Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 13(1): 3998, 2022 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-35810180

RESUMEN

Basic processes of the fatty acid metabolism have an important impact on the function of intestinal epithelial cells (IEC). However, while the role of cellular fatty acid oxidation is well appreciated, it is not clear how de novo fatty acid synthesis (FAS) influences the biology of IECs. We report here that interfering with de novo FAS by deletion of the enzyme Acetyl-CoA-Carboxylase (ACC)1 in IECs results in the loss of epithelial crypt structures and a specific decline in Lgr5+ intestinal epithelial stem cells (ISC). Mechanistically, ACC1-mediated de novo FAS supports the formation of intestinal organoids and the differentiation of complex crypt structures by sustaining the nuclear accumulation of PPARδ/ß-catenin in ISCs. The dependency of ISCs on cellular de novo FAS is tuned by the availability of environmental lipids, as an excess delivery of external fatty acids is sufficient to rescue the defect in crypt formation. Finally, inhibition of ACC1 reduces the formation of tumors in colitis-associated colon cancer, together highlighting the importance of cellular lipogenesis for sustaining ISC function and providing a potential perspective to colon cancer therapy.


Asunto(s)
Acetil-CoA Carboxilasa , Lipogénesis , Acetilcoenzima A/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Ácidos Grasos/metabolismo , Lipogénesis/fisiología , Células Madre/metabolismo
2.
Eur J Immunol ; 49(5): 747-757, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30802297

RESUMEN

Infection with Clostridium difficile is one of the major causes of health care acquired diarrhea and colitis. Signaling though MyD88 downstream of TLRs is critical for initiating the early protective host response in mouse models of C. difficile infection (CDI). In the intestine, MyD88 is expressed in various tissues and cell types, such as the intestinal epithelium and mononuclear phagocytes (MNP), including DC or macrophages. Using a genetic gain-of-function system, we demonstrate here that restricting functional MyD88 signaling to the intestinal epithelium, but also to MNPs is sufficient to protect mice during acute CDI by upregulation of the intestinal barrier function and recruitment of neutrophils. Nevertheless, we also show that mice depleted for CD11c-expressing MNPs in the intestine display no major defects in mounting an effective inflammatory response, indicating that the absence of these cells is irrelevant for inducing host protection during acute infection. Together, our results highlight the importance of epithelial-specific MyD88 signaling and demonstrate that although functional MyD88 signaling in DC and macrophages alone is sufficient to correct the phenotype of MyD88-deficiency, these cells do not seem to be essential for host protection in MyD88-sufficient animals during acute infection with C. difficile.


Asunto(s)
Clostridioides difficile/inmunología , Enterocolitis Seudomembranosa/inmunología , Enterocolitis Seudomembranosa/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Enterocolitis Seudomembranosa/microbiología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Mucosa Intestinal/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones
3.
PLoS Pathog ; 13(5): e1006357, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28520792

RESUMEN

MyD88-mediated signaling downstream of Toll-like receptors and the IL-1 receptor family is critically involved in the induction of protective host responses upon infections. Although it is known that MyD88-deficient mice are highly susceptible to a wide range of bacterial infections, the cell type-specific contribution of MyD88 in protecting the host against intestinal bacterial infection is only poorly understood. In order to investigate the importance of MyD88 in specific immune and nonimmune cell types during intestinal infection, we employed a novel murine knock-in model for MyD88 that enables the cell type-specific reactivation of functional MyD88 expression in otherwise MyD88-deficient mice. We report here that functional MyD88 signaling in CD11c+ cells was sufficient to activate intestinal dendritic cells (DC) and to induce the early group 3 innate lymphoid cell (ILC3) response as well as the development of colonic Th17/Th1 cells in response to infection with the intestinal pathogen C. rodentium. In contrast, restricting MyD88 signaling to several other cell types, including macrophages (MO), T cells or ILC3 did not induce efficient intestinal immune responses upon infection. However, we observed that the functional expression of MyD88 in intestinal epithelial cells (IEC) also partially protected the mice during intestinal infection, which was associated with enhanced epithelial barrier integrity and increased expression of the antimicrobial peptide RegIIIγ and the acute phase protein SAA1 by epithelial cells. Together, our data suggest that MyD88 signaling in DC and IEC is both essential and sufficient to induce a full spectrum of host responses upon intestinal infection with C. rodentium.


Asunto(s)
Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Animales , Colon/inmunología , Colon/microbiología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Técnicas de Sustitución del Gen , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Receptores de Interleucina-1/metabolismo , Células TH1/inmunología , Células TH1/microbiología , Células Th17/inmunología , Células Th17/microbiología , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
4.
Eur J Immunol ; 46(9): 2233-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27338930

RESUMEN

Upon antigen-specific or allogeneic activation, T cells sharply increase their metabolic activity to cope with augmented needs for proliferation and effector functions. Therefore, enzymes involved in energy metabolism constitute attractive targets to modulate the activity of pathogenic effector T cells in the setting of graft-versus-host-disease (GVHD). Here, we show that T cells deficient for acetyl-CoA carboxylase 1 (TACC1) are dramatically less pathogenic than wild-type (WT) T cells in a lethal C57BL/6 into BALB/c model of acute GVHD and permitted sustained survival of recipient mice. In line with this clinical observation, higher frequencies of GVHD-suppressing Foxp3(+) regulatory T (Treg) cells were detected in the colon of TACC T-cell recipients. In vitro, T-cell stimulation with allogeneic DCs induced higher proportions of Treg cells but also led to diminished proliferation of TACC1 T cells compared to WT T cells. Furthermore, TACC1 T cells activated by allogeneic DCs showed impaired glycolysis and lipid synthesis. Thus, targeting de novo fatty acid synthesis via acetyl-CoA carboxylase inhibition may be a promising new strategy to prevent GVHD.


Asunto(s)
Acetil-CoA Carboxilasa/antagonistas & inhibidores , Ácidos Grasos/biosíntesis , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Acetil-CoA Carboxilasa/deficiencia , Traslado Adoptivo , Animales , Biomarcadores , Trasplante de Médula Ósea , Diferenciación Celular , Modelos Animales de Enfermedad , Eliminación de Gen , Enfermedad Injerto contra Huésped/mortalidad , Inmunofenotipificación , Macrólidos/farmacología , Masculino , Ratones , Fenotipo , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Homólogo
5.
PLoS One ; 6(9): e24241, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21935389

RESUMEN

Interleukin(IL)-18 is a pleiotrophic cytokine with functions in immune modulation, angiogenesis and bone metabolism. In this study, the potential of IL-18 as an immunotherapy for prostate cancer (PCa) was examined using the murine model of prostate carcinoma, RM1 and a bone metastatic variant RM1(BM)/B4H7-luc. RM1 and RM1(BM)/B4H7-luc cells were stably transfected to express bioactive IL-18. These cells were implanted into syngeneic immunocompetent mice, with or without an IL-18-neutralising antibody (αIL-18, SK113AE4). IL-18 significantly inhibited the growth of both subcutaneous and orthotopic RM1 tumors and the IL-18 neutralizing antibody abrogated the tumor growth-inhibition. In vivo neutralization of interferon-gamma (IFN-γ) completely eliminated the anti-tumor effects of IL-18 confirming an essential role of IFN-γ as a down-stream mediator of the anti-tumor activity of IL-18. Tumors from mice in which IL-18 and/or IFN-γ was neutralized contained significantly fewer CD4(+) and CD8(+) T cells than those with functional IL-18. The essential role of adaptive immunity was demonstrated as tumors grew more rapidly in RAG1(-/-) mice or in mice depleted of CD4(+) and/or CD8(+) cells than in normal mice. The tumors in RAG1(-/-) mice were also significantly smaller when IL-18 was present, indicating that innate immune mechanisms are involved. IL-18 also induced an increase in tumor infiltration of macrophages and neutrophils but not NK cells. In other experiments, direct injection of recombinant IL-18 into established tumors also inhibited tumor growth, which was associated with an increase in intratumoral macrophages, but not T cells. These results suggest that local IL-18 in the tumor environment can significantly potentiate anti-tumor immunity in the prostate and clearly demonstrate that this effect is mediated by innate and adaptive immune mechanisms.


Asunto(s)
Inmunidad Adaptativa/inmunología , Carcinoma/metabolismo , Inmunidad Innata/inmunología , Interleucina-18/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Quimiocina CCL5/metabolismo , Femenino , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunohistoquímica , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Interleucina-4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
6.
J Immunol ; 182(9): 5789-99, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380827

RESUMEN

Stromal cells in lymphoid tissues regulate lymphocyte recruitment and survival through the expression of specific chemokines and cytokines. During inflammation, the same signals recruit lymphocytes to the site of injury; however, the "lymphoid" stromal (LS) cells producing these signals remain poorly characterized. We find that mouse inflammatory lesions and tumors develop gp38(+) LS cells, in recapitulation of the development of LS cells early during the ontogeny of lymphoid organs and the intestine, and express a set of genes that promotes the development of lymphocyte-permissive tissues. These gp38(+) LS cells are induced by a robust pathway that requires myeloid cells but not known Toll- or NOD-like receptors, the inflammasome, or adaptive immunity. Parabiosis and inducible genetic cell fate mapping experiments indicate that local precursors, presumably resident fibroblasts rather that circulating precursors, massively proliferate and give rise to LS cells during inflammation. Our results show that LS cells are both programmed during ontogeny and reinduced during inflammation.


Asunto(s)
Movimiento Celular/inmunología , Inflamación/inmunología , Inflamación/patología , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Animales , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Inflamación/embriología , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Mucosa Intestinal/embriología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/patología , Tejido Linfoide/embriología , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Células del Estroma/inmunología , Células del Estroma/metabolismo , Células del Estroma/patología
7.
Immunity ; 29(6): 958-70, 2008 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-19084435

RESUMEN

Natural killer (NK) cells are innate lymphocytes with spontaneous antitumor activity, and they produce interferon-gamma (IFN-gamma) that primes immune responses. Whereas T helper cell subsets differentiate from naive T cells via specific transcription factors, evidence for NK cell diversification is limited. In this report, we characterized intestinal lymphocytes expressing the NK cell natural cytotoxicity receptor NKp46. Gut NKp46+ cells were distinguished from classical NK cells by limited IFN-gamma production and absence of perforin, whereas several subsets expressed the nuclear hormone receptor retinoic acid receptor-related orphan receptor t (RORgammat) and interleukin-22 (IL-22). Intestinal NKp46+IL-22+ cells were generated via a local process that was conditioned by commensal bacteria and required RORgammat. Mice lacking IL-22-producing NKp46+ cells showed heightened susceptibility to the pathogen Citrobacter rodentium, consistent with a role for intestinal NKp46+ cells in immune protection. RORgammat-driven diversification of intestinal NKp46+ cells thereby specifies an innate cellular defense mechanism that operates at mucosal surfaces.


Asunto(s)
Antígenos Ly/inmunología , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Interleucinas/inmunología , Intestinos/inmunología , Células Asesinas Naturales/inmunología , Receptor 1 Gatillante de la Citotoxidad Natural/inmunología , Animales , Antígenos Ly/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Inmunidad Innata , Inmunidad Mucosa/inmunología , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Perforina/inmunología , Perforina/metabolismo , Receptores de Ácido Retinoico/inmunología , Receptores de Ácido Retinoico/metabolismo , Receptores de Hormona Tiroidea/inmunología , Receptores de Hormona Tiroidea/metabolismo , Transducción de Señal/inmunología , Interleucina-22
8.
Infect Immun ; 76(9): 3881-90, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18573894

RESUMEN

The induction of proinflammatory cytokines such as gamma interferon (IFN-gamma) and tumor necrosis factor alpha is crucial for the early control of bacterial infections. Since interleukin-18 (IL-18) acts as a potent inducer of IFN-gamma, it might play an important role in the induction of a protective immune response in listeriosis. We used a murine model of systemic Listeria monocytogenes infection to study the immune response to these intracellular bacteria in the absence of IL-18. For this purpose, IL-18-deficient mice and mice treated with anti-IL-18 neutralizing antibody were infected with L. monocytogenes, and their innate and adaptive immune responses were compared to those of control mice. Unexpectedly, we found that mice deficient in IL-18 were partially resistant to primary infection with L. monocytogenes. At day 3 after infection, the numbers of listeriae in the livers and spleens of control mice were up to 500 times higher than those in IL-18-deficient or anti-IL-18 antibody-treated mice. In addition, the level of proinflammatory cytokines was markedly reduced in IL-18-deficient mice. Enhanced resistance to L. monocytogenes infection in IL-18-deficient mice was accompanied by increased numbers of leukocytes and reduced apoptosis in the spleen 48 to 72 h after infection. In contrast, control and IL-18-deficient mice showed no significant differences in their abilities to mount a protective L. monocytogenes-specific T-cell response.


Asunto(s)
Susceptibilidad a Enfermedades , Interleucina-18/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Animales , Apoptosis , Citocinas/biosíntesis , Citocinas/sangre , Interleucina-18/deficiencia , Leucocitos/inmunología , Hígado/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/microbiología , Bazo/patología , Linfocitos T/inmunología
9.
J Exp Med ; 205(6): 1381-93, 2008 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-18504307

RESUMEN

The nuclear hormone receptor retinoic acid receptor-related orphan receptor gamma t (RORgamma t) is required for the generation of T helper 17 cells expressing the proinflammatory cytokine interleukin (IL)-17. In vivo, however, less than half of RORgamma t(+) T cells express IL-17. We report here that RORgamma t(+) T alphabeta cells include Foxp3(+) cells that coexist with IL-17-producing RORgamma t(+) T alphabeta cells in all tissues examined. The Foxp3(+) RORgamma t(+) T alphabeta express IL-10 and CCL20, and function as regulatory T cells. Furthermore, the ratio of Foxp3(+) to IL-17-producing RORgamma t(+) T alphabeta cells remains remarkably constant in mice enduring infection and inflammation. This equilibrium is tuned in favor of IL-10 production by Foxp3 and CCL20, and in favor of IL-17 production by IL-6 and IL-23. In the lung and skin, the largest population of RORgamma t(+) T cells express the gammadelta T cell receptor and produce the highest levels of IL-17 independently of IL-6. Thus, potentially antagonistic proinflammatory IL-17-producing and regulatory Foxp3(+) RORgamma t(+) T cells coexist and are tightly controlled, suggesting that a perturbed equilibrium in RORgamma t(+) T cells might lead to decreased immunoreactivity or, in contrast, to pathological inflammation.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Interleucina-10/inmunología , Interleucina-17/inmunología , Receptores de Ácido Retinoico/inmunología , Receptores de Hormona Tiroidea/inmunología , Linfocitos T/inmunología , Animales , Citometría de Flujo , Genes Reporteros , Variación Genética , Inflamación/inmunología , Interleucina-6/inmunología , Ratones , Ratones Transgénicos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Receptores de Interleucina-12/inmunología , Linfocitos T Reguladores/inmunología
10.
Eur J Immunol ; 34(11): 3197-207, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15368276

RESUMEN

Oral infection of C57BL/6 mice with Toxoplasma gondii results in small intestinal Th1-type immunopathology mediated by local production of IFN-gamma, TNF-alpha, and NO. To analyze whether the proinflammatory cytokines IL-12 and IL-18 play a role in the induction of immunopathology, IL-12p35/p40(-/-) and IL-18(-/-) mice were orally infected with T. gondii. Wild-type mice developed massive necrosis in their small intestines and died 7-10 days post infection. Even though IL-12p35/40(-/-) mice did not develop the necrosis they all died between day 9 and 11 after infection. In contrast, 50% of IL-18(-/-) mice died during the acute phase of infection. Compared to wild-type mice, IL-12p35/p40(-/-) but not IL-18(-/-) mice showed significantly higher parasite numbers in their small intestines and significantly higher numbers of parasite-associated inflammatory foci in their livers. IFN-gamma production was similar in infected wild-type and IL-18(-/-) mice but significantly decreased in IL-12p35/p40(-/-) mice. Treatment of mice with anti-IL-12- or anti-IL-18 antibodies after infection prevented the development of intestinal necrosis. These results reveal that both IL-12 and IL-18 play an important role in the development of intestinal immunopathology following oral infection with T. gondii. However, IL-12 is dominant over IL-18 in the host defense against parasite replication. Therefore, neutralization of IL-18 (rather than TNF-alpha, IL-12, and IFN-gamma) may be a safe strategy for the treatment of Th1-associated diseases.


Asunto(s)
Interleucina-12/inmunología , Interleucina-18/inmunología , Intestino Delgado/inmunología , Subunidades de Proteína/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Encéfalo/parasitología , Encéfalo/patología , Femenino , Histocitoquímica , Interferón gamma/inmunología , Subunidad p35 de la Interleucina-12 , Subunidad p40 de la Interleucina-12 , Intestino Delgado/parasitología , Intestino Delgado/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/inmunología , Organismos Libres de Patógenos Específicos , Bazo/parasitología , Bazo/patología , Toxoplasmosis/parasitología , Toxoplasmosis/patología , Factor de Necrosis Tumoral alfa/inmunología
11.
Pathobiology ; 70(3): 164-9, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12571421

RESUMEN

Interleukin (IL)-18 is a cytokine with a broad array of effector functions, the most prominent of which is to act synergistically with IL-12 in interferon-gamma production and the induction of a strong T-helper-1-mediated immune response. In addition, IL-18 also upregulates the production of proinflammatory cytokines such as IL-1 and tumor necrosis factor-alpha. Analysis of IL-18-deficient mice revealed an important role of IL-18 in the activation of macrophages and natural killer cells in the context of infection with intracellular bacteria or parasites. In humans, it was reported that IL-18 is elevated at sites of inflammation in inflammatory bowel disease (IBD), particularly in Crohn's disease, suggesting a possible role for IL-18 in the development and persistence of IBD. In this review we summarize recent findings on the functional role of IL-18 in the pathogenesis of colitis with a special focus on murine models of IBD. The neutralizing mouse anti-mouse IL-18 antibodies generated in our group should facilitate the evaluation of the efficiency of therapeutic blockade of endogenous IL-18 in chronic mouse models of colitis besides the use of recombinant forms of the inhibitory IL-18-binding protein.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Colitis Ulcerosa/terapia , Modelos Animales de Enfermedad , Interleucina-18/antagonistas & inhibidores , Animales , Anticuerpos Bloqueadores/biosíntesis , Colitis Ulcerosa/genética , Colitis Ulcerosa/inmunología , Inmunosupresores/uso terapéutico , Inmunoterapia , Interleucina-18/genética , Interleucina-18/inmunología , Ratones , Ratones Endogámicos , Ratones Noqueados , ARN sin Sentido/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA