Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 917
Filtrar
Más filtros

Tipo del documento
Publication year range
1.
Annu Rev Immunol ; 38: 23-48, 2020 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-32340570

RESUMEN

The gastrointestinal tract harbors numerous commensal bacteria, referred to as the microbiota, that benefit host health by digesting dietary components and eliminating pathogens. The intestinal microbiota maintains epithelial barrier integrity and shapes the mucosal immune system, balancing host defense and oral tolerance with microbial metabolites, components, and attachment to host cells. To avoid aberrant immune responses, epithelial cells segregate the intestinal microbiota from immune cells by constructing chemical and physical barriers, leading to the establishment of host-commensal mutualism. Furthermore, intestinal immune cells participate in the maintenance of a healthy microbiota community and reinforce epithelial barrier functions. Perturbations of the microbiota composition are commonly observed in patients with autoimmune diseases and chronic inflammatory disorders. An understanding of the intimate interactions between the intestinal microbiota, epithelial cells, and immune cells that are crucial for the maintenance of intestinal homeostasis might promote advances in diagnostic and therapeutic approaches for various diseases.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Susceptibilidad a Enfermedades , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo
2.
Annu Rev Immunol ; 35: 119-147, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28125357

RESUMEN

The intestinal epithelial barrier includes columnar epithelial, Paneth, goblet, enteroendocrine, and tuft cells as well as other cell populations, all of which contribute properties essential for gastrointestinal homeostasis. The intestinal mucosa is covered by mucin, which contains antimicrobial peptides and secretory IgA and prevents luminal bacteria, fungi, and viruses from stimulating intestinal immune responses. Conversely, the transport of luminal microorganisms-mediated by M, dendritic, and goblet cells-into intestinal tissues facilitates the harmonization of active and quiescent mucosal immune responses. The bacterial population within gut-associated lymphoid tissues creates the intratissue cohabitations for harmonized mucosal immunity. Intermolecular and intercellular communication among epithelial, immune, and mesenchymal cells creates an environment conducive for epithelial regeneration and mucosal healing. This review summarizes the so-called intestinal mucosal ecological network-the complex but vital molecular and cellular interactions of epithelial mesenchymal cells, immune cells, and commensal microbiota that achieve intestinal homeostasis, regeneration, and healing.


Asunto(s)
Células Epiteliales/fisiología , Microbioma Gastrointestinal/inmunología , Mucosa Intestinal/inmunología , Animales , Comunicación Celular , Homeostasis , Humanos , Inmunidad Innata , Inmunoglobulina A/metabolismo , Mucosa Intestinal/patología , Cicatrización de Heridas
3.
Cell ; 187(1): 62-78.e20, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-38096822

RESUMEN

The microbiota influences intestinal health and physiology, yet the contributions of commensal protists to the gut environment have been largely overlooked. Here, we discover human- and rodent-associated parabasalid protists, revealing substantial diversity and prevalence in nonindustrialized human populations. Genomic and metabolomic analyses of murine parabasalids from the genus Tritrichomonas revealed species-level differences in excretion of the metabolite succinate, which results in distinct small intestinal immune responses. Metabolic differences between Tritrichomonas species also determine their ecological niche within the microbiota. By manipulating dietary fibers and developing in vitro protist culture, we show that different Tritrichomonas species prefer dietary polysaccharides or mucus glycans. These polysaccharide preferences drive trans-kingdom competition with specific commensal bacteria, which affects intestinal immunity in a diet-dependent manner. Our findings reveal unappreciated diversity in commensal parabasalids, elucidate differences in commensal protist metabolism, and suggest how dietary interventions could regulate their impact on gut health.


Asunto(s)
Microbioma Gastrointestinal , Parabasalidea , Polisacáridos , Animales , Humanos , Ratones , Fibras de la Dieta , Intestino Delgado/metabolismo , Polisacáridos/metabolismo , Parabasalidea/metabolismo , Carbohidratos de la Dieta/metabolismo , Biodiversidad
4.
Cell ; 186(3): 607-620.e17, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36640762

RESUMEN

Tissue immunity and responses to injury depend on the coordinated action and communication among physiological systems. Here, we show that, upon injury, adaptive responses to the microbiota directly promote sensory neuron regeneration. At homeostasis, tissue-resident commensal-specific T cells colocalize with sensory nerve fibers within the dermis, express a transcriptional program associated with neuronal interaction and repair, and promote axon growth and local nerve regeneration following injury. Mechanistically, our data reveal that the cytokine interleukin-17A (IL-17A) released by commensal-specific Th17 cells upon injury directly signals to sensory neurons via IL-17 receptor A, the transcription of which is specifically upregulated in injured neurons. Collectively, our work reveals that in the context of tissue damage, preemptive immunity to the microbiota can rapidly bridge biological systems by directly promoting neuronal repair, while also identifying IL-17A as a major determinant of this fundamental process.


Asunto(s)
Interleucina-17 , Microbiota , Regeneración Nerviosa , Células Th17 , Axones , Regeneración Nerviosa/fisiología , Células Receptoras Sensoriales , Animales , Ratones , Células Th17/citología
5.
Cell ; 183(6): 1562-1571.e12, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33306955

RESUMEN

Ticks transmit a diverse array of microbes to vertebrate hosts, including human pathogens, which has led to a human-centric focus in this vector system. Far less is known about pathogens of ticks themselves. Here, we discover that a toxin in blacklegged ticks (Ixodes scapularis) horizontally acquired from bacteria-called domesticated amidase effector 2 (dae2)-has evolved to kill mammalian skin microbes with remarkable efficiency. Secreted into the saliva and gut of ticks, Dae2 limits skin-associated staphylococci in ticks while feeding. In contrast, Dae2 has no intrinsic ability to kill Borrelia burgdorferi, the tick-borne Lyme disease bacterial pathogen. These findings suggest ticks resist their own pathogens while tolerating symbionts. Thus, just as tick symbionts can be pathogenic to humans, mammalian commensals can be harmful to ticks. Our study underscores how virulence is context-dependent and bolsters the idea that "pathogen" is a status and not an identity.


Asunto(s)
Bacterias/metabolismo , Factores Inmunológicos/metabolismo , Ixodes/fisiología , Piel/microbiología , Simbiosis , Animales , Antibacterianos/farmacología , Biocatálisis , Pared Celular/metabolismo , Conducta Alimentaria , Femenino , Tracto Gastrointestinal/metabolismo , Interacciones Huésped-Patógeno , Ratones , Modelos Moleculares , Peptidoglicano/metabolismo , Filogenia , Saliva/metabolismo , Glándulas Salivales/metabolismo , Staphylococcus epidermidis/fisiología , Homología Estructural de Proteína , Especificidad por Sustrato , Regulación hacia Arriba
6.
Immunity ; 57(8): 1893-1907.e6, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39096910

RESUMEN

Naive CD4+ T cells in specific pathogen-free (SPF) mice are characterized by transcriptional heterogeneity and subpopulations distinguished by the expression of quiescence, the extracellular matrix (ECM) and cytoskeleton, type I interferon (IFN-I) response, memory-like, and T cell receptor (TCR) activation genes. We demonstrate that this constitutive heterogeneity, including the presence of the IFN-I response cluster, is commensal independent insofar as being identical in germ-free and SPF mice. By contrast, Nippostrongylus brasiliensis infection altered this constitutive heterogeneity. Naive T cell-intrinsic transcriptional changes acquired during helminth infection correlated with and accounted for decreased immunization response to an unrelated antigen. These compositional and functional changes were dependent variables of helminth infection, as they disappeared at the established time point of its clearance in mice. Collectively, our results indicate that the naive T cell pool is subject to dynamic transcriptional changes in response to certain environmental cues, which in turn permutes the magnitude of the immune response.


Asunto(s)
Linfocitos T CD4-Positivos , Nippostrongylus , Animales , Ratones , Linfocitos T CD4-Positivos/inmunología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Organismos Libres de Patógenos Específicos , Transcripción Genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Helmintiasis/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Ratones Endogámicos C57BL , Activación de Linfocitos/inmunología
7.
Immunity ; 57(6): 1428-1441.e8, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38723638

RESUMEN

Induction of commensal-specific immunity contributes to tissue homeostasis, yet the mechanisms underlying induction of commensal-specific B cells remain poorly understood in part due to a lack of tools to identify these cells. Using phage display, we identified segmented filamentous bacteria (SFB) antigens targeted by serum and intestinal antibodies and generated B cell tetramers to track SFB-specific B cells in gut-associated lymphoid tissues. We revealed a compartmentalized response in SFB-specific B cell activation, with a gradient of immunoglobulin A (IgA), IgG1, and IgG2b isotype production along Peyer's patches contrasted by selective production of IgG2b within mesenteric lymph nodes. V(D)J sequencing and monoclonal antibody generation identified somatic hypermutation driven affinity maturation to SFB antigens under homeostatic conditions. Combining phage display and B cell tetramers will enable investigation of the ontogeny and function of commensal-specific B cell responses in tissue immunity, inflammation, and repair.


Asunto(s)
Linfocitos B , Animales , Linfocitos B/inmunología , Ratones , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados/inmunología , Activación de Linfocitos/inmunología , Antígenos Bacterianos/inmunología , Hipermutación Somática de Inmunoglobulina , Biblioteca de Péptidos , Ganglios Linfáticos/inmunología , Técnicas de Visualización de Superficie Celular , Simbiosis/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina A/inmunología
8.
Immunity ; 56(5): 1115-1131.e9, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-36917985

RESUMEN

Intestinal IL-17-producing T helper (Th17) cells are dependent on adherent microbes in the gut for their development. However, how microbial adherence to intestinal epithelial cells (IECs) promotes Th17 cell differentiation remains enigmatic. Here, we found that Th17 cell-inducing gut bacteria generated an unfolded protein response (UPR) in IECs. Furthermore, subtilase cytotoxin expression or genetic removal of X-box binding protein 1 (Xbp1) in IECs caused a UPR and increased Th17 cells, even in antibiotic-treated or germ-free conditions. Mechanistically, UPR activation in IECs enhanced their production of both reactive oxygen species (ROS) and purine metabolites. Treating mice with N-acetyl-cysteine or allopurinol to reduce ROS production and xanthine, respectively, decreased Th17 cells that were associated with an elevated UPR. Th17-related genes also correlated with ER stress and the UPR in humans with inflammatory bowel disease. Overall, we identify a mechanism of intestinal Th17 cell differentiation that emerges from an IEC-associated UPR.


Asunto(s)
Estrés del Retículo Endoplásmico , Mucosa Intestinal , Células Th17 , Estrés del Retículo Endoplásmico/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Células Th17/citología , Células Th17/metabolismo , Diferenciación Celular , Humanos , Animales , Ratones , Ratones Transgénicos , Antibacterianos/farmacología
9.
Immunity ; 56(12): 2719-2735.e7, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38039966

RESUMEN

Commensal microbes induce cytokine-producing effector tissue-resident CD4+ T cells, but the function of these T cells in mucosal homeostasis is not well understood. Here, we report that commensal-specific intestinal Th17 cells possess an anti-inflammatory phenotype marked by expression of interleukin (IL)-10 and co-inhibitory receptors. The anti-inflammatory phenotype of gut-resident commensal-specific Th17 cells was driven by the transcription factor c-MAF. IL-10-producing commensal-specific Th17 cells were heterogeneous and derived from a TCF1+ gut-resident progenitor Th17 cell population. Th17 cells acquired IL-10 expression and anti-inflammatory phenotype in the small-intestinal lamina propria. IL-10 production by CD4+ T cells and IL-10 signaling in intestinal macrophages drove IL-10 expression by commensal-specific Th17 cells. Intestinal commensal-specific Th17 cells possessed immunoregulatory functions and curbed effector T cell activity in vitro and in vivo in an IL-10-dependent and c-MAF-dependent manner. Our results suggest that tissue-resident commensal-specific Th17 cells perform regulatory functions in mucosal homeostasis.


Asunto(s)
Microbioma Gastrointestinal , Células Th17 , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Antiinflamatorios
10.
Immunity ; 56(6): 1239-1254.e7, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37028427

RESUMEN

Early-life establishment of tolerance to commensal bacteria at barrier surfaces carries enduring implications for immune health but remains poorly understood. Here, we showed that tolerance in skin was controlled by microbial interaction with a specialized subset of antigen-presenting cells. More particularly, CD301b+ type 2 conventional dendritic cells (DCs) in neonatal skin were specifically capable of uptake and presentation of commensal antigens for the generation of regulatory T (Treg) cells. CD301b+ DC2 were enriched for phagocytosis and maturation programs, while also expressing tolerogenic markers. In both human and murine skin, these signatures were reinforced by microbial uptake. In contrast to their adult counterparts or other early-life DC subsets, neonatal CD301b+ DC2 highly expressed the retinoic-acid-producing enzyme, RALDH2, the deletion of which limited commensal-specific Treg cell generation. Thus, synergistic interactions between bacteria and a specialized DC subset critically support early-life tolerance at the cutaneous interface.


Asunto(s)
Células Dendríticas , Piel , Animales , Ratones , Humanos , Linfocitos T Reguladores , Tolerancia Inmunológica , Aldehído Oxidorreductasas/metabolismo
11.
Cell ; 169(5): 849-861.e13, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28502769

RESUMEN

We examined the evolutionary history of leading multidrug resistant hospital pathogens, the enterococci, to their origin hundreds of millions of years ago. Our goal was to understand why, among the vast diversity of gut flora, enterococci are so well adapted to the modern hospital environment. Molecular clock estimation, together with analysis of their environmental distribution, phenotypic diversity, and concordance with host fossil records, place the origins of the enterococci around the time of animal terrestrialization, 425-500 mya. Speciation appears to parallel the diversification of hosts, including the rapid emergence of new enterococcal species following the End Permian Extinction. Major drivers of speciation include changing carbohydrate availability in the host gut. Life on land would have selected for the precise traits that now allow pathogenic enterococci to survive desiccation, starvation, and disinfection in the modern hospital, foreordaining their emergence as leading hospital pathogens.


Asunto(s)
Evolución Biológica , Enterococcus/genética , Animales , Enfermedades Transmisibles Emergentes/microbiología , Infección Hospitalaria/microbiología , Farmacorresistencia Bacteriana , Enterococcus/clasificación , Enterococcus/citología , Enterococcus/efectos de los fármacos , Especiación Genética , Interacciones Huésped-Patógeno , Larva/microbiología , Mariposas Nocturnas/crecimiento & desarrollo , Mariposas Nocturnas/microbiología , Filogenia , ARN Ribosómico 16S/genética
12.
Immunity ; 55(9): 1594-1608.e6, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36029766

RESUMEN

Tumor-induced host wasting and mortality are general phenomena across species. Many groups have previously demonstrated endocrinal impacts of malignant tumors on host wasting in rodents and Drosophila. Whether and how environmental factors and host immune response contribute to tumor-associated host wasting and survival, however, are largely unknown. Here, we report that flies bearing malignant yki3SA-gut tumors exhibited the exponential increase of commensal bacteria, which were mostly acquired from the environment, and systemic IMD-NF-κB activation due to suppression of a gut antibacterial amidase PGRP-SC2. Either gut microbial elimination or specific IMD-NF-κB blockade in the renal-like Malpighian tubules potently improved mortality of yki3SA-tumor-bearing flies in a manner independent of host wasting. We further indicate that renal IMD-NF-κB activation caused uric acid (UA) overload to reduce survival of tumor-bearing flies. Therefore, our results uncover a fundamental mechanism whereby gut commensal dysbiosis, renal immune activation, and UA imbalance potentiate tumor-associated host death.


Asunto(s)
FN-kappa B , Neoplasias , Animales , Proteínas Portadoras , Drosophila , Homeostasis , FN-kappa B/metabolismo , Ácido Úrico
13.
Cell ; 167(2): 444-456.e14, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27716507

RESUMEN

While conventional pathogenic protists have been extensively studied, there is an underappreciated constitutive protist microbiota that is an integral part of the vertebrate microbiome. The impact of these species on the host and their potential contributions to mucosal immune homeostasis remain poorly studied. Here, we show that the protozoan Tritrichomonas musculis activates the host epithelial inflammasome to induce IL-18 release. Epithelial-derived IL-18 promotes dendritic cell-driven Th1 and Th17 immunity and confers dramatic protection from mucosal bacterial infections. Along with its role as a "protistic" antibiotic, colonization with T. musculis exacerbates the development of T-cell-driven colitis and sporadic colorectal tumors. Our findings demonstrate a novel mutualistic host-protozoan interaction that increases mucosal host defenses at the cost of an increased risk of inflammatory disease.


Asunto(s)
Colitis/inmunología , Colitis/parasitología , Interacciones Huésped-Parásitos , Inflamasomas/inmunología , Mucosa Intestinal/parasitología , Microbiota/inmunología , Tricomoniasis/inmunología , Trichomonas/inmunología , Animales , Colitis/microbiología , Dientamoeba/inmunología , Inmunidad Mucosa , Interleucina-18/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Simbiosis , Células TH1/inmunología , Células Th17/inmunología
14.
Immunity ; 54(6): 1137-1153.e8, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34051146

RESUMEN

Alterations in the cGAS-STING DNA-sensing pathway affect intestinal homeostasis. We sought to delineate the functional role of STING in intestinal inflammation. Increased STING expression was a feature of intestinal inflammation in mice with colitis and in humans afflicted with inflammatory bowel disease. Mice bearing an allele rendering STING constitutively active exhibited spontaneous colitis and dysbiosis, as well as progressive chronic intestinal inflammation and fibrosis. Bone marrow chimera experiments revealed STING accumulation in intestinal macrophages and monocytes as the initial driver of inflammation. Depletion of Gram-negative bacteria prevented STING accumulation in these cells and alleviated intestinal inflammation. STING accumulation occurred at the protein rather than transcript level, suggesting post-translational stabilization. We found that STING was ubiquitinated in myeloid cells, and this K63-linked ubiquitination could be elicited by bacterial products, including cyclic di-GMP. Our findings suggest a positive feedback loop wherein dysbiosis foments the accumulation of STING in intestinal myeloid cells, driving intestinal inflammation.


Asunto(s)
Colitis/inmunología , Disbiosis/inmunología , Inmunidad Innata/inmunología , Proteínas de la Membrana/inmunología , Células Mieloides/inmunología , Ubiquitinación/inmunología , Animales , Estudios de Casos y Controles , Femenino , Humanos , Inflamación/inmunología , Intestinos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología
15.
Immunity ; 54(11): 2547-2564.e7, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34715017

RESUMEN

Cryptosporidium can cause severe diarrhea and morbidity, but many infections are asymptomatic. Here, we studied the immune response to a commensal strain of Cryptosporidium tyzzeri (Ct-STL) serendipitously discovered when conventional type 1 dendritic cell (cDC1)-deficient mice developed cryptosporidiosis. Ct-STL was vertically transmitted without negative health effects in wild-type mice. Yet, Ct-STL provoked profound changes in the intestinal immune system, including induction of an IFN-γ-producing Th1 response. TCR sequencing coupled with in vitro and in vivo analysis of common Th1 TCRs revealed that Ct-STL elicited a dominant antigen-specific Th1 response. In contrast, deficiency in cDC1s skewed the Ct-STL CD4 T cell response toward Th17 and regulatory T cells. Although Ct-STL predominantly colonized the small intestine, colon Th1 responses were enhanced and associated with protection against Citrobacter rodentium infection and exacerbation of dextran sodium sulfate and anti-IL10R-triggered colitis. Thus, Ct-STL represents a commensal pathobiont that elicits Th1-mediated intestinal homeostasis that may reflect asymptomatic human Cryptosporidium infection.


Asunto(s)
Criptosporidiosis/inmunología , Criptosporidiosis/parasitología , Cryptosporidium/inmunología , Células Dendríticas/inmunología , Interacciones Huésped-Parásitos/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Células TH1/inmunología , Animales , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Homeostasis , Mucosa Intestinal/metabolismo , Ratones , Microbiota , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células TH1/metabolismo
16.
Immunity ; 52(4): 635-649.e4, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32240600

RESUMEN

The intestinal microbiota shapes and directs immune development locally and systemically, but little is known about whether commensal microbes in the stomach can impact their immunological microenvironment. Here, we report that group 2 innate lymphoid cells (ILC2s) were the predominant ILC subset in the stomach and show that their homeostasis and effector functions were regulated by local commensal communities. Microbes elicited interleukin-7 (IL-7) and IL-33 production in the stomach, which in turn triggered the propagation and activation of ILC2. Stomach ILC2s were also rapidly induced following infection with Helicobacter pylori. ILC2-derived IL-5 resulted in the production of IgA, which coated stomach bacteria in both specific pathogen-free (SPF) and H. pylori-infected mice. Our study thus identifies ILC2-dependent IgA response that is regulated by the commensal microbiota, which is implicated in stomach protection by eliminating IgA-coated bacteria including pathogenic H. pylori.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter pylori/patogenicidad , Inmunoglobulina A/biosíntesis , Interleucina-5/inmunología , Estómago/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Femenino , Regulación de la Expresión Génica , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/crecimiento & desarrollo , Helicobacter pylori/inmunología , Inmunidad Humoral , Inmunidad Innata , Interleucina-33/genética , Interleucina-33/inmunología , Interleucina-5/genética , Interleucina-7/genética , Interleucina-7/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Transducción de Señal , Estómago/microbiología , Simbiosis/inmunología , Subgrupos de Linfocitos T/clasificación
17.
Immunol Rev ; 326(1): 203-218, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39285525

RESUMEN

The increasing prevalence of immune-mediated non-communicable chronic diseases, such as food allergies, has prompted a deeper investigation into the role of the gut microbiome in modulating immune responses. Here, we explore the complex interactions between commensal microbes and the host immune system, highlighting the critical role of gut bacteria in maintaining immune homeostasis. We examine how modern lifestyle practices and environmental factors have disrupted co-evolved host-microbe interactions and discuss how changes in microbiome composition impact epithelial barrier function, responses to food allergens, and susceptibility to allergic diseases. Finally, we examine the potential of bioengineered microbiome-based therapies, and live biotherapeutic products, for reestablishing immune homeostasis to prevent or treat food allergies.


Asunto(s)
Hipersensibilidad a los Alimentos , Microbioma Gastrointestinal , Simbiosis , Humanos , Animales , Microbioma Gastrointestinal/inmunología , Hipersensibilidad a los Alimentos/inmunología , Simbiosis/inmunología , Homeostasis , Alérgenos/inmunología , Alimentos , Inmunomodulación , Interacciones Microbiota-Huesped/inmunología , Probióticos/uso terapéutico
18.
Annu Rev Microbiol ; 76: 757-782, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36075093

RESUMEN

Identified in the late nineteenth century as a single species residing on human skin, Malassezia is now recognized as a diverse genus comprising 18 species inhabiting not only skin but human gut, hospital environments, and even deep-sea sponges. All cultivated Malassezia species are lipid dependent, having lost genes for lipid synthesis and carbohydrate metabolism. The surging interest in Malassezia results from development of tools to improve sampling, culture, identification, and genetic engineering, which has led to findings implicating it in numerous skin diseases, Crohn disease, and pancreatic cancer. However, it has become clear that Malassezia plays a multifaceted role in human health, with mutualistic activity in atopic dermatitis and a preventive effect against other skin infections due to its potential to compete with skin pathogens such as Candida auris. Improved understanding of complex microbe-microbe and host-microbe interactions will be required to define Malassezia's role in human and animal health and disease so as to design targeted interventions.


Asunto(s)
Dermatitis Atópica , Malassezia , Animales , Humanos , Lípidos , Malassezia/genética , Piel , Simbiosis
19.
Immunity ; 49(6): 1103-1115.e6, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30566883

RESUMEN

Retinoic acid (RA), a vitamin A metabolite, regulates transcriptional programs that drive protective or pathogenic immune responses in the intestine, in a manner dependent on RA concentration. Vitamin A is obtained from diet and is metabolized by intestinal epithelial cells (IECs), which operate in intimate association with microbes and immune cells. Here we found that commensal bacteria belonging to class Clostridia modulate RA concentration in the gut by suppressing the expression of retinol dehydrogenase 7 (Rdh7) in IECs. Rdh7 expression and associated RA amounts were lower in the intestinal tissue of conventional mice, as compared to germ-free mice. Deletion of Rdh7 in IECs diminished RA signaling in immune cells, reduced the IL-22-dependent antimicrobial response, and enhanced resistance to colonization by Salmonella Typhimurium. Our findings define a regulatory circuit wherein bacterial regulation of IEC-intrinsic RA synthesis protects microbial communities in the gut from excessive immune activity, achieving a balance that prevents colonization by enteric pathogens.


Asunto(s)
Disbiosis/metabolismo , Células Epiteliales/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Tretinoina/metabolismo , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Bacterias/clasificación , Bacterias/genética , Disbiosis/microbiología , Células Epiteliales/microbiología , Interacciones Microbiota-Huesped , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Linfocitos/metabolismo , Linfocitos/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota/genética , Microbiota/fisiología , ARN Ribosómico 16S/genética , Salmonella typhimurium/genética , Salmonella typhimurium/fisiología , Simbiosis , Interleucina-22
20.
Proc Natl Acad Sci U S A ; 121(11): e2315540121, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38437561

RESUMEN

Insects lack acquired immunity and were thought to have no immune memory, but recent studies reported a phenomenon called immune priming, wherein sublethal dose of pathogens or nonpathogenic microbes stimulates immunity and prevents subsequential pathogen infection. Although the evidence for insect immune priming is accumulating, the underlying mechanisms are still unclear. The bean bug Riptortus pedestris acquires its gut microbiota from ambient soil and spatially structures them into a multispecies and variable community in the anterior midgut and a specific, monospecies Caballeronia symbiont population in the posterior region. We demonstrate that a particular Burkholderia strain colonizing the anterior midgut stimulates systemic immunity by penetrating gut epithelia and migrating into the hemolymph. The activated immunity, consisting of a humoral and a cellular response, had no negative effect on the host fitness, but on the contrary protected the insect from subsequent infection by pathogenic bacteria. Interruption of contact between the Burkholderia strain and epithelia of the gut weakened the host immunity back to preinfection levels and made the insects more vulnerable to microbial infection, demonstrating that persistent acquisition of environmental bacteria is important to maintain an efficient immunity.


Asunto(s)
Burkholderia , Burkholderiaceae , Animales , Endodermo , Insectos , Suelo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda