Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nature ; 631(8022): 857-866, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38987586

RESUMEN

Systemic lupus erythematosus (SLE) is prototypical autoimmune disease driven by pathological T cell-B cell interactions1,2. Expansion of T follicular helper (TFH) and T peripheral helper (TPH) cells, two T cell populations that provide help to B cells, is a prominent feature of SLE3,4. Human TFH and TPH cells characteristically produce high levels of the B cell chemoattractant CXCL13 (refs. 5,6), yet regulation of T cell CXCL13 production and the relationship between CXCL13+ T cells and other T cell states remains unclear. Here, we identify an imbalance in CD4+ T cell phenotypes in patients with SLE, with expansion of PD-1+/ICOS+ CXCL13+ T cells and reduction of CD96hi IL-22+ T cells. Using CRISPR screens, we identify the aryl hydrocarbon receptor (AHR) as a potent negative regulator of CXCL13 production by human CD4+ T cells. Transcriptomic, epigenetic and functional studies demonstrate that AHR coordinates with AP-1 family member JUN to prevent CXCL13+ TPH/TFH cell differentiation and promote an IL-22+ phenotype. Type I interferon, a pathogenic driver of SLE7, opposes AHR and JUN to promote T cell production of CXCL13. These results place CXCL13+ TPH/TFH cells on a polarization axis opposite from T helper 22 (TH22) cells and reveal AHR, JUN and interferon as key regulators of these divergent T cell states.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Linfocitos T CD4-Positivos , Quimiocina CXCL13 , Interferón Tipo I , Lupus Eritematoso Sistémico , Proteínas Proto-Oncogénicas c-jun , Receptores de Hidrocarburo de Aril , Femenino , Humanos , Masculino , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Quimiocina CXCL13/metabolismo , Epigenómica , Perfilación de la Expresión Génica , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interleucina-22/inmunología , Interleucina-22/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Lupus Eritematoso Sistémico/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
2.
Cell Rep ; 43(6): 114292, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38823020

RESUMEN

Interleukin (IL)-22 promotes host-microbiota homeostasis. We sought to identify microbiota metabolite(s) that drive intestinal IL-22 production. We observed that exposing Peyer's patch cells (PPCs), ex vivo, to fecal supernatants (FSs) recapitulates fermentable fiber- and microbiota-dependent IL-22 production, and cellular sources thereof, thus supporting the use of this model. An interrogation of FSs generated from mice fed the fermentable fiber inulin (FS-Inu) revealed that its IL-22-inducing activity is mediated by heat-labile protein. Fractionation of FS-Inu by ion-exchange chromatography, and subsequent proteomic analysis of IL-22-inducing fractions, indicates that outer membrane protein A (OmpA) might be a microbial driver of IL-22 expression. Concomitantly, recombinant OmpA from Parabacteroides goldsteinii, which is enriched by an inulin diet, induces IL-22 production and expression of the IL-22-dependent genes REG3γ and -ß, in PPCs and mice. Thus, OmpA is one bacterial inducer of IL-22 expression, potentially linking diet, mucosal immune homeostasis, and gut health.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Interleucina-22 , Animales , Ratones , Proteínas de la Membrana Bacteriana Externa/metabolismo , Heces/microbiología , Microbioma Gastrointestinal , Interleucina-22/metabolismo , Interleucinas/metabolismo , Inulina/metabolismo , Ratones Endogámicos C57BL , Proteínas Asociadas a Pancreatitis/metabolismo
3.
Nature ; 629(8012): 669-678, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600382

RESUMEN

Interleukin 22 (IL-22) has a non-redundant role in immune defence of the intestinal barrier1-3. T cells, but not innate lymphoid cells, have an indispensable role in sustaining the IL-22 signalling that is required for the protection of colonic crypts against invasion during infection by the enteropathogen Citrobacter rodentium4 (Cr). However, the intestinal epithelial cell (IEC) subsets targeted by T cell-derived IL-22, and how T cell-derived IL-22 sustains activation in IECs, remain undefined. Here we identify a subset of absorptive IECs in the mid-distal colon that are specifically targeted by Cr and are differentially responsive to IL-22 signalling. Major histocompatibility complex class II (MHCII) expression by these colonocytes was required to elicit sustained IL-22 signalling from Cr-specific T cells, which was required to restrain Cr invasion. Our findings explain the basis for the regionalization of the host response to Cr and demonstrate that epithelial cells must elicit MHCII-dependent help from IL-22-producing T cells to orchestrate immune protection in the intestine.


Asunto(s)
Citrobacter rodentium , Colon , Células Epiteliales , Mucosa Intestinal , Linfocitos T , Animales , Femenino , Masculino , Ratones , Citrobacter rodentium/inmunología , Colon/citología , Colon/inmunología , Colon/microbiología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Células Epiteliales/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Interleucina-22/inmunología , Interleucina-22/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/citología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Transducción de Señal/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
4.
Infect Immun ; 92(5): e0009924, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38557196

RESUMEN

The mouse pathogen Citrobacter rodentium is utilized as a model organism for studying infections caused by the human pathogens enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) and to elucidate mechanisms of mucosal immunity. In response to C. rodentium infection, innate lymphoid cells and T cells secrete interleukin (IL)-22, a cytokine that promotes mucosal barrier function. IL-22 plays a pivotal role in enabling mice to survive and recover from C. rodentium infection, although the exact mechanisms involved remain incompletely understood. Here, we investigated whether particular components of the host response downstream of IL-22 contribute to the cytokine's protective effects during C. rodentium infection. In line with previous research, mice lacking the IL-22 gene (Il22-/- mice) were highly susceptible to C. rodentium infection. To elucidate the role of specific antimicrobial proteins modulated by IL-22, we infected the following knockout mice: S100A9-/- (calprotectin), Lcn2-/- (lipocalin-2), Reg3b-/- (Reg3ß), Reg3g-/- (Reg3γ), and C3-/- (C3). All knockout mice tested displayed a considerable level of resistance to C. rodentium infection, and none phenocopied the lethality observed in Il22-/- mice. By investigating another arm of the IL-22 response, we observed that C. rodentium-infected Il22-/- mice exhibited an overall decrease in gene expression related to intestinal barrier integrity as well as significantly elevated colonic inflammation, gut permeability, and pathogen levels in the spleen. Taken together, these results indicate that host resistance to lethal C. rodentium infection may depend on multiple antimicrobial responses acting in concert, or that other IL-22-regulated processes, such as tissue repair and maintenance of epithelial integrity, play crucial roles in host defense to attaching and effacing pathogens.


Asunto(s)
Citrobacter rodentium , Infecciones por Enterobacteriaceae , Interleucina-22 , Animales , Ratones , Citrobacter rodentium/inmunología , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Interleucina-22/genética , Interleucina-22/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/metabolismo , Proteínas Asociadas a Pancreatitis/inmunología
5.
Food Funct ; 15(10): 5364-5381, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38639049

RESUMEN

Invasive candidiasis may be caused by Candida albicans (C. albicans) colonization of the intestinal tract. Preventing intestinal damage caused by Candida albicans infection and protecting intestinal barrier function have become a critical issue. Integrated analyses of the microbiome with metabolome revealed a remarkable shift of the gut microbiota and tryptophan metabolites, kynurenic acid (KynA), and indolacrylic acid (IA) in mice infected with C. albicans. The transcriptome sequencing indicated that differentially expressed genes were significantly associated with innate immune responses and inflammatory responses. The results of this study suggest that KynA and IA (KI) can alleviate intestinal damage caused by Candida albicans infection in mice by reducing intestinal permeability, increasing intestinal firmness, alleviating intestinal inflammation, and reducing the secretion of interleukin-22 (IL-22) in the 3 groups of colon innate lymphoid cells (ILC3). We performed a fecal microbiota transplantation (FMT) experiment and found that the intestinal barrier function, inflammation, and IL-22 secretion of ILC3 in the colon lamina propria of the recipient mice subjected to C. albicans infection and KI treatment were consistent with the trends of the donor mice. Our results suggest that tryptophan metabolites may directly regulate colon lamina ILC3 to promote intestinal resistance to C. albicans invasion, or indirectly regulate the ILC3 secretion of IL-22 to play a protective role in the intestinal barrier by affecting intestinal microorganisms, which may become a potential target for alleviating intestine borne C. albicans infection.


Asunto(s)
Candida albicans , Candidiasis , Colon , Microbioma Gastrointestinal , Interleucina-22 , Interleucinas , Mucosa Intestinal , Triptófano , Animales , Ratones , Candida albicans/efectos de los fármacos , Candidiasis/tratamiento farmacológico , Candidiasis/inmunología , Candidiasis/microbiología , Colon/microbiología , Colon/inmunología , Colon/metabolismo , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad Innata , Interleucina-22/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Triptófano/metabolismo , Triptófano/farmacología
6.
Dig Dis Sci ; 69(5): 1691-1700, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38466463

RESUMEN

BACKGROUND: Acute pancreatitis (AP) is one of the most common acute abdominal disorders; due to the lack of specific treatment, the treatment of acute pancreatitis, especially serious acute pancreatitis (SAP), is difficult and challenging. We will observe the changes of Interleukin -22 levels in acute pancreatitis animal models, and explore the mechanism of Interleukin -22 in acute pancreatitis. OBJECTIVE: This study aims to assess the potential protective effect of Interleukin -22 on caerulein-induced acute pancreatitis and to explore its mechanism. METHODS: Blood levels of amylase and lipase and Interleukin -22 were assessed in mice with acute pancreatitis. In animal model and cell model of caerulein-induced acute pancreatitis, the mRNA levels of P62 and Beclin-1 were determined using PCR, and the protein expression of P62, LC3-II, mTOR, AKT, p-mTOR, and p-AKT were evaluated through Western blot analysis. RESULTS: Interleukin -22 administration reduced blood amylase and lipase levels and mitigated tissue damage in acute pancreatitis mice model. Interleukin -22 inhibited the relative mRNA levels of P62 and Beclin-1, and the Interleukin -22 group showed a decreased protein expression of LC3-II and P62 and the phosphorylation of the AKT/mTOR pathway. Furthermore, we obtained similar results in the cell model of acute pancreatitis. CONCLUSION: This study suggests that Interleukin -22 administration could alleviate pancreatic damage in caerulein-induced acute pancreatitis. This effect may result from the activation of the AKT/mTOR pathway, leading to the inhibition of autophagy. Consequently, Interleukin -22 shows potential as a treatment.


Asunto(s)
Ceruletida , Interleucina-22 , Interleucinas , Pancreatitis , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Masculino , Ratones , Enfermedad Aguda , Amilasas/sangre , Amilasas/metabolismo , Autofagia/efectos de los fármacos , Beclina-1/metabolismo , Beclina-1/genética , Ceruletida/efectos adversos , Ceruletida/metabolismo , Modelos Animales de Enfermedad , Interleucina-22/metabolismo , Interleucina-22/farmacología , Interleucinas/metabolismo , Interleucinas/farmacología , Lipasa/sangre , Lipasa/metabolismo , Ratones Endogámicos C57BL , Páncreas/metabolismo , Páncreas/patología , Páncreas/efectos de los fármacos , Pancreatitis/inducido químicamente , Pancreatitis/metabolismo , Pancreatitis/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
7.
Immun Inflamm Dis ; 12(2): e1144, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38363052

RESUMEN

Interleukin-22 (IL-22) is an important cytokine in the intestinal environment. IL-22 is mainly produced by immune cells and targeted at nonimmune cells such as epithelial and stromal cells in a broad array of tissues such as -but not restricted to- the liver and adipose tissue. IL-22 therefore connects immune functions with metabolic functions of the host, and since it is induced by the microbiota, connects host functioning to the outside environment. IL-22 induces epithelial cell proliferation aiding in rapid epithelium regeneration and wound healing. Additionally, IL-22 activates antiapoptotic genes and DNA damage response pathways, enhancing epithelial cell survival. Recently, it has also been shown that IL-22 induces Paneth cell differentiation in humans. However, IL-22 can also contribute to intestinal epithelium damage and reduces microbial diversity in the intestine directly or indirectly by inducing excessive antimicrobial peptide production by epithelial cells. Moreover, IL-22 enhances angiogenesis and may therefore support tumorigenesis in the intestine. In conclusion, it appears that whether IL-22 has a beneficial or harmful effect in the mammalian intestine largely depends on its regulation. This review aims to provide a comprehensive overview of the current literature and emphasizes that IL-22 signaling outcome depends on the timing and duration of IL-22 production, the presence of it regulators such as IL-22BP, and the specific location of the cytokine production in the gastrointestinal tract.


Asunto(s)
Interleucina-22 , Intestinos , Animales , Humanos , Homeostasis , Interleucina-22/metabolismo , Mucosa Intestinal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA