Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 15(11): 1055-1063, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25282159

RESUMEN

TRPV1 is a Ca(2+)-permeable channel studied mostly as a pain receptor in sensory neurons. However, its role in other cell types is poorly understood. Here we found that TRPV1 was functionally expressed in CD4(+) T cells, where it acted as a non-store-operated Ca(2+) channel and contributed to T cell antigen receptor (TCR)-induced Ca(2+) influx, TCR signaling and T cell activation. In models of T cell-mediated colitis, TRPV1 promoted colitogenic T cell responses and intestinal inflammation. Furthermore, genetic and pharmacological inhibition of TRPV1 in human CD4(+) T cells recapitulated the phenotype of mouse Trpv1(-/-) CD4(+) T cells. Our findings suggest that inhibition of TRPV1 could represent a new therapeutic strategy for restraining proinflammatory T cell responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inflamación/inmunología , Activación de Linfocitos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Canales Catiónicos TRPV/genética , Anilidas/farmacología , Animales , Linfocitos T CD4-Positivos/citología , Calcio/metabolismo , Canales de Calcio/inmunología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/inmunología , Capsaicina/farmacología , Células Cultivadas , Cinamatos/farmacología , Colitis/inmunología , Humanos , Interleucina-10/genética , Intestinos/inmunología , Intestinos/patología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fármacos del Sistema Sensorial/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/biosíntesis
2.
Proc Natl Acad Sci U S A ; 117(35): 21432-21440, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817524

RESUMEN

Approximately half of eukaryotic proteins reside in organelles. To reach their correct destination, such proteins harbor targeting signals recognized by dedicated targeting pathways. It has been shown that differences in targeting signals alter the efficiency in which proteins are recognized and targeted. Since multiple proteins compete for any single pathway, such differences can affect the priority for which a protein is catered. However, to date the entire repertoire of proteins with targeting priority, and the mechanisms underlying it, have not been explored for any pathway. Here we developed a systematic tool to study targeting priority and used the Pex5-mediated targeting to yeast peroxisomes as a model. We titrated Pex5 out by expressing high levels of a Pex5-cargo protein and examined how the localization of each peroxisomal protein is affected. We found that while most known Pex5 cargo proteins were outcompeted, several cargo proteins were not affected, implying that they have high targeting priority. This priority group was dependent on metabolic conditions. We dissected the mechanism of priority for these proteins and suggest that targeting priority is governed by different parameters, including binding affinity of the targeting signal to the cargo factor, the number of binding interfaces to the cargo factor, and more. This approach can be modified to study targeting priority in various organelles, cell types, and organisms.


Asunto(s)
Señales de Direccionamiento al Peroxisoma , Receptor de la Señal 1 de Direccionamiento al Peroxisoma/metabolismo , Peroxisomas/metabolismo , Prueba de Estudio Conceptual , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/metabolismo
3.
Nature ; 519(7541): 57-62, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25731159

RESUMEN

Inflammation promotes regeneration of injured tissues through poorly understood mechanisms, some of which involve interleukin (IL)-6 family members, the expression of which is elevated in many diseases including inflammatory bowel diseases and colorectal cancer. Here we show in mice and human cells that gp130, a co-receptor for IL-6 cytokines, triggers activation of YAP and Notch, transcriptional regulators that control tissue growth and regeneration, independently of the gp130 effector STAT3. Through YAP and Notch, intestinal gp130 signalling stimulates epithelial cell proliferation, causes aberrant differentiation and confers resistance to mucosal erosion. gp130 associates with the related tyrosine kinases Src and Yes, which are activated on receptor engagement to phosphorylate YAP and induce its stabilization and nuclear translocation. This signalling module is strongly activated upon mucosal injury to promote healing and maintain barrier function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Receptor gp130 de Citocinas/metabolismo , Células Epiteliales/citología , Inflamación/metabolismo , Mucosa Intestinal/citología , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Regeneración , Animales , Peso Corporal , Proteínas de Ciclo Celular , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Activación Enzimática , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células HEK293 , Homeostasis , Humanos , Inflamación/patología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Proteínas Proto-Oncogénicas c-yes/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Regulación hacia Arriba , Proteínas Señalizadoras YAP
4.
Proc Natl Acad Sci U S A ; 114(7): 1643-1648, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28130546

RESUMEN

Loss of tumor suppressor adenomatous polyposis coli (APC) activates ß-catenin to initiate colorectal tumorigenesis. However, ß-catenin (CTNNB1) activating mutations rarely occur in human colorectal cancer (CRC). We found that APC loss also results in up-regulation of IL-6 signal transducer (IL-6ST/gp130), thereby activating Src family kinases (SFKs), YAP, and STAT3, which are simultaneously up-regulated in the majority of human CRC. Although, initial YAP activation, which stimulates IL6ST gene transcription, may be caused by reduced serine phosphorylation, sustained YAP activation depends on tyrosine phosphorylation by SFKs, whose inhibition, along with STAT3-activating JAK kinases, causes regression of established colorectal tumors. These results explain why APC loss is a more potent initiating event than the mere activation of CTNNB1.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Receptor gp130 de Citocinas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Adulto , Anciano , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptor gp130 de Citocinas/genética , Femenino , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Mutación , beta Catenina/genética , beta Catenina/metabolismo
6.
Infect Immun ; 86(6)2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29610256

RESUMEN

Rodents are a natural host for the dimorphic pathogenic fungi Coccidioides immitis and Coccidioides posadasii, and mice are a good model for human infection. Humans and rodents both express Dectin-1 and Toll-like receptor 2 (TLR2) on myeloid cells, and those receptors collaborate to maximize the cytokine/chemokine responses to spherules (the tissue form of the fungi) and to formalin-killed spherules (FKS). We showed that Dectin-1 is necessary for resistance to pulmonary coccidioidomycosis, but the importance of TLR2 in vivo is uncertain. Myeloid differentiation factor 88 (MyD88) is the adapter protein for TLR2 and -4, interleukin-1R1 (IL-1R1), and IL-18R1. MyD88/TRIF-/- and MyD88-/- mice were equally susceptible to C. immitis infection, in contrast to C57BL/6 (B6) controls. Of the four surface receptors, only IL-1R1 was required for resistance to C. immitis, partially explaining the susceptibility of MyD88-/- mice. We also found that FKS stimulated production of IL-1Ra by bone marrow-derived dendritic cells (BMDCs), independent of MyD88 and Dectin-1. There also was a very high concentration of IL-1Ra in the lungs of infected B6 mice, supporting the potential importance of this regulatory IL-1 family protein in the largely ineffective response of B6 mice to coccidioidomycosis. These results suggest that IL-1R1 signaling is important for defense against C. immitis infection.


Asunto(s)
Coccidioides/inmunología , Coccidioidomicosis/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Animales , Células Dendríticas , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Receptores Tipo I de Interleucina-1/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(5): 1529-34, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25605931

RESUMEN

The inductive role of dendritic cells (DC) in Th2 differentiation has not been fully defined. We addressed this gap in knowledge by focusing on signaling events mediated by the heterotrimeric GTP binding proteins Gαs, and Gαi, which respectively stimulate and inhibit the activation of adenylyl cyclases and the synthesis of cAMP. We show here that deletion of Gnas, the gene that encodes Gαs in mouse CD11c(+) cells (Gnas(ΔCD11c) mice), and the accompanying decrease in cAMP provoke Th2 polarization and yields a prominent allergic phenotype, whereas increases in cAMP inhibit these responses. The effects of cAMP on DC can be demonstrated in vitro and in vivo and are mediated via PKA. Certain gene products made by Gnas(ΔCD11c) DC affect the Th2 bias. These findings imply that G protein-coupled receptors, the physiological regulators of Gαs and Gαi activation and cAMP formation, act via PKA to regulate Th bias in DC and in turn, Th2-mediated immunopathologies.


Asunto(s)
Asma/inmunología , AMP Cíclico/metabolismo , Células Dendríticas/metabolismo , Hipersensibilidad/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Cromograninas , Células Dendríticas/citología , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Ratones
8.
Gut ; 66(9): 1584-1596, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27325418

RESUMEN

OBJECTIVE: Transient receptor potential ankyrin-1 (TRPA1) and transient receptor potential vanilloid-1 (TRPV1) are calcium (Ca2+)-permeable ion channels mostly known as pain receptors in sensory neurons. However, growing evidence suggests their crucial involvement in the pathogenesis of IBD. We explored the possible contribution of TRPA1 and TRPV1 to T-cell-mediated colitis. DESIGN: We evaluated the role of Trpa1 gene deletion in two models of experimental colitis (ie, interleukin-10 knockout and T-cell-adoptive transfer models). We performed electrophysiological and Ca2+ imaging studies to analyse TRPA1 and TRPV1 functions in CD4+ T cells. We used genetic and pharmacological approaches to evaluate TRPV1 contribution to the phenotype of Trpa1-/- CD4+ T cells. We also analysed TRPA1 and TRPV1 gene expression and TRPA1+TRPV1+ T cell infiltration in colonic biopsies from patients with IBD. RESULTS: We identified a protective role for TRPA1 in T-cell-mediated colitis. We demonstrated the functional expression of TRPA1 on the plasma membrane of CD4+ T cells and identified that Trpa1-/- CD4+ T cells have increased T-cell receptor-induced Ca2+ influx, activation profile and differentiation into Th1-effector cells. This phenotype was abrogated upon genetic deletion or pharmacological inhibition of the TRPV1 channel in mouse and human CD4+ T cells. Finally, we found differential regulation of TRPA1 and TRPV1 gene expression as well as increased infiltration of TRPA1+TRPV1+ T cells in the colon of patients with IBD. CONCLUSIONS: Our study indicates that TRPA1 inhibits TRPV1 channel activity in CD4+ T cells, and consequently restrains CD4+ T-cell activation and colitogenic responses. These findings may therefore have therapeutic implications for human IBD.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Canales Catiónicos TRPV , Canales de Potencial de Receptor Transitorio , Animales , Biopsia/métodos , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Colitis/genética , Colitis/metabolismo , Colitis/patología , Colon/metabolismo , Colon/patología , Modelos Animales de Enfermedad , Expresión Génica/fisiología , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Ratones , Factores Protectores , Estadística como Asunto , Canal Catiónico TRPA1 , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
9.
FEMS Yeast Res ; 16(7)2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27650704

RESUMEN

The minimal description of a growing cell consists of self-replicating ribosomes translating the cellular proteome. While neglecting all other cellular components, this model provides key insights into the control and limitations of growth rate. It shows, for example, that growth rate is maximized when ribosomes work at full capacity, explains the linear relation between growth rate and the ribosome fraction of the proteome and defines the maximal possible growth rate. This ribosome-centered model also highlights the challenge of coordinating cell growth with related processes such as cell division or nutrient production. Coordination is promoted when ribosomes don't translate at maximal capacity, as it allows escaping strict exponential growth. Recent data support the notion that multiple cellular processes limit growth. In particular, increasing transcriptional demand may be as deleterious as increasing translational demand, depending on growth conditions. Consistent with the idea of trade-off, cells may forgo maximal growth to enable more efficient interprocess coordination and faster adaptation to changing conditions.


Asunto(s)
Biosíntesis de Proteínas , Ribosomas/metabolismo , Adaptación Fisiológica , Modelos Biológicos
10.
Eur J Immunol ; 44(9): 2536-49, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24965684

RESUMEN

The innate immune system detects infection and tissue injury through different families of pattern-recognition receptors (PRRs), such as Toll-like receptors. Most PRR-mediated responses initiate elaborate processes of signaling, transcription, translation, and secretion of effector mediators, which together require time to achieve. Therefore, PRR-mediated processes are not active in the early phases of infection. These considerations raise the question of how the host limits microbial replication and invasion during this critical period. Here, we examine the crucial defense mechanisms, such as antimicrobial peptides or extracellular traps, typically activated within minutes of the initial infection phase, which we term the "immediate protective response". Deficiencies in different components of the immediate protective response are often associated with severe and recurrent infectious diseases in humans, highlighting their physiologic importance.


Asunto(s)
Inmunidad Innata/fisiología , Infecciones/inmunología , Receptores Toll-Like/inmunología , Animales , Humanos
12.
Exp Parasitol ; 156: 68-78, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26071205

RESUMEN

Giardia lamblia is a leading protozoan cause of diarrheal disease worldwide. It colonizes the lumen and epithelial surface of the small intestine, but does not invade the mucosa. Acute infection causes only minimal mucosal inflammation. Effective immune defenses exist, yet their identity and mechanisms remain incompletely understood. Interleukin (IL)-17A has emerged as an important cytokine involved in inflammation and antimicrobial defense against bacterial pathogens at mucosal surfaces. In this study, we demonstrate that IL-17A has a crucial function in host defense against Giardia infection. Using murine infection models with G. muris and G. lamblia, we observed marked and selective induction of intestinal IL-17A with peak expression after 2 weeks. Th17 cells in the lamina propria and innate immune cells in the epithelial compartment of the small intestine were responsible for the IL-17A response. Experiments in gene-targeted mice revealed that the cytokine, and its cognate receptor IL-17RA, were required for eradication of the parasite. The actions of the cytokine were mediated by hematopoietic cells, and were required for the transport of IgA into the intestinal lumen, since IL-17A deficiency led to marked reduction of fecal IgA levels, as well as for increased intestinal expression of several other potential effectors, including ß-defensin 1 and resistin-like molecule ß. In contrast, intestinal hypermotility, another major antigiardial defense mechanism, was not impacted by IL-17A loss. Taken together, these findings demonstrate that IL-17A and IL-17 receptor signaling are essential for intestinal defense against the important lumen-dwelling intestinal parasite Giardia.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Giardia/inmunología , Giardiasis/inmunología , Inmunoglobulina A/biosíntesis , Interleucina-17/metabolismo , Animales , Anticuerpos Antiprotozoarios/inmunología , Linfocitos T CD4-Positivos/inmunología , Quimera , Giardia lamblia/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunoglobulina A/inmunología , Interleucina-17/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Intestino Delgado/inmunología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Transducción de Señal/inmunología , Organismos Libres de Patógenos Específicos , Células Th17/inmunología
13.
bioRxiv ; 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39229034

RESUMEN

Neurons are long-lived, terminally differentiated cells with limited regenerative capacity. Cellular stressors such as endoplasmic reticulum (ER) protein folding stress and membrane trafficking stress accumulate as neurons age and accompany age-dependent neurodegeneration. Current strategies to improve neuronal resilience are focused on using factors to reprogram neurons or targeting specific proteostasis pathways. We discovered a different approach. In an unbiased screen for modifiers of neuronal membrane trafficking defects, we unexpectedly identified a role for histone deacetylases (HDACs) in limiting cellular flexibility in choosing cellular pathways to respond to diverse types of stress. Genetic or pharmacological inactivation of HDACs resulted in improved neuronal health in response to ER protein folding stress and endosomal membrane trafficking stress in C. elegans and mammalian neurons. Surprisingly, HDAC inhibition enabled neurons to activate latent proteostasis pathways tailored to the nature of the individual stress, instead of generalized transcriptional upregulation. These findings shape our understanding of neuronal stress responses and suggest new therapeutic strategies to enhance neuronal resilience.

14.
J Biol Chem ; 287(6): 4033-40, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22167182

RESUMEN

ATG16L1 is an essential component of the autophagasome. The T300A allele of ATG16L1 is associated with the increased susceptibility to Crohn disease. In this study, we identified a novel function of ATG16L1, which suppresses signaling of the pro-inflammatory cytokine IL-1ß. Deletion of ATG16L1 in mouse embryonic fibroblasts significantly amplifies IL-1ß signal transduction cascades. This amplification is due to elevated p62 levels in ATG16L1-deficient cells. We found that ATG16L1 regulates p62 levels via both autolysosomal and proteasomal pathways. For proteasomal degradation, we found that Cullin-3 (Cul-3) is a E3 ubiquitin ligase of p62 and that ATG16L1 is essential for neddylation of Cul-3, a step required for Cul-3 activation. Taken together our data indicate that loss-of-function of ATG16L1 results in a hyper-responsiveness to the IL-1ß signaling because of the increased p62 level.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia/fisiología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteínas de Choque Térmico/metabolismo , Interleucina-1beta/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Células Cultivadas , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Embrión de Mamíferos/citología , Activación Enzimática/fisiología , Fibroblastos/citología , Eliminación de Gen , Proteínas de Choque Térmico/genética , Interleucina-1beta/genética , Ratones , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína Sequestosoma-1
15.
J Biol Chem ; 287(22): 18182-9, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22496368

RESUMEN

STAT3 was recently reported to suppress tumor invasion in Apc(min)(/+) mice. We investigated the mechanisms by which STAT3 inhibits intestinal epithelial tumors using Apc(min)(/+)/Stat3(IEC-KO) mice (intestinal epithelial cell (IEC)-specific deletion of STAT3 in the Apc(min)(/+) background) to determine the role of STAT3 in carcinogenesis in vivo as well as colorectal cancer cell lines in vitro. To inhibit invasion of IEC tumors, STAT3 functions as a molecular adaptor rather than a transcription factor. Accordingly, the tumors in Apc(min)(/+)/Stat3(IEC-KO) mice undergo adenoma-to-carcinoma transition and acquire an invasive phenotype. Similarly, STAT3 knockdown in a colorectal cell line enhances IEC invasion. We demonstrate that STAT3 down-regulates SNAI (Snail-1) expression levels and hence suppresses epithelial-mesenchymal transition of colorectal cancer cells. Mechanistically, STAT3 facilitates glycogen synthase kinase (GSK) 3ß-mediated degradation of SNAI by regulating phosphorylation of GSK3ß. Our data identified a new role for STAT3 in the adenoma-to-carcinoma sequence of intestinal tumors.


Asunto(s)
Adenoma/patología , Carcinoma/patología , Genes APC , Factor de Transcripción STAT3/fisiología , Factores de Transcripción/fisiología , Animales , Secuencia de Bases , Proliferación Celular , Cartilla de ADN , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Neoplasias Intestinales/patología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño , Factor de Transcripción STAT3/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética
16.
Gastroenterology ; 143(1): 145-54, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22475534

RESUMEN

BACKGROUND & AIMS: Foxp3(+) T-regulatory cells (Tregs) maintain intestinal homeostasis under conditions of continuous challenge with inflammatory microbes. However, plasticity of the Treg population under certain conditions has been reported; Foxp3(+) Tregs can be converted to Foxp3(-) CD4(+) T cells. METHODS: We used mice with a T cell-induced colitis model to study the regulatory role of type I interferons (IFNs) in adaptive immunity. We transferred CD4(+)CD45RB(hi) (RB(hi)) T cells, with or without CD4(+)CD45RB(lo) CD25(+) T cells, from wild-type or IFN-αßR(-/-) mice into Rag1(-/-) recipients. We analyzed induction of colitis by flow cytometry, confocal microscopy, and enzyme-linked immunosorbent assay and reverse-transcription polymerase chain reaction analyses. IFN-αßR(-/-)Rag(-/-) mice were given injections of recombinant IFN-α following transfer of IFN-αßR(-/-) RB(hi) T cells and CD4(+)Foxp3(+) cells from Foxp3-eGFP mice. RESULTS: Signaling by type I IFNs was required for maintenance of Foxp3 expression and the suppressive activity of Tregs in mice. Transfer of CD4(+)CD45RB(lo)CD25(+) Tregs from IFN-αßR(-/-) mice did not prevent T-cell induction of colitis in mice. Foxp3 expression by Tregs transferred from IFN-αßR(-/-) mice was significantly lower than that of Tregs from wild-type mice. Administration of recombinant IFN-α reduced T cell-mediated colitis by increasing the number of Foxp3(+) Tregs and their suppressive functions. CONCLUSIONS: Type I IFNs regulate intestinal homeostasis by maintaining Foxp3 expression on Tregs in colons of mice under inflammatory conditions.


Asunto(s)
Colitis/inmunología , Factores de Transcripción Forkhead/biosíntesis , Interferón Tipo I/inmunología , Linfocitos T Reguladores/inmunología , Inmunidad Adaptativa , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL
17.
Nat Cell Biol ; 8(12): 1327-36, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17128265

RESUMEN

The mechanisms by which commensal bacteria suppress inflammatory signalling in the gut are still unclear. Here, we present a cellular mechanism whereby the polarity of intestinal epithelial cells (IECs) has a major role in colonic homeostasis. TLR9 activation through apical and basolateral surface domains have distinct transcriptional responses, evident by NF-kappaB activation and cDNA microarray analysis. Whereas basolateral TLR9 signals IkappaBalpha degradation and activation of the NF-kappaB pathway, apical TLR9 stimulation invokes a unique response in which ubiquitinated IkappaB accumulates in the cytoplasm preventing NF-kappaB activation. Furthermore, apical TLR9 stimulation confers intracellular tolerance to subsequent TLR challenges. IECs in TLR9-deficient mice, when compared with wild-type and TLR2-deficient mice, display a lower NF-kappaB activation threshold and these mice are highly susceptible to experimental colitis. Our data provide a case for organ-specific innate immunity in which TLR expression in polarized IECs has uniquely evolved to maintain colonic homeostasis and regulate tolerance and inflammation.


Asunto(s)
Polaridad Celular , Colon/citología , Enterocitos/citología , Homeostasis , Transducción de Señal , Receptor Toll-Like 9/metabolismo , Animales , Células CACO-2 , Cloroquina/farmacología , Colon/efectos de los fármacos , Colon/patología , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Ligandos , Ratones , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 9/química
18.
Proc Natl Acad Sci U S A ; 107(23): 10638-43, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20479237

RESUMEN

Cholera toxin (CT) elicits a mucosal immune response in mice when used as a vaccine adjuvant. The mechanisms by which CT exerts its adjuvant effects are incompletely understood. We show that protection against inhalation anthrax by an irradiated spore vaccine depends on CT-mediated induction of IL-17-producing CD4 Th17 cells. Furthermore, IL-17 is involved in the induction of serum and mucosal antibody responses by CT. Th17 cells induced by CT have a unique cytokine profile compared with those induced by IL-6 and TGF-beta, and their induction by CT requires cAMP-dependent secretion of IL-1beta and beta-calcitonin gene-related peptide by dendritic cells. These findings demonstrate that Th17 cells mediate mucosal adjuvant effects of CT and identify previously unexplored pathways involved in Th17 induction that could be targeted for development of unique mucosal adjuvants.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra el Carbunco/inmunología , Toxina del Cólera/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Formación de Anticuerpos , Toxina del Cólera/farmacología , Inmunidad Mucosa , Inhalación , Interleucina-17/inmunología , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/inmunología
19.
Diabetes ; 72(9): 1235-1250, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37257047

RESUMEN

In obesity, CD11c+ innate immune cells are recruited to adipose tissue and create an inflammatory state that causes both insulin and catecholamine resistance. We found that ablation of Gnas, the gene that encodes Gαs, in CD11c expressing cells protects mice from obesity, glucose intolerance, and insulin resistance. Transplantation studies showed that the lean phenotype was conferred by bone marrow-derived cells and did not require adaptive immunity. Loss of cAMP signaling was associated with increased adipose tissue norepinephrine and cAMP signaling, and prevention of catecholamine resistance. The adipose tissue had reduced expression of catecholamine transport and degradation enzymes, suggesting that the elevated norepinephrine resulted from decreased catabolism. Collectively, our results identified an important role for cAMP signaling in CD11c+ innate immune cells in whole-body metabolism by controlling norepinephrine levels in white adipose tissue, modulating catecholamine-induced lipolysis and increasing thermogenesis, which, together, created a lean phenotype. ARTICLE HIGHLIGHTS: We undertook this study to understand how immune cells communicate with adipocytes, specifically, whether cAMP signaling in the immune cell and the adipocyte are connected. We identified a reciprocal interaction between CD11c+ innate immune cells and adipocytes in which high cAMP signaling in the immune cell compartment induces low cAMP signaling in adipocytes and vice versa. This interaction regulates lipolysis in adipocytes and inflammation in immune cells, resulting in either a lean, obesity-resistant, and insulin-sensitive phenotype, or an obese, insulin-resistant phenotype.


Asunto(s)
Dieta Alta en Grasa , Resistencia a la Insulina , Obesidad , Animales , Ratones , Tejido Adiposo Blanco/metabolismo , Catecolaminas/metabolismo , Dieta Alta en Grasa/efectos adversos , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Ratones Endogámicos C57BL , Norepinefrina/metabolismo , Obesidad/etiología , Obesidad/metabolismo
20.
J Exp Clin Cancer Res ; 42(1): 26, 2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36670473

RESUMEN

BACKGROUND: Individuals with certain chronic inflammatory lung diseases have a higher risk of developing lung cancer (LC). However, the underlying mechanisms remain largely unknown. Here, we hypothesized that chronic exposure to house dust mites (HDM), a common indoor aeroallergen associated with the development of asthma, accelerates LC development through the induction of chronic lung inflammation (CLI).  METHODS: The effects of HDM and heat-inactivated HDM (HI-HDM) extracts were evaluated in two preclinical mouse models of LC (a chemically-induced model using the carcinogen urethane and a genetically-driven model with oncogenic KrasG12D activation in lung epithelial cells) and on murine macrophages in vitro. Pharmacological blockade or genetic deletion of the Nod-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome, caspase-1, interleukin-1ß (IL-1ß), and C-C motif chemokine ligand 2 (CCL2) or treatment with an inhaled corticosteroid (ICS) was used to uncover the pro-tumorigenic effect of HDM.  RESULTS: Chronic intranasal (i.n) instillation of HDM accelerated LC development in the two mouse models. Mechanistically, HDM caused a particular subtype of CLI, in which the NLRP3/IL-1ß signaling pathway is chronically activated in macrophages, and made the lung microenvironment conducive to tumor development. The tumor-promoting effect of HDM was significantly decreased by heat treatment of the HDM extract and was inhibited by NLRP3, IL-1ß, and CCL2 neutralization, or ICS treatment. CONCLUSIONS: Collectively, these data indicate that long-term exposure to HDM can accelerate lung tumorigenesis in susceptible hosts (e.g., mice and potentially humans exposed to lung carcinogens or genetically predisposed to develop LC).


Asunto(s)
Asma , Neoplasias Pulmonares , Humanos , Animales , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Pyroglyphidae , Pulmón/patología , Asma/metabolismo , Asma/patología , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/metabolismo , Modelos Animales de Enfermedad , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA