immune- and tissue-homeostasis. Chitin is a major component of the fungal cell wall, and fibrinogen C containing domain 1 (FIBCD1) is a chitin-binding protein; however, the role of this molecule in influencing host-mycobiome interactions in vivo has never been examined. Here, we identify direct binding of FIBCD1 to intestinal-derived fungi and demonstrate that epithelial-specific expression of FIBCD1 results in significantly reduced fungal colonization and amelioration of fungal-driven intestinal inflammation. Collectively, these results identify FIBCD1 as a previously unrecognized microbial pattern recognition receptor through which intestinal epithelial cells can recognize and control fungal colonization, limit fungal dysbiosis, and dampen intestinal inflammation.


Asunto(s)
Hongos/fisiología , Interacciones Microbianas , Micobioma , Receptores de Superficie Celular/metabolismo , Animales , Quitina/metabolismo , ADN Espaciador Ribosómico , Modelos Animales de Enfermedad , Enteritis/etiología , Enteritis/metabolismo , Enteritis/patología , Microbioma Gastrointestinal , Expresión Génica , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Metagenómica , Ratones , Ratones Transgénicos , Unión Proteica , ARN Ribosómico 16S
10.
Science ; 365(6452)2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31371577

RESUMEN

Laboratory mouse studies are paramount for understanding basic biological phenomena but also have limitations. These include conflicting results caused by divergent microbiota and limited translational research value. To address both shortcomings, we transferred C57BL/6 embryos into wild mice, creating "wildlings." These mice have a natural microbiota and pathogens at all body sites and the tractable genetics of C57BL/6 mice. The bacterial microbiome, mycobiome, and virome of wildlings affect the immune landscape of multiple organs. Their gut microbiota outcompete laboratory microbiota and demonstrate resilience to environmental challenges. Wildlings, but not conventional laboratory mice, phenocopied human immune responses in two preclinical studies. A combined natural microbiota- and pathogen-based model may enhance the reproducibility of biomedical studies and increase the bench-to-bedside safety and success of immunological studies.


Asunto(s)
Animales Salvajes/microbiología , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Investigación Biomédica Traslacional/normas
11.
Immunity ; 50(6): 1365-1379, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31216461

RESUMEN

The mammalian intestine is colonized by a wealth of microorganisms-including bacteria, viruses, protozoa, and fungi-that are all integrated into a functional trans-kingdom community. Characterization of the composition of the fungal community-the mycobiota-has advanced further than the much-needed mechanistic studies. Recent findings have revealed roles for the gut mycobiota in the regulation of host immunity and in the development and progression of human diseases of inflammatory origin. We review these findings here while placing them in the context of the current understanding of the pathways and cellular networks that induce local and systemic immune responses to fungi in the gastrointestinal tract. We discuss gaps in knowledge and argue for the importance of considering bacteria-fungal interactions as we aim to define the roles of mycobiota in immune homeostasis and immune-associated pathologies.


Asunto(s)
Susceptibilidad a Enfermedades , Gastroenteritis/etiología , Microbioma Gastrointestinal/inmunología , Inmunidad , Inmunidad Adaptativa , Animales , Susceptibilidad a Enfermedades/inmunología , Gastroenteritis/metabolismo , Homeostasis , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata
12.
PLoS Biol ; 17(5): e3000271, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31083650

RESUMEN

Malaria parasites possess the remarkable ability to maintain chronic infections that fail to elicit a protective immune response, characteristics that have stymied vaccine development and cause people living in endemic regions to remain at risk of malaria despite previous exposure to the disease. These traits stem from the tremendous antigenic diversity displayed by parasites circulating in the field. For Plasmodium falciparum, the most virulent of the human malaria parasites, this diversity is exemplified by the variant gene family called var, which encodes the major surface antigen displayed on infected red blood cells (RBCs). This gene family exhibits virtually limitless diversity when var gene repertoires from different parasite isolates are compared. Previous studies indicated that this remarkable genome plasticity results from extensive ectopic recombination between var genes during mitotic replication; however, the molecular mechanisms that direct this process to antigen-encoding loci while the rest of the genome remains relatively stable were not determined. Using targeted DNA double-strand breaks (DSBs) and long-read whole-genome sequencing, we show that a single break within an antigen-encoding region of the genome can result in a cascade of recombination events leading to the generation of multiple chimeric var genes, a process that can greatly accelerate the generation of diversity within this family. We also found that recombinations did not occur randomly, but rather high-probability, specific recombination products were observed repeatedly. These results provide a molecular basis for previously described structured rearrangements that drive diversification of this highly polymorphic gene family.


Asunto(s)
Variación Antigénica/genética , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Mitosis/genética , Parásitos/genética , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Recombinación Genética , Animales , Secuencia de Bases , Cromosomas/genética , Roturas del ADN de Doble Cadena , Humanos , Telómero/genética
13.
Fungal Genet Biol ; 127: 45-49, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30849443

RESUMEN

Gut mycobiota dysbiosis can negatively impact the outcome of several diseases of inflammatory origin, suggesting a role of the mycobiota in influencing the host immunity. However, it is unknown whether the gut mycobiota composition can create an immune environment that would influence the immune response to a newly introduced intestinal fungus. Using ITS1 deep sequencing, we evaluated the mycobiome structure of C57BL/6J mice acquired from Jackson (JAX) or bred in a controlled environment at a dedicated room in our own mouse facility (WCM-CE) for several generations. We found that C57BL/6J mice from these segregated mouse colonies harbor dramatically different mycobiota. To assess whether the mycobiota make up can influence immune responses to colonization with a fungus foreign to the murine GI tract, we colonized JAX and WCM-CE mice with the human commensal C. albicans and measured Th17 responses in the gut. We found that independent of mycobiota composition, mice produced strong Th17 responses to gastrointestinal C. albicans colonization. Our data suggest that different mouse colonies can carry dramatically different mycobiota. Nevertheless, strong Th17 responses to a newly introduced opportunistic commensal fungus are potently induced independent of the mycobiota background in this experimental setting.


Asunto(s)
Hongos/clasificación , Tracto Gastrointestinal/microbiología , Micobioma , Simbiosis , Células Th17/inmunología , Animales , ADN Intergénico/genética , ADN Ribosómico/genética , Hongos/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno , Ratones , Ratones Endogámicos C57BL
14.
Cell Host Microbe ; 24(6): 847-856.e4, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30503509

RESUMEN

Sensing of the gut microbiota, including fungi, regulates mucosal immunity. Whether fungal sensing in the gut can influence immunity at other body sites is unknown. Here we show that fluconazole-induced gut fungal dysbiosis has persistent effects on allergic airway disease in a house dust mite challenge model. Mice with a defined community of bacteria, but lacking intestinal fungi were not susceptible to fluconazole-induced dysbiosis, while colonization with a fungal mixture recapitulated the detrimental effects. Gut-resident mononuclear phagocytes (MNPs) expressing the fractalkine receptor CX3CR1 were essential for the effect of gut fungal dysbiosis on peripheral immunity. Depletion of CX3CR1+ MNPs or selective inhibition of Syk signaling downstream of fungal sensing in these cells ameliorated lung allergy. These results indicate that disruption of intestinal fungal communities can have persistent effects on peripheral immunity and aggravate disease severity through fungal sensing by gut-resident CX3CR1+ MNPs.


Asunto(s)
Disbiosis , Hipersensibilidad , Animales , Receptor 1 de Quimiocinas CX3C , Hongos , Intestinos , Ratones , Fagocitos
15.
Curr Opin Gastroenterol ; 34(6): 392-397, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30239343

RESUMEN

PURPOSE OF REVIEW: In this review, we discuss recent advances into delineating the dual role of intestinal phagocytes in health and during intestinal disease. We further discuss the key role of gut-resident macrophages in recognition of bacterial and fungal microbiota in the gut. RECENT FINDINGS: Inflammatory bowel disease (IBD) commonly manifests with pathologic changes in the composition of gut bacterial and fungal microbiota. Intestinal macrophages are key regulators of the balance between tolerogenic immunity and inflammation. Recent studies have highlighted the role of resident intestinal macrophages in the control of commensal fungi and bacteria in the steady state and during dysbiosis. The dual role of these cells in maintaining intestinal homeostasis and responding to microbiota dysbiosis during inflammation is being increasingly studied. SUMMARY: It is becoming increasingly clear that an aberrant proinflammatory response to microbiota by infiltrating monocytes plays a role in the development of intestinal inflammation. Intestinal mononuclear phagocytes with characteristics of macrophages play an important role in limiting fungal and bacterial overgrowth under these conditions, but can be influenced by the inflammatory environment to further propel inflammation. Better understanding of the interaction of intestinal macrophages with host microbiota including commensal fungi and bacteria, provides an opportunity for the development of more targeted therapies for IBD.


Asunto(s)
Bacterias/inmunología , Hongos/inmunología , Microbioma Gastrointestinal/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Macrófagos/inmunología , Disbiosis/inmunología , Disbiosis/microbiología , Interacciones Microbiota-Huesped/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/inmunología
16.
Immunity ; 48(3): 584-598.e5, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29548673

RESUMEN

Live vaccines historically afford superior protection, yet the cellular and molecular mechanisms mediating protective immunity remain unclear. Here we found that vaccination of mice with live, but not dead, Gram-negative bacteria heightened follicular T helper cell (Tfh) differentiation, germinal center formation, and protective antibody production through the signaling adaptor TRIF. Complementing the dead vaccine with an innate signature of bacterial viability, bacterial RNA, recapitulated these responses. The interferon (IFN) and inflammasome pathways downstream of TRIF orchestrated Tfh responses extrinsically to B cells and classical dendritic cells. Instead, CX3CR1+CCR2- monocytes instructed Tfh differentiation through interleukin-1ß (IL-1ß), a tightly regulated cytokine secreted upon TRIF-dependent IFN licensing of the inflammasome. Hierarchical production of IFN-ß and IL-1ß dictated Tfh differentiation and elicited the augmented humoral responses characteristic of live vaccines. These findings identify bacterial RNA, an innate signature of microbial viability, as a trigger for Tfh differentiation and suggest new approaches toward vaccine formulations for coordinating augmented Tfh and B cell responses.


Asunto(s)
Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Activación de Linfocitos/inmunología , Viabilidad Microbiana/inmunología , ARN Bacteriano/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Anticuerpos Neutralizantes/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos B/metabolismo , Vacunas Bacterianas/inmunología , Biomarcadores , Diferenciación Celular/inmunología , Citocinas/metabolismo , Centro Germinal , Interacciones Huésped-Patógeno/inmunología , Inmunidad Celular , Inmunidad Innata , Inflamasomas/metabolismo , Ratones , Monocitos/inmunología , Monocitos/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Transducción de Señal , Linfocitos T Colaboradores-Inductores/metabolismo
17.
Science ; 359(6372): 232-236, 2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29326275

RESUMEN

Intestinal fungi are an important component of the microbiota, and recent studies have unveiled their potential in modulating host immune homeostasis and inflammatory disease. Nonetheless, the mechanisms governing immunity to gut fungal communities (mycobiota) remain unknown. We identified CX3CR1+ mononuclear phagocytes (MNPs) as being essential for the initiation of innate and adaptive immune responses to intestinal fungi. CX3CR1+ MNPs express antifungal receptors and activate antifungal responses in a Syk-dependent manner. Genetic ablation of CX3CR1+ MNPs in mice led to changes in gut fungal communities and to severe colitis that was rescued by antifungal treatment. In Crohn's disease patients, a missense mutation in the gene encoding CX3CR1 was identified and found to be associated with impaired antifungal responses. These results unravel a role of CX3CR1+ MNPs in mediating interactions between intestinal mycobiota and host immunity at steady state and during inflammatory disease.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/análisis , Receptor 1 de Quimiocinas CX3C/genética , Candida albicans/inmunología , Microbioma Gastrointestinal/inmunología , Intestinos/microbiología , Micobioma/inmunología , Fagocitos/inmunología , Animales , Anticuerpos Antifúngicos/biosíntesis , Anticuerpos Antifúngicos/sangre , Candida albicans/crecimiento & desarrollo , Colitis/tratamiento farmacológico , Colitis/microbiología , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Células Dendríticas/inmunología , Microbioma Gastrointestinal/fisiología , Humanos , Inmunidad Mucosa , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Intestinos/inmunología , Ratones , Mutación Missense , Micobioma/fisiología , Fagocitos/microbiología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
18.
J Crohns Colitis ; 12(3): 355-368, 2018 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-29136128

RESUMEN

BACKGROUND AND AIMS: During active inflammation, intraluminal intestinal pH is decreased in patients with inflammatory bowel disease [IBD]. Acidic pH may play a role in IBD pathophysiology. Recently, proton-sensing G-protein coupled receptors were identified, including GPR4, OGR1 [GPR68], and TDAG8 [GPR65]. We investigated whether GPR4 is involved in intestinal inflammation. METHODS: The role of GPR4 was assessed in murine colitis models by chronic dextran sulphate sodium [DSS] administration and by cross-breeding into an IL-10 deficient background for development of spontaneous colitis. Colitis severity was assessed by body weight, colonoscopy, colon length, histological score, cytokine mRNA expression, and myeloperoxidase [MPO] activity. In the spontaneous Il-10-/- colitis model, the incidence of rectal prolapse and characteristics of lamina propria leukocytes [LPLs] were analysed. RESULTS: Gpr4-/- mice showed reduced body weight loss and histology score after induction of chronic DSS colitis. In Gpr4-/-/Il-10-/- double knock-outs, the onset and progression of rectal prolapse were significantly delayed and mitigated compared with Gpr4+/+/Il-10-/- mice. Double knock-out mice showed lower histology scores, MPO activity, CD4+ T helper cell infiltration, IFN-γ, iNOS, MCP-1 [CCL2], CXCL1, and CXCL2 expression compared with controls. In colon, GPR4 mRNA was detected in endothelial cells, some smooth muscle cells, and some macrophages. CONCLUSIONS: Absence of GPR4 ameliorates colitis in IBD animal models, indicating an important regulatory role in mucosal inflammation, thus providing a new link between tissue pH and the immune system. Therapeutic inhibition of GPR4 may be beneficial for the treatment of IBD.


Asunto(s)
Colitis/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Prolapso Rectal/etiología , Animales , Quimiocina CCL2/metabolismo , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/patología , Sulfato de Dextran , Células Endoteliales/metabolismo , Femenino , Concentración de Iones de Hidrógeno , Interferón gamma/metabolismo , Interleucina-10/genética , Mucosa Intestinal/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Peroxidasa/metabolismo , Protones , ARN Mensajero/metabolismo , Prolapso Rectal/genética , Linfocitos T Colaboradores-Inductores/patología
19.
Sci Rep ; 7(1): 16500, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29184071

RESUMEN

Trichuris suis ova (TSO) have been tested for therapeutic application in inflammatory bowel diseases (IBD) yet understanding of the underlying mechanisms and safety in an immunocompromised host is limited due to lack of a suitable animal model. We used a recently established rabbit model of dextran sodium sulphate (DSS) induced colitis to study the efficacy, mechanisms and safety of TSO therapy in immunocompetent and immunosuppressed animals. TSO treatment prevented the DSS induced weight loss, delayed the onset of DSS induced symptoms by 2 days and significantly reduced the disease activity (DAI). TSO treatment protected caecal histology and prevented the colitis-associated loss in faecal microbiota diversity. Mainly the transcriptome of lamina propria mononuclear cells (LPMC) was affected by TSO treatment, showing dampened innate and adaptive inflammatory responses. The protective effect of TSO was lost in immunosuppressed rabbits, where TSO exacerbated colitis. Our data show that preventive TSO treatment ameliorates colitis severity in immunocompetent rabbits, modulates LPMC immune responses and reduces faecal dysbiosis. In contrast, the same TSO treatment exacerbates colitis in immunosuppressed animals. Our data provide further evidence for a therapeutic effect of TSO in IBD, yet caution is required with regard to TSO treatment in immunosuppressed patients.


Asunto(s)
Terapia Biológica , Colitis/etiología , Colitis/prevención & control , Terapia de Inmunosupresión , Trichuris/inmunología , Animales , Terapia Biológica/métodos , Colitis/tratamiento farmacológico , Colitis/patología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Humanos , Huésped Inmunocomprometido , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/terapia , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Conejos , Transcriptoma
20.
Sci Immunol ; 2(17)2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-29101210

RESUMEN

Candidalysin-induced epithelial cell damage promotes expansion of innate TCRαß+ cells during oropharyngeal candidiasis.


Asunto(s)
Candida albicans , Proteínas Fúngicas , Células Epiteliales , Factores de Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA