Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(5): 114153, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38687643

RESUMEN

Gut-draining mesenteric and celiac lymph nodes (mLNs and celLNs) critically contribute to peripheral tolerance toward food and microbial antigens by supporting the de novo induction of regulatory T cells (Tregs). These tolerogenic properties of mLNs and celLNs are stably imprinted within stromal cells (SCs) by microbial signals and vitamin A (VA), respectively. Here, we report that a single, transient gastrointestinal infection in the neonatal, but not adult, period durably abrogates the efficient Treg-inducing capacity of celLNs by altering the subset composition and gene expression profile of celLNSCs. These cells carry information about the early-life pathogen encounter until adulthood and durably instruct migratory dendritic cells entering the celLN with reduced tolerogenic properties. Mechanistically, transiently reduced VA levels cause long-lasting celLN functional impairment, which can be rescued by early-life treatment with VA. Together, our data highlight the therapeutic potential of VA to prevent sequelae post gastrointestinal infections in infants.


Asunto(s)
Ganglios Linfáticos , Linfocitos T Reguladores , Vitamina A , Animales , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos/efectos de los fármacos , Vitamina A/farmacología , Vitamina A/uso terapéutico , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Ratones , Animales Recién Nacidos , Tolerancia Inmunológica/efectos de los fármacos , Células Dendríticas/inmunología , Ratones Endogámicos C57BL , Femenino
2.
Front Immunol ; 13: 1082055, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36569861

RESUMEN

Regulatory T cells in non-lymphoid tissues are not only critical for maintaining self-tolerance, but are also important for promoting organ homeostasis and tissue repair. It is proposed that the generation of tissue Treg cells is a stepwise, multi-site process, accompanied by extensive epigenome remodeling, finally leading to the acquisition of unique tissue-specific epigenetic signatures. This process is initiated in the thymus, where Treg cells acquire core phenotypic and functional properties, followed by a priming step in secondary lymphoid organs that permits Treg cells to exit the lymphoid organs and seed into non-lymphoid tissues. There, a final specialization process takes place in response to unique microenvironmental cues in the respective tissue. In this review, we will summarize recent findings on this multi-site tissue Treg cell differentiation and highlight the importance of epigenetic remodeling during these stepwise events.


Asunto(s)
Tolerancia Inmunológica , Linfocitos T Reguladores , Diferenciación Celular , Autotolerancia , Epigénesis Genética
3.
Nat Commun ; 13(1): 7227, 2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36433946

RESUMEN

Gut-draining mesenteric lymph nodes (LN) provide the framework to shape intestinal adaptive immune responses. Based on the transcriptional signatures established by our previous work, the composition and immunomodulatory function of LN stromal cells (SC) vary according to location. Here, we describe the single-cell composition and development of the SC compartment within mesenteric LNs derived from postnatal to aged mice. We identify CD34+ SC and fibroblastic reticular stromal cell (FRC) progenitors as putative progenitors, both supplying the typical rapid postnatal mesenteric LN expansion. We further establish the location-specific chromatin accessibility and DNA methylation landscape of non-endothelial SCs and identify a microbiota-independent core epigenomic signature, showing characteristic differences between SCs from mesenteric and skin-draining peripheral LNs. The epigenomic landscape of SCs points to dynamic expression of Irf3 along the differentiation trajectories of FRCs. Accordingly, a mesenchymal stem cell line acquires a Cxcl9+ FRC molecular phenotype upon lentiviral overexpression of Irf3, and the relevance of Irf3 for SC biology is further underscored by the diminished proportion of Ccl19+ and Cxcl9+ FRCs in LNs of Irf3-/- mice. Together, our data constitute a comprehensive transcriptional and epigenomic map of mesenteric LNSC development in early life and dissect location-specific, microbiota-independent properties of non-endothelial SCs.


Asunto(s)
Ganglios Linfáticos , Células del Estroma , Ratones , Animales , Ratones Endogámicos C57BL , Células del Estroma/metabolismo , Ganglios Linfáticos/patología , Moléculas de Adhesión Celular/metabolismo , Antígenos CD34/metabolismo
4.
Front Immunol ; 13: 991671, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36119090

RESUMEN

The first wave of Foxp3+ regulatory T cells (Tregs) generated in neonates is critical for the life-long prevention of autoimmunity. Although it is widely accepted that neonates are highly susceptible to infections, the impact of neonatal infections on this first wave of Tregs is completely unknown. Here, we challenged newborn Treg fate-mapping mice (Foxp3eGFPCreERT2xROSA26STOP-eYFP) with the Toll-like receptor (TLR) agonists LPS and poly I:C to mimic inflammatory perturbations upon neonatal bacterial or viral infections, respectively, and subsequently administrated tamoxifen during the first 8 days of life to selectively label the first wave of Tregs. Neonatally-tagged Tregs preferentially accumulated in non-lymphoid tissues (NLTs) when compared to secondary lymphoid organs (SLOs) irrespective of the treatment. One week post challenge, no differences in the frequency and phenotypes of neonatally-tagged Tregs were observed between challenged mice and untreated controls. However, upon aging, a decreased frequency of neonatally-tagged Tregs in both NLTs and SLOs was detected in challenged mice when compared to untreated controls. This decrease became significant 12 weeks post challenge, with no signs of altered Foxp3 stability. Remarkably, this late decrease in the frequency of neonatally-tagged Tregs only occurred when newborns were challenged, as treating 8-days-old mice with TLR agonists did not result in long-lasting alterations of the first wave of Tregs. Combined single-cell T cell receptor (TCR)-seq and RNA-seq revealed that neonatal inflammatory perturbations drastically diminished TCR diversity and long-lastingly altered the transcriptome of neonatally-tagged Tregs, exemplified by lower expression of Tigit, Foxp3, and Il2ra. Together, our data demonstrate that a single, transient encounter with a pathogen in early life can have long-lasting consequences for the first wave of Tregs, which might affect immunological tolerance, prevention of autoimmunity, and other non-canonical functions of tissue-resident Tregs in adulthood.


Asunto(s)
Linfocitos T Reguladores , Transcriptoma , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Lipopolisacáridos/metabolismo , Ratones , Poli I/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Tamoxifeno/metabolismo , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
5.
Immunity ; 55(10): 1813-1828.e9, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36002023

RESUMEN

Lymphatic transport of molecules and migration of myeloid cells to lymph nodes (LNs) continuously inform lymphocytes on changes in drained tissues. Here, using LN transplantation, single-cell RNA-seq, spectral flow cytometry, and a transgenic mouse model for photolabeling, we showed that tissue-derived unconventional T cells (UTCs) migrate via the lymphatic route to locally draining LNs. As each tissue harbored a distinct spectrum of UTCs with locally adapted differentiation states and distinct T cell receptor repertoires, every draining LN was thus populated by a distinctive tissue-determined mix of these lymphocytes. By making use of single UTC lineage-deficient mouse models, we found that UTCs functionally cooperated in interconnected units and generated and shaped characteristic innate and adaptive immune responses that differed between LNs that drained distinct tissues. Lymphatic migration of UTCs is, therefore, a key determinant of site-specific immunity initiated in distinct LNs with potential implications for vaccination strategies and immunotherapeutic approaches.


Asunto(s)
Ganglios Linfáticos , Linfocitos T , Animales , Modelos Animales de Enfermedad , Inmunidad , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T
6.
Cell Mol Immunol ; 18(5): 1211-1221, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33762684

RESUMEN

Intestinal Foxp3+ regulatory T cell (Treg) subsets are crucial players in tolerance to microbiota-derived and food-borne antigens, and compelling evidence suggests that the intestinal microbiota modulates their generation, functional specialization, and maintenance. Selected bacterial species and microbiota-derived metabolites, such as short-chain fatty acids (SCFAs), have been reported to promote Treg homeostasis in the intestinal lamina propria. Furthermore, gut-draining mesenteric lymph nodes (mLNs) are particularly efficient sites for the generation of peripherally induced Tregs (pTregs). Despite this knowledge, the direct role of the microbiota and their metabolites in the early stages of pTreg induction within mLNs is not fully elucidated. Here, using an adoptive transfer-based pTreg induction system, we demonstrate that neither transfer of a dysbiotic microbiota nor dietary SCFA supplementation modulated the pTreg induction capacity of mLNs. Even mice housed under germ-free (GF) conditions displayed equivalent pTreg induction within mLNs. Further molecular characterization of these de novo induced pTregs from mLNs by dissection of their transcriptomes and accessible chromatin regions revealed that the microbiota indeed has a limited impact and does not contribute to the initialization of the Treg-specific epigenetic landscape. Overall, our data suggest that the microbiota is dispensable for the early stages of pTreg induction within mLNs.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Ganglios Linfáticos/inmunología , Mesenterio/inmunología , Linfocitos T Reguladores/inmunología , Animales , Cromatina/metabolismo , Disbiosis/microbiología , Disbiosis/patología , Epigénesis Genética/efectos de los fármacos , Ácidos Grasos Volátiles/farmacología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Perfilación de la Expresión Génica , Ganglios Linfáticos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Reguladores/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transcriptoma/genética
7.
Int J Med Microbiol ; 311(3): 151492, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33676241

RESUMEN

The effective priming of adaptive immune responses depends on the precise dispatching of lymphocytes and antigens into and within lymph nodes (LNs), which are strategically dispersed throughout the body. Over the past decade, a growing body of evidence has advanced our understanding of lymph node stromal cells (LNSCs) from viewing them as mere accessory cells to seeing them as critical cellular players for the modulation of adaptive immune responses. In this review, we summarize current advances on the pivotal roles that LNSCs play in orchestrating adaptive immune responses during homeostasis and infection, and highlight the imprinting of location-specific information by micro-environmental cues into LNSCs, thereby tailoring tissue-specific immune responses.


Asunto(s)
Especialización , Células del Estroma , Homeostasis , Inmunidad , Ganglios Linfáticos
8.
Eur J Microbiol Immunol (Bp) ; 10(2): 98-106, 2020 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-32644940

RESUMEN

Listeria monocytogenes (Lm) is a food-borne pathogen with a high chance of infecting neonates, pregnant women, elderly and immunocompromised individuals. Lm infection in neonates can cause neonatal meningitis and sepsis with a high risk of severe neurological and developmental sequelae and high mortality rates. However, whether an acute neonatal Lm infection causes long-term effects on the immune system persisting until adulthood has not been fully elucidated. Here, we established a neonatal Lm infection model and monitored the composition of major immune cell subsets at defined time points post infection (p.i.) in secondary lymphoid organs and the intestine. Twelve weeks p.i., the CD8+ T cell population was decreased in colon and mesenteric lymph nodes (mLNs) with an opposing increase in the spleen. In the colon, we observed an accumulation of CD4+ and CD8+ effector/memory T cells with an increase of T-bet+ T helper 1 (Th1) cells. In addition, 12 weeks p.i. an altered composition of innate lymphoid cell (ILC) and dendritic cell (DC) subsets was still observed in colon and mLNs, respectively. Together, these findings highlight organ-specific long-term consequences of an acute neonatal Lm infection on both the adaptive and innate immune system.

9.
Front Immunol ; 10: 426, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30906299

RESUMEN

Mammals co-exist with resident microbial ecosystem that is composed of an incredible number and diversity of bacteria, viruses and fungi. Owing to direct contact between resident microbes and mucosal surfaces, both parties are in continuous and complex interactions resulting in important functional consequences. These interactions govern immune homeostasis, host response to infection, vaccination and cancer, as well as predisposition to metabolic, inflammatory and neurological disorders. Here, we discuss recent studies on direct and indirect effects of resident microbiota on regulatory T cells (Tregs) and Th17 cells at the cellular and molecular level. We review mechanisms by which commensal microbes influence mucosa in the context of bioactive molecules derived from resident bacteria, immune senescence, chronic inflammation and cancer. Lastly, we discuss potential therapeutic applications of microbiota alterations and microbial derivatives, for improving resilience of mucosal immunity and combating immunopathology.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Mucosa Intestinal , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología
10.
J Mol Med (Berl) ; 96(12): 1387-1394, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30357435

RESUMEN

Foxp3+ regulatory T cells (Tregs) co-expressing the Th17-lineage specification factor RORγt represent a unique Treg subpopulation that has been reported to be induced upon response to gut microbiota within the intestinal immune system. Hence, RORγt+ Tregs are considered to solely consist of peripherally induced Foxp3+ Tregs (pTregs), and the possibility that also thymus-derived Treg (tTregs) can upregulate RORγt expression and contribute to the pool of RORγt+ Tregs was largely ignored. Here, we expand our knowledge on the origin of RORγt+ Tregs by demonstrating that also tTregs can attain RORγt expression. In transgenic Foxp3∆CNS1-Cre reporter mice, a substantial fraction of CNS1-independent Tregs, predominantly consisting of tTregs, was found to co-express RORγt. In addition, genuine tTregs isolated from thymi of Foxp3hCD2RAGGFP reporter mice initiated RORγt expression both in vitro and in vivo, particularly under inflammatory conditions. In conclusion, our data demonstrate that tTregs can upregulate RORγt expression under inflammatory conditions and that hence RORγt+ Tregs can be regarded as a heterogeneous population consisting of both pTregs and tTregs. KEY MESSAGES: RORγt cannot be considered as a marker for pTregs. CNS1-independent tTregs within the colon display RORγt expression. RORγt can be induced in genuine tTregs, particularly under inflammatory conditions. RORγt+ Tregs are a heterogeneous population consisting of both pTregs and tTregs.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Colitis/inmunología , Colitis/terapia , Inflamación/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Linfocitos T Reguladores/trasplante , Regulación hacia Arriba
11.
Nat Commun ; 9(1): 3903, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254319

RESUMEN

Gut-draining mesenteric lymph nodes (mLNs) are important for inducing peripheral tolerance towards food and commensal antigens by providing an optimal microenvironment for de novo generation of Foxp3+ regulatory T cells (Tregs). We previously identified microbiota-imprinted mLN stromal cells as a critical component in tolerance induction. Here we show that this imprinting process already takes place in the neonatal phase, and renders the mLN stromal cell compartment resistant to inflammatory perturbations later in life. LN transplantation and single-cell RNA-seq uncover stably imprinted expression signatures in mLN fibroblastic stromal cells. Subsetting common stromal cells across gut-draining mLNs and skin-draining LNs further refine their location-specific immunomodulatory functions, such as subset-specific expression of Aldh1a2/3. Finally, we demonstrate that mLN stromal cells shape resident dendritic cells to attain high Treg-inducing capacity in a Bmp2-dependent manner. Thus, crosstalk between mLN stromal and resident dendritic cells provides a robust regulatory mechanism for the maintenance of intestinal tolerance.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Ganglios Linfáticos/inmunología , Células del Estroma/inmunología , Animales , Animales Recién Nacidos , Microambiente Celular/genética , Microambiente Celular/inmunología , Células Dendríticas/metabolismo , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Tolerancia Inmunológica/genética , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/trasplante , Mesenterio/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células del Estroma/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
12.
PLoS One ; 11(6): e0158432, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27348426

RESUMEN

Wnt/ß-catenin signaling pathway plays essential roles in mammalian development and tissue homeostasis. MicroRNAs (miRNAs) are a class of regulators involved in modulating this pathway. In this study, we screened miRNAs regulating Wnt/ß-catenin signaling by using a TopFlash based luciferase reporter. Surprisingly, we found that miR-142 inhibited Wnt/ß-catenin signaling, which was inconsistent with a recent study showing that miR-142-3p targeted Adenomatous Polyposis Coli (APC) to upregulate Wnt/ß-catenin signaling. Due to the discordance, we elaborated experiments by using extensive mutagenesis, which demonstrated that the stem-loop structure was important for miR-142 to efficiently suppress Wnt/ß-catenin signaling. Moreover, the inhibitory effect of miR-142 relies on miR-142-3p rather than miR-142-5p. Further, we found that miR-142-3p directly modulated translation of Ctnnb1 mRNA (encoding ß-catenin) through binding to its 3' untranslated region (3' UTR). Finally, miR-142 was able to repress cell cycle progression by inhibiting active Wnt/ß-catenin signaling. Thus, our findings highlight the inhibitory role of miR-142-3p in Wnt/ß-catenin signaling, which help to understand the complex regulation of Wnt/ß-catenin signaling.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Vía de Señalización Wnt , Regiones no Traducidas 3' , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Proliferación Celular , Humanos , Ratones , MicroARNs/química , Conformación de Ácido Nucleico , Biosíntesis de Proteínas/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...