Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
mBio ; 14(2): e0276422, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37017530

RESUMEN

The conserved ESX-1 type VII secretion system is a major virulence determinant of pathogenic mycobacteria, including Mycobacterium tuberculosis and Mycobacterium marinum. ESX-1 is known to interact with infected macrophages, but its potential roles in regulating other host cells and immunopathology have remained largely unexplored. Using a murine M. marinum infection model, we identify neutrophils and Ly6C+MHCII+ monocytes as the main cellular reservoirs for the bacteria. We show that ESX-1 promotes intragranuloma accumulation of neutrophils and that neutrophils have a previously unrecognized required role in executing ESX-1-mediated pathology. To explore if ESX-1 also regulates the function of recruited neutrophils, we performed a single-cell RNA-sequencing analysis that indicated that ESX-1 drives newly recruited uninfected neutrophils into an inflammatory phenotype via an extrinsic mechanism. In contrast, monocytes restricted the accumulation of neutrophils and immunopathology, demonstrating a major host-protective function for monocytes specifically by suppressing ESX-1-dependent neutrophilic inflammation. Inducible nitric oxide synthase (iNOS) activity was required for the suppressive mechanism, and we identified Ly6C+MHCII+ monocytes as the main iNOS-expressing cell type in the infected tissue. These results suggest that ESX-1 mediates immunopathology by promoting neutrophil accumulation and phenotypic differentiation in the infected tissue, and they demonstrate an antagonistic interplay between monocytes and neutrophils by which monocytes suppress host-detrimental neutrophilic inflammation. IMPORTANCE The ESX-1 type VII secretion system is required for virulence of pathogenic mycobacteria, including Mycobacterium tuberculosis. ESX-1 interacts with infected macrophages, but its potential roles in regulating other host cells and immunopathology have remained largely unexplored. We demonstrate that ESX-1 promotes immunopathology by driving intragranuloma accumulation of neutrophils, which upon arrival adopt an inflammatory phenotype in an ESX-1-dependent manner. In contrast, monocytes limited the accumulation of neutrophils and neutrophil-mediated pathology via an iNOS-dependent mechanism, suggesting a major host-protective function for monocytes specifically by restricting ESX-1-dependent neutrophilic inflammation. These findings provide insight into how ESX-1 promotes disease, and they reveal an antagonistic functional relationship between monocytes and neutrophils that might regulate immunopathology not only in mycobacterial infection but also in other infections as well as in inflammatory conditions and cancer.


Asunto(s)
Mycobacterium marinum , Mycobacterium tuberculosis , Sistemas de Secreción Tipo VII , Animales , Ratones , Neutrófilos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sistemas de Secreción Tipo VII/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium marinum/genética , Inflamación/microbiología , Diferenciación Celular
2.
Nat Commun ; 14(1): 2307, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-37085516

RESUMEN

The intestinal lamina propria contains a diverse network of fibroblasts that provide key support functions to cells within their local environment. Despite this, our understanding of the diversity, location and ontogeny of fibroblasts within and along the length of the intestine remains incomplete. Here we show that the small and large intestinal lamina propria contain similar fibroblast subsets that locate in specific anatomical niches. Nevertheless, we find that the transcriptional profile of similar fibroblast subsets differs markedly between the small intestine and colon suggesting region specific functions. We perform in vivo transplantation and lineage-tracing experiments to demonstrate that adult intestinal fibroblast subsets, smooth muscle cells and pericytes derive from Gli1-expressing precursors present in embryonic day 12.5 intestine. Trajectory analysis of single cell RNA-seq datasets of E12.5 and adult mesenchymal cells suggest that adult smooth muscle cells and fibroblasts derive from distinct embryonic intermediates and that adult fibroblast subsets develop in a linear trajectory from CD81+ fibroblasts. Finally, we provide evidence that colonic subepithelial PDGFRαhi fibroblasts comprise several functionally distinct populations that originate from an Fgfr2-expressing fibroblast intermediate. Our results provide insights into intestinal stromal cell diversity, location, function, and ontogeny, with implications for intestinal development and homeostasis.


Asunto(s)
Intestino Grueso , Células Madre Mesenquimatosas , Colon , Fibroblastos/metabolismo , Intestino Grueso/anatomía & histología , Intestino Grueso/citología , Intestino Delgado , Intestinos/anatomía & histología , Intestinos/citología , Proteína con Dedos de Zinc GLI1/genética , Células Madre Mesenquimatosas/metabolismo
3.
J Exp Med ; 220(3)2023 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-36515659

RESUMEN

The small intestinal lamina propria contains large numbers of IFNγ-producing T helper (Th1) cells that play important roles in intestinal homeostasis and host defense, but the mechanisms underlying their development remain poorly understood. Here, we demonstrate that Th1 cells accumulate in the SI-LP after weaning and are maintained there long term. While both Th17 and Th1 cell accumulation in the SI-LP was microbiota dependent, Th1 cell accumulation uniquely required IL-27 and MHCII expression by cDC1. This reflected a requirement for IL-27 signaling in the priming of Th1 cells rather than for their maintenance once in the mucosa. cDC1-derived IL-27 was essential for maintaining the Th1-Th17 balance within the SI-LP, and in its absence, remaining Th1 cells expressed enhanced levels of Th17 signature genes. In conclusion, we identify cDC1-derived IL-27 as a key regulator of SI-LP Th1-Th17 cell homeostasis.


Asunto(s)
Linfocitos T CD4-Positivos , Interleucina-27 , Ratones , Animales , Linfocitos T CD4-Positivos/metabolismo , Interleucina-27/metabolismo , Interleucina-17/metabolismo , Células Th17/metabolismo , Células TH1/metabolismo , Mucosa Intestinal/metabolismo , Homeostasis
4.
Immunity ; 55(10): 1829-1842.e6, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36115337

RESUMEN

The adult immune system consists of cells that emerged at various times during ontogeny. We aimed to define the relationship between developmental origin and composition of the adult B cell pool during unperturbed hematopoiesis. Lineage tracing stratified murine adult B cells based on the timing of output, revealing that a substantial portion originated within a restricted neonatal window. In addition to B-1a cells, early-life time-stamped B cells included clonally interrelated IgA plasma cells in the gut and bone marrow. These were actively maintained by B cell memory within gut chronic germinal centers and contained commensal microbiota reactivity. Neonatal rotavirus infection recruited recurrent IgA clones that were distinct from those arising by infection with the same antigen in adults. Finally, gut IgA plasma cells arose from the same hematopoietic progenitors as B-1a cells during ontogeny. Thus, a complex layer of neonatally imprinted B cells confer unique antibody responses later in life.


Asunto(s)
Inmunoglobulina A , Microbiota , Animales , Linfocitos B , Centro Germinal , Ratones , Células Plasmáticas
5.
Immunity ; 55(8): 1431-1447.e11, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35830859

RESUMEN

Conventional dendritic cells (cDCs) consist of two major functionally and phenotypically distinct subsets, cDC1 and cDC2, whose development is dependent on distinct sets of transcription factors. Interferon regulatory factor 8 (IRF8) is required at multiple stages of cDC1 development, but its role in committed cDC1 remains unclear. Here, we used Xcr1-cre to delete Irf8 in committed cDC1 and demonstrate that Irf8 is required for maintaining the identity of cDC1. In the absence of Irf8, committed cDC1 acquired the transcriptional, functional, and chromatin accessibility properties of cDC2. This conversion was independent of Irf4 and was associated with the decreased accessibility of putative IRF8, Batf3, and composite AP-1-IRF (AICE)-binding elements, together with increased accessibility of cDC2-associated transcription-factor-binding elements. Thus, IRF8 expression by committed cDC1 is required for preventing their conversion into cDC2-like cells.


Asunto(s)
Células Dendríticas , Factores Reguladores del Interferón , Células Dendríticas/metabolismo , Epigénesis Genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo
6.
Mucosal Immunol ; 15(4): 745-761, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35418673

RESUMEN

Migratory dendritic cells expressing CD103 are the targets for mucosal vaccines. These belong to either of two lineage-restricted subsets, cDC1 or cDC2 cells, which have been linked to priming of functionally distinct CD4 T cells. However, recent studies have identified plasticity in cDC2 cells with overlapping functions with cDC1 cells, while the converse has not been reported. We genetically engineered a vaccine adjuvant platform that targeted the cholera toxin A1 (CTA1) ADP-ribosylating enzyme to CD103+ cDC1 and cDC2 cells using a single-chain antibody (scFv) to CD103. Unexpectedly, intranasal immunization with the CTA1-svFcCD103 adjuvant modified cDC1 cells to effectively prime Th17 cells, a function previously limited to cDC2 cells. In fact, cDC2 cells were dispensible, while cDC1 cells, lacking in Batf3-/- mice, were critical. Following intranasal immunizations isolated cDC1 cells from mLN exclusively promoted Rorgt+ T cells and IL-17, IL-21, and IL-22 production. Strong CD8 T cell responses through antigen cross presentation by cDC1 cells were also observed. Single-cell RNAseq analysis revealed upregulation of Th17-promoting gene signatures in sorted cDC1 cells. Gene expression in isolated cDC2 cells was largely unaffected. Our finding represents a major shift of paradigm as we have documented functional plasticity in cDC1 cells.


Asunto(s)
Gripe Humana , Infecciones por Orthomyxoviridae , Adenosina Difosfato/metabolismo , Adyuvantes Inmunológicos , Animales , Toxina del Cólera/metabolismo , Células Dendríticas , Humanos , Gripe Humana/metabolismo , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Células Th17
7.
Dev Comp Immunol ; 131: 104375, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35219758

RESUMEN

Gut-associated lymphoid tissues (GALT) serve as key priming sites for intestinal adaptive immune responses. Most of our understanding of GALT function and development arises from studies in mice. However, the diversity, structure and cellular composition of GALT differs markedly between mammalian species and the developmental window in which distinct GALT structures develop in large mammals remains poorly understood. Given the importance of pigs as models of human disease, as well as their role in livestock production, we adapted a recently developed protocol for the isolation of human GALT to assess the diversity, development and immune composition of large intestinal GALT in neonatal and adult pigs. We demonstrate that the large intestine of adult pigs contains two major GALT types; multifollicular submucosal GALT that we term submucosal lymphoid clusters (SLC) which develop prenatally, and as yet undescribed mucosal isolated lymphoid follicles (M-ILF), which arise after birth. Using confocal laser microscopy and flow cytometry, we additionally assess the microanatomy and lymphocyte composition of SLC and M-ILF, compare them to jejunal Peyer's patches (PP), and describe the maturation of these structures. Collectively, our results provide a deeper understanding of the diversity and development of GALT within the porcine large intestine.


Asunto(s)
Inmunidad Mucosa , Ganglios Linfáticos Agregados , Animales , Mucosa Intestinal , Intestino Grueso , Intestinos , Tejido Linfoide , Mamíferos , Ratones , Porcinos
8.
Scand J Immunol ; 96(5): e13219, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37807915

RESUMEN

The intestinal lamina propria (LP) contains distinct subsets of classical dendritic cells (cDC), each playing key non-redundant roles in intestinal immune homeostasis. Here, we show that glycoprotein 2 (GP2), a GPI-anchored protein and receptor for bacterial type-I fimbriae, is selectively expressed by CD103+CD11b+ cDC in the murine small intestine (SI). GP2 expression was induced on CD103+CD11b+ cDC within the SI-LP and was regulated by IRF4, TGFßR1- and retinoic acid signalling. Mice selectively lacking Gp2 on CD103+CD11b+ cDC (huLang-Cre.gp2fl/fl mice) had normal numbers and proportions of innate and adaptive immune cells in the SI-LP suggesting that GP2 expression by CD103+CD11b+ cDC is not required for intestinal immune homoeostasis.


Asunto(s)
Cadenas alfa de Integrinas , Intestinos , Ratones , Animales , Mucosa Intestinal , Intestino Delgado , Transducción de Señal , Células Dendríticas , Ratones Endogámicos C57BL
9.
Nat Microbiol ; 6(11): 1367-1382, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34675385

RESUMEN

Breastfeeding profoundly shapes the infant gut microbiota, which is critical for early life immune development, and the gut microbiota can impact host physiology in various ways, such as through the production of metabolites. However, few breastmilk-dependent microbial metabolites mediating host-microbiota interactions are currently known. Here, we demonstrate that breastmilk-promoted Bifidobacterium species convert aromatic amino acids (tryptophan, phenylalanine and tyrosine) into their respective aromatic lactic acids (indolelactic acid, phenyllactic acid and 4-hydroxyphenyllactic acid) via a previously unrecognized aromatic lactate dehydrogenase (ALDH). The ability of Bifidobacterium species to convert aromatic amino acids to their lactic acid derivatives was confirmed using monocolonized mice. Longitudinal profiling of the faecal microbiota composition and metabolome of Danish infants (n = 25), from birth until 6 months of age, showed that faecal concentrations of aromatic lactic acids are correlated positively with the abundance of human milk oligosaccharide-degrading Bifidobacterium species containing the ALDH, including Bifidobacterium longum, B. breve and B. bifidum. We further demonstrate that faecal concentrations of Bifidobacterium-derived indolelactic acid are associated with the capacity of these samples to activate in vitro the aryl hydrocarbon receptor (AhR), a receptor important for controlling intestinal homoeostasis and immune responses. Finally, we show that indolelactic acid modulates ex vivo immune responses of human CD4+ T cells and monocytes in a dose-dependent manner by acting as an agonist of both the AhR and hydroxycarboxylic acid receptor 3 (HCA3). Our findings reveal that breastmilk-promoted Bifidobacterium species produce aromatic lactic acids in the gut of infants and suggest that these microbial metabolites may impact immune function in early life.


Asunto(s)
Bifidobacterium/metabolismo , Microbioma Gastrointestinal , Ácido Láctico/metabolismo , Adulto , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bifidobacterium/química , Bifidobacterium/clasificación , Bifidobacterium/genética , Lactancia Materna , Estudios de Cohortes , Heces/microbiología , Femenino , Humanos , Lactante , Ácido Láctico/química , Masculino , Ratones , Receptores de Hidrocarburo de Aril/metabolismo , Adulto Joven
10.
Sci Immunol ; 6(60)2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34088744

RESUMEN

Although CD8+ T cell tolerance to tissue-specific antigen (TSA) is essential for host homeostasis, the mechanisms underlying peripheral cross-tolerance and whether they may differ between tissue sites remain to be fully elucidated. Here, we demonstrate that peripheral cross-tolerance to intestinal epithelial cell (IEC)-derived antigen involves the generation and suppressive function of FoxP3+CD8+ T cells. FoxP3+CD8+ Treg generation was dependent on intestinal cDC1, whose absence led to a break of tolerance and epithelial destruction. Mechanistically, intestinal cDC1-derived PD-L1, TGFß, and retinoic acid contributed to the generation of gut-tropic CCR9+CD103+FoxP3+CD8+ Tregs Last, CD103-deficient CD8+ T cells lacked tolerogenic activity in vivo, indicating a role for CD103 in FoxP3+CD8+ Treg function. Our results describe a role for FoxP3+CD8+ Tregs in cross-tolerance in the intestine for which development requires intestinal cDC1.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Tolerancia Periférica , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno , Autoantígenos/inmunología , Autoantígenos/metabolismo , Autoinmunidad , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Femenino , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Yeyuno/citología , Yeyuno/inmunología , Ratones , Modelos Animales , Cultivo Primario de Células , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Quimera por Trasplante
11.
Mucosal Immunol ; 14(4): 793-802, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33753873

RESUMEN

Gut-associated lymphoid tissues (GALT) are the key antigen sampling and adaptive immune inductive sites within the intestinal wall. Human GALT includes the multi-follicular Peyer's patches of the ileum, the vermiform appendix, and the numerous isolated lymphoid follicles (ILF) which are distributed along the length of the intestine. Our current understanding of GALT diversity and function derives primarily from studies in mice, and the relevance of many of these findings to human GALT remains unclear. Here we review our current understanding of human GALT diversity, structure, and composition as well as their potential for regulating intestinal immune responses during homeostasis and inflammatory bowel disease (IBD). Finally, we outline some key remaining questions regarding human GALT, the answers to which will advance our understanding of intestinal immune responses and provide potential opportunities to improve the treatment of intestinal diseases.


Asunto(s)
Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ganglios Linfáticos Agregados/fisiología , Animales , Biomarcadores/metabolismo , Susceptibilidad a Enfermedades , Homeostasis , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Mucosa , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/patología , Especificidad de Órganos , Ganglios Linfáticos Agregados/citología
12.
Nat Protoc ; 16(4): 2051-2067, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33619391

RESUMEN

Gut-associated lymphoid tissues (GALTs) comprise key intestinal immune inductive sites, including the Peyer's patches of the small intestine and different types of isolated lymphoid follicle (ILF) found along the length of the gut. Our understanding of human GALT is limited due to a lack of protocols for their isolation. Here we describe a technique that, uniquely among intestinal cell isolation protocols, allows identification and isolation of all human GALT, as well as GALT-free intestinal lamina propria (LP). The technique involves the mechanical separation of intestinal mucosa from the submucosa, allowing the identification and isolation of submucosal ILF (SM-ILF), LP-embedded mucosal ILF (M-ILF) and LP free of contaminating lymphoid tissue. Individual SM-ILF, M-ILF and Peyer's patch follicles can be subsequently digested for downstream cellular and molecular characterization. The technique, which takes 4-10 h, will be useful for researchers interested in intestinal immune development and function in health and disease.


Asunto(s)
Tracto Gastrointestinal/fisiología , Tejido Linfoide/fisiología , Técnicas de Cultivo de Tejidos/métodos , Recuento de Células , Supervivencia Celular , Colon/fisiología , Enfermedad de Crohn/patología , Humanos , Inmunidad Innata , Mucosa Intestinal/citología , Antígenos Comunes de Leucocito/metabolismo
13.
Nat Commun ; 12(1): 1093, 2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33597537

RESUMEN

Interactions between host and gut microbial communities are modulated by diets and play pivotal roles in immunological homeostasis and health. We show that exchanging the protein source in a high fat, high sugar, westernized diet from casein to whole-cell lysates of the non-commensal bacterium Methylococcus capsulatus Bath is sufficient to reverse western diet-induced changes in the gut microbiota to a state resembling that of lean, low fat diet-fed mice, both under mild thermal stress (T22 °C) and at thermoneutrality (T30 °C). Concomitant with microbiota changes, mice fed the Methylococcus-based western diet exhibit improved glucose regulation, reduced body and liver fat, and diminished hepatic immune infiltration. Intake of the Methylococcu-based diet markedly boosts Parabacteroides abundances in a manner depending on adaptive immunity, and upregulates triple positive (Foxp3+RORγt+IL-17+) regulatory T cells in the small and large intestine. Collectively, these data point to the potential for leveraging the use of McB lysates to improve immunometabolic homeostasis.


Asunto(s)
Intestino Grueso/inmunología , Intestino Delgado/inmunología , Methylococcus capsulatus/inmunología , Microbiota/inmunología , Proteínas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Dieta , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Homeostasis/inmunología , Interleucina-17/inmunología , Interleucina-17/metabolismo , Intestino Grueso/metabolismo , Intestino Grueso/microbiología , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Masculino , Methylococcus capsulatus/química , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Obesidad/inmunología , Proteínas/metabolismo , Linfocitos T Reguladores/metabolismo
14.
Cell Rep ; 31(11): 107763, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32553166

RESUMEN

The network topology of a protein interactome is shaped by the function of each protein, making it a resource of functional knowledge in tissues and in single cells. Today, this resource is underused, as complete network topology characterization has proved difficult for large protein interactomes. We apply a matrix visualization and decoding approach to a physical protein interactome of a dendritic cell, thereby characterizing its topology with no prior assumptions of structure. We discover 294 proteins, each forming topological motifs called "bow-ties" that tie together the majority of observed protein complexes. The central proteins of these bow-ties have unique network properties, display multifunctional capabilities, are enriched for essential proteins, and are widely expressed in other cells and tissues. Collectively, the bow-tie motifs are a pervasive and previously unnoted topological trend in cellular interactomes. As such, these results provide fundamental knowledge on how intracellular protein connectivity is organized and operates.


Asunto(s)
Modelos Biológicos , Mapeo de Interacción de Proteínas , Proteínas/metabolismo , Transducción de Señal/fisiología , Algoritmos , Animales , Biología Computacional/métodos , Humanos , Ratones , Mapeo de Interacción de Proteínas/métodos
15.
Eur J Immunol ; 50(10): 1525-1536, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32383212

RESUMEN

Initiation of adaptive immunity to particulate antigens in lymph nodes largely depends on their presentation by migratory dendritic cells (DCs). DC subsets differ in their capacity to induce specific types of immunity, allowing subset-specific DC-targeting to influence vaccination and therapy outcomes. Faithful drug design, however, requires exact understanding of subset-specific versus global activation mechanisms. cDC1, the subset of DCs that excel in supporting immunity toward viruses, intracellular bacteria, and tumors, express uniquely high levels of the pattern recognition receptor TLR3. Using various murine genetic models, we show here that both, the cDC1 and cDC2 subsets of cDCs are activated and migrate equally well in response to TLR3 stimulation in a cell extrinsic and TNF-α dependent manner, but that cDC1 show a unique requirement for type I interferon signaling. Our findings reveal common and differing pathways regulating DC subset migration, offering important insights for the design of DC-based vaccination and therapy approaches.


Asunto(s)
Células Dendríticas/inmunología , Intestinos/inmunología , Receptor Toll-Like 3/metabolismo , Animales , Vacunas contra el Cáncer , Movimiento Celular , Células Cultivadas , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Receptor Toll-Like 3/inmunología
16.
Immunity ; 52(3): 557-570.e6, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32160523

RESUMEN

The intestine contains some of the most diverse and complex immune compartments in the body. Here we describe a method for isolating human gut-associated lymphoid tissues (GALTs) that allows unprecedented profiling of the adaptive immune system in submucosal and mucosal isolated lymphoid follicles (SM-ILFs and M-ILFs, respectively) as well as in GALT-free intestinal lamina propria (LP). SM-ILF and M-ILF showed distinct patterns of distribution along the length of the intestine, were linked to the systemic circulation through MAdCAM-1+ high endothelial venules and efferent lymphatics, and had immune profiles consistent with immune-inductive sites. IgA sequencing analysis indicated that human ILFs are sites where intestinal adaptive immune responses are initiated in an anatomically restricted manner. Our findings position ILFs as key inductive hubs for regional immunity in the human intestine, and the methods presented will allow future assessment of these compartments in health and disease.


Asunto(s)
Inmunidad Adaptativa/inmunología , Inmunidad Mucosa/inmunología , Mucosa Intestinal/inmunología , Intestinos/inmunología , Tejido Linfoide/inmunología , Inmunidad Adaptativa/genética , Animales , Citometría de Flujo , Mucosa Gástrica/inmunología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/ultraestructura , Humanos , Inmunidad Mucosa/genética , Inmunoglobulina A/genética , Inmunoglobulina A/inmunología , Inmunoglobulina M/genética , Inmunoglobulina M/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestructura , Intestinos/ultraestructura , Linfocitos/inmunología , Linfocitos/metabolismo , Tejido Linfoide/metabolismo , Tejido Linfoide/ultraestructura , Microscopía Confocal , Microscopía Electrónica de Rastreo , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/ultraestructura , Análisis de Secuencia de ADN
17.
J Immunol ; 204(4): 1047-1055, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31900340

RESUMEN

Classical dendritic cells (cDC) can be classified into two major subsets: Irf8-dependent cDC1 and Irf4-expressing cDC2. Although these subsets play distinct roles in intestinal immune homeostasis, their functions in T cell-driven colitis remain unknown. To assess the role of IRF4 expression in cDC2 in T cell-driven colitis, CD11c-Cre.Irf4 fl/fl and Irf4 fl/fl mice were backcrossed onto a Rag-1 -/- background and used as recipients of CD45RBhiCD4+ T cells. Colitis score and innate immune cell influx were reduced in Cre+ mice 4 wk posttransfer, and these changes were associated with reduced CD4+ T cell counts in both the mesenteric lymph nodes and colon. By 7 wk, colitis score and colon CD4+ T cell numbers were similar in Cre+ and Cre- mice despite a selective reduction in Th17 cells in the colon of Cre+ mice and a continued reduction in CD4+ T cell numbers in mesenteric lymph nodes. Cotransfer of CD25+CD45RBlo CD4+ T cells prevented CD45RBhiCD4+ T cell-driven colitis in both Cre+ and Cre- recipients, demonstrating that IRF4 expression by cDC is not required for CD4+ regulatory T cell-mediated control of colitis. Collectively these results suggest a role for IRF4 expression in cDC2 in the generation of colitogenic CD4+ T cells, which becomes redundant as colitis progresses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Colitis/inmunología , Colon/inmunología , Células Dendríticas/inmunología , Factores Reguladores del Interferón/metabolismo , Mucosa Intestinal/inmunología , Animales , Linfocitos T CD4-Positivos/trasplante , Colitis/patología , Colon/patología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteínas de Homeodominio/genética , Humanos , Factores Reguladores del Interferón/genética , Mucosa Intestinal/patología , Ratones , Ratones Noqueados
18.
Eur J Immunol ; 49(10): 1457-1973, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31633216

RESUMEN

These guidelines are a consensus work of a considerable number of members of the immunology and flow cytometry community. They provide the theory and key practical aspects of flow cytometry enabling immunologists to avoid the common errors that often undermine immunological data. Notably, there are comprehensive sections of all major immune cell types with helpful Tables detailing phenotypes in murine and human cells. The latest flow cytometry techniques and applications are also described, featuring examples of the data that can be generated and, importantly, how the data can be analysed. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid, all written and peer-reviewed by leading experts in the field, making this an essential research companion.


Asunto(s)
Alergia e Inmunología/normas , Separación Celular/métodos , Separación Celular/normas , Citometría de Flujo/métodos , Citometría de Flujo/normas , Consenso , Humanos , Fenotipo
19.
Sci Signal ; 12(596)2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31455723

RESUMEN

Second mitochondria-derived activator of caspase (SMAC) mimetics (SMs) are selective antagonists of the inhibitor of apoptosis proteins (IAPs), which activate noncanonical NF-κB signaling and promote tumor cell death. Through gene expression analysis, we found that treatment of CD4+ T cells with SMs during T helper 17 (TH17) cell differentiation disrupted the balance between two antagonistic transcription factor modules. Moreover, proteomics analysis revealed that SMs altered the abundance of proteins associated with cell cycle, mitochondrial activity, and the balance between canonical and noncanonical NF-κB signaling. Whereas SMs inhibited interleukin-17 (IL-17) production and ameliorated TH17 cell-driven inflammation, they stimulated IL-22 secretion. Mechanistically, SM-mediated activation of NF-κB-inducing kinase (NIK) and the transcription factors RelB and p52 directly suppressed Il17a expression and IL-17A protein production, as well as the expression of a number of other immune genes. Induction of IL-22 production correlated with the NIK-dependent reduction in cMAF protein abundance and the enhanced activity of the aryl hydrocarbon receptor. Last, SMs also increased IL-9 and IL-13 production and, under competing conditions, favored the differentiation of naïve CD4+ T cells into TH2 cells rather than TH17 cells. These results demonstrate that SMs shape the gene expression and protein profiles of TH17 cells and inhibit TH17 cell-driven autoimmunity.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Materiales Biomiméticos/farmacología , Diferenciación Celular/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Mitocondriales , Proteínas Serina-Treonina Quinasas/inmunología , Células Th17/inmunología , Animales , Regulación de la Expresión Génica/inmunología , Ratones , Ratones Transgénicos , Células Th17/citología , Células Th2/citología , Células Th2/inmunología , Quinasa de Factor Nuclear kappa B
20.
Front Immunol ; 9: 2409, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30386346

RESUMEN

Systemic immunization with soluble flagellin (sFliC) from Salmonella Typhimurium induces mucosal responses, offering potential as an adjuvant platform for vaccines. Moreover, this engagement of mucosal immunity is necessary for optimal systemic immunity, demonstrating an interaction between these two semi-autonomous immune systems. Although TLR5 and CD103+CD11b+ cDC2 contribute to this process, the relationship between these is unclear in the early activation of CD4+ T cells and the development of antigen-specific B cell responses. In this work, we use TLR5-deficient mice and CD11c-cre.Irf4fl/fl mice (which have reduced numbers of cDC2, particularly intestinal CD103+CD11b+ cDCs), to address these points by studying the responses concurrently in the spleen and the mesenteric lymph nodes (MLN). We show that CD103+CD11b+ cDC2 respond rapidly and accumulate in the MLN after immunization with sFliC in a TLR5-dependent manner. Furthermore, we identify that whilst CD103+CD11b+ cDC2 are essential for the induction of primary T and B cell responses in the mucosa, they do not play such a central role for the induction of these responses in the spleen. Additionally, we show the involvement of CD103+CD11b+ cDC2 in the induction of Th2-associated responses. CD11c-cre.Irf4fl/fl mice showed a reduced primary FliC-specific Th2-associated IgG1 responses, but enhanced Th1-associated IgG2c responses. These data expand our current understanding of the mucosal immune responses promoted by sFliC and highlights the potential of this adjuvant for vaccine usage by taking advantage of the functionality of mucosal CD103+CD11b+ cDC2.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Dendríticas/metabolismo , Flagelina/inmunología , Animales , Antígenos CD/metabolismo , Antígeno CD11c/metabolismo , Técnica del Anticuerpo Fluorescente , Inmunización , Inmunohistoquímica , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Receptor Toll-Like 5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA