Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 115(14): E3173-E3181, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29559533

RESUMEN

Wnts and R-spondins (RSPOs) support intestinal homeostasis by regulating crypt cell proliferation and differentiation. Ex vivo, Wnts secreted by Paneth cells in organoids can regulate the proliferation and differentiation of Lgr5-expressing intestinal stem cells. However, in vivo, Paneth cell and indeed all epithelial Wnt production is completely dispensable, and the cellular source of Wnts and RSPOs that maintain the intestinal stem-cell niche is not known. Here we investigated both the source and the functional role of stromal Wnts and RSPO3 in regulation of intestinal homeostasis. RSPO3 is highly expressed in pericryptal myofibroblasts in the lamina propria and is several orders of magnitude more potent than RSPO1 in stimulating both Wnt/ß-catenin signaling and organoid growth. Stromal Rspo3 ablation ex vivo resulted in markedly decreased organoid growth that was rescued by exogenous RSPO3 protein. Pdgf receptor alpha (PdgfRα) is known to be expressed in pericryptal myofibroblasts. We therefore evaluated if PdgfRα identified the key stromal niche cells. In vivo, Porcn excision in PdgfRα+ cells blocked intestinal crypt formation, demonstrating that Wnt production in the stroma is both necessary and sufficient to support the intestinal stem-cell niche. Mice with Rspo3 excision in the PdgfRα+ cells had decreased intestinal crypt Wnt/ß-catenin signaling and Paneth cell differentiation and were hypersensitive when stressed with dextran sodium sulfate. The data support a model of the intestinal stem-cell niche regulated by both Wnts and RSPO3 supplied predominantly by stromal pericryptal myofibroblasts marked by PdgfRα.


Asunto(s)
Células Epiteliales/citología , Intestinos/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Nicho de Células Madre/fisiología , Células Madre/citología , Células del Estroma/citología , Trombospondinas/metabolismo , Proteína Wnt1/metabolismo , Aciltransferasas/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Células Epiteliales/metabolismo , Homeostasis , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides/citología , Organoides/metabolismo , Células Madre/metabolismo , Células del Estroma/metabolismo , Trombospondinas/genética , Proteína Wnt1/genética
2.
Differentiation ; 108: 8-16, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30683451

RESUMEN

Intestinal homeostasis is dependent on the continuous production of differentiated epithelial cells from a sustainable and resilient stem cell compartment. Wnt/ß-catenin signaling plays a central role in this process, cooperating with R-spondins, growth factors and regulators of the TGF-ß/BMP pathway to generate a specialized tissue microenvironment that regulates the intestinal stem cell niche. Recent studies revealed that many of these factors are produced in a paracrine manner by specialized cell populations that reside in the subepithelial stroma. These stromal signal-producing cells, variously called telocytes and myofibroblasts, can be identified by expression of specific genes including PdgfRa, Gli1 and FoxL1. In this review we discuss how the intestinal stem cell niche is established during development and then sustained during adult intestinal homeostasis by these stromal cell populations. The signaling stroma cells regulate intestinal stem cell development into different epithelial lineages and play an important role in the response to environmental stresses.


Asunto(s)
Autorrenovación de las Células , Mucosa Intestinal/metabolismo , Nicho de Células Madre , Animales , Humanos , Mucosa Intestinal/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Vía de Señalización Wnt
3.
Development ; 141(11): 2206-15, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24821987

RESUMEN

Wnt/ß-catenin signaling supports intestinal homeostasis by regulating proliferation in the crypt. Multiple Wnts are expressed in Paneth cells as well as other intestinal epithelial and stromal cells. Ex vivo, Wnts secreted by Paneth cells can support intestinal stem cells when Wnt signaling is enhanced with supplemental R-Spondin 1 (RSPO1). However, in vivo, the source of Wnts in the stem cell niche is less clear. Genetic ablation of Porcn, an endoplasmic reticulum resident O-acyltransferase that is essential for the secretion and activity of all vertebrate Wnts, confirmed the role of intestinal epithelial Wnts in ex vivo culture. Unexpectedly, mice lacking epithelial Wnt activity (Porcn(Del)/Villin-Cre mice) had normal intestinal proliferation and differentiation, as well as successful regeneration after radiation injury, indicating that epithelial Wnts are dispensable for these processes. Consistent with a key role for stroma in the crypt niche, intestinal stromal cells endogenously expressing Wnts and Rspo3 support the growth of Porcn(Del) organoids ex vivo without RSPO1 supplementation. Conversely, increasing pharmacologic PORCN inhibition, affecting both stroma and epithelium, reduced Lgr5 intestinal stem cells, inhibited recovery from radiation injury, and at the highest dose fully blocked intestinal proliferation. We conclude that epithelial Wnts are dispensable and that stromal production of Wnts can fully support normal murine intestinal homeostasis.


Asunto(s)
Epitelio/metabolismo , Proteínas de la Membrana/genética , Nicho de Células Madre , Células del Estroma/citología , Proteínas Wnt/metabolismo , Aciltransferasas , Animales , Apoptosis , Proliferación Celular , Retículo Endoplásmico/metabolismo , Células Epiteliales/citología , Fibroblastos/metabolismo , Eliminación de Gen , Células HEK293 , Homeostasis , Humanos , Intestinos/citología , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Células de Paneth/citología , Transducción de Señal , Células Madre/citología , Trombospondinas/metabolismo
4.
Carcinogenesis ; 33(6): 1231-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22461519

RESUMEN

Chronic inflammation is increasingly recognized as a major contributor of human colorectal cancer (CRC). While gut microbiota can trigger inflammation in the intestinal tract, the precise signaling pathways through which host cells respond to inflammatory bacterial stimulation are unclear. Here, we show that gut microbiota enhances intestinal tumor load in the APC(Min/+) mouse model of CRC. Furthermore, systemic anemia occurs coincident with rapid tumor growth, suggesting a role for intestinal barrier damage and erythropoiesis-stimulating mitogens. Short-term stimulation assays of murine colonic tumor cells reveal that lipopolysaccharide, a microbial cell wall component, can accelerate cell growth via a c-Jun/JNK activation pathway. Colonic tumors are also infiltrated by CD11b+ myeloid cells expressing high levels of phospho-STAT3 (p-Tyr705). Our results implicate the role of gut microbiota, through triggering the c-Jun/JNK and STAT3 signaling pathways in combination with anemia, in the acceleration of tumor growth in APC(Min/+) mice.


Asunto(s)
Neoplasias Colorrectales/microbiología , Intestinos/microbiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metagenoma/fisiología , Factor de Transcripción STAT3/metabolismo , Anemia , Animales , Antígeno CD11b/biosíntesis , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Eritropoyetina/farmacología , Genes APC , Inflamación/microbiología , Mucosa Intestinal/metabolismo , Intestinos/patología , Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/metabolismo , Fosforilación , Transducción de Señal , Carga Tumoral
5.
Proc Natl Acad Sci U S A ; 105(6): 1943-8, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18234854

RESUMEN

The postembryonic development of the gastrointestinal tract is subject to regulation by the colonizing microbiota. This maturation process requires the commensal bacteria to cross-talk with host cells by way of recognizing receptors and inducing signaling pathways to activate transcription factors such as the nuclear receptors. Here, we show that in colonic cell lines and in primary colonic cells, Enterococcus faecalis isolated from newborn babies possess the ability to regulate peroxisome proliferator-activated receptor-gamma1 (PPARgamma1) activity through phosphorylation. This results in elevated DNA binding and transcriptional activation of downstream target genes, including IL-10, a cytokine known to modulate innate immune function. Furthermore, phosphorylation appears tightly regulated as phospho-PPARgamma1 becomes an immediate substrate for degradation possibly to curtail any extended transactivation. The involvement of PPARgamma1 in a myriad of physiological processes further confirms that microflora-driven regulation might be important for a number of homeostatic strategies in the gut.


Asunto(s)
Colon/metabolismo , Enterococcus faecalis/fisiología , Interleucina-10/metabolismo , PPAR gamma/metabolismo , Colon/citología , Colon/microbiología , ADN/metabolismo , Expresión Génica , Células HT29 , Humanos , Recién Nacido , Interleucina-10/genética , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ligandos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Perilipina-2 , Fosforilación , Unión Proteica
6.
Nat Commun ; 12(1): 2058, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824332

RESUMEN

Wnt signaling regulates cell proliferation and cell differentiation as well as migration and polarity during development. However, it is still unclear how the Wnt ligand distribution is precisely controlled to fulfil these functions. Here, we show that the planar cell polarity protein Vangl2 regulates the distribution of Wnt by cytonemes. In zebrafish epiblast cells, mouse intestinal telocytes and human gastric cancer cells, Vangl2 activation generates extremely long cytonemes, which branch and deliver Wnt protein to multiple cells. The Vangl2-activated cytonemes increase Wnt/ß-catenin signaling in the surrounding cells. Concordantly, Vangl2 inhibition causes fewer and shorter cytonemes to be formed and reduces paracrine Wnt/ß-catenin signaling. A mathematical model simulating these Vangl2 functions on cytonemes in zebrafish gastrulation predicts a shift of the signaling gradient, altered tissue patterning, and a loss of tissue domain sharpness. We confirmed these predictions during anteroposterior patterning in the zebrafish neural plate. In summary, we demonstrate that Vangl2 is fundamental to paracrine Wnt/ß-catenin signaling by controlling cytoneme behaviour.


Asunto(s)
Proteínas de la Membrana/metabolismo , Seudópodos/metabolismo , Vía de Señalización Wnt , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo , Embrión no Mamífero/metabolismo , Activación Enzimática , Fibroblastos/metabolismo , Gastrulación , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones Endogámicos C57BL , Placa Neural/embriología , Placa Neural/metabolismo , Neurogénesis , Comunicación Paracrina , Análisis de Sistemas , Telocitos/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo
7.
J Clin Invest ; 128(9): 3806-3812, 2018 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-30059017

RESUMEN

Intestinal homeostasis depends on a slowly proliferating stem cell compartment in crypt cells, followed by rapid proliferation of committed progenitor cells in the transit amplifying (TA) compartment. The balance between proliferation and differentiation in intestinal stem cells (ISCs) is regulated by Wnt/ß-catenin signaling, although the mechanism remains unclear. We previously targeted PORCN, an enzyme essential for all Wnt secretion, and demonstrated that stromal production of Wnts was required for intestinal homeostasis. Here, a PORCN inhibitor was used to acutely suppress Wnt signaling. Unexpectedly, the treatment induced an initial burst of proliferation in the stem cell compartment of the small intestine, due to conversion of ISCs into TA cells with a loss of intrinsic ISC self-renewal. This process involved MAPK pathway activation, as the proliferating cells in the base of the intestinal crypt contained phosphorylated ERK1/2, and a MEK inhibitor attenuated the proliferation of ISCs and their differentiation into TA cells. These findings suggest a role for Wnt signaling in suppressing the MAPK pathway at the crypt base to maintain a pool of ISCs. The interaction between Wnt and MAPK pathways in vivo has potential therapeutic applications in cancer and regenerative medicine.


Asunto(s)
Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Sistema de Señalización de MAP Quinasas , Células Madre/citología , Células Madre/metabolismo , Vía de Señalización Wnt , Aciltransferasas/antagonistas & inhibidores , Animales , Bencenoacetamidas/farmacología , Diferenciación Celular , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Piridinas/farmacología , Células Madre/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
8.
Elife ; 72018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30060804

RESUMEN

Signaling filopodia, termed cytonemes, are dynamic actin-based membrane structures that regulate the exchange of signaling molecules and their receptors within tissues. However, how cytoneme formation is regulated remains unclear. Here, we show that Wnt/planar cell polarity (PCP) autocrine signaling controls the emergence of cytonemes, and that cytonemes subsequently control paracrine Wnt/ß-catenin signal activation. Upon binding of the Wnt family member Wnt8a, the receptor tyrosine kinase Ror2 becomes activated. Ror2/PCP signaling leads to the induction of cytonemes, which mediate the transport of Wnt8a to neighboring cells. In the Wnt-receiving cells, Wnt8a on cytonemes triggers Wnt/ß-catenin-dependent gene transcription and proliferation. We show that cytoneme-based Wnt transport operates in diverse processes, including zebrafish development, murine intestinal crypt and human cancer organoids, demonstrating that Wnt transport by cytonemes and its control via the Ror2 pathway is highly conserved in vertebrates.


Asunto(s)
Proteínas del Citoesqueleto/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Proteínas Wnt/genética , Proteínas de Pez Cebra/genética , beta Catenina/genética , Animales , Comunicación Autocrina/genética , Polaridad Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Comunicación Paracrina/genética , Seudópodos/genética , Seudópodos/metabolismo , Vía de Señalización Wnt/genética , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
9.
J Leukoc Biol ; 74(1): 126-34, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12832451

RESUMEN

Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1, CD66a) is a member of the immunoglobulin (Ig) superfamily, previously characterized as an adhesion and signaling molecule in epithelial, endothelial, and hematopoietic cells. Here, we show that the CEACAM1 isoform expression pattern is different in nonactivated and activated primary mouse B lymphocytes and that CEACAM1 influences B cell receptor complex-mediated activation. A CEACAM1-specific monoclonal antibody strongly triggered proliferation of mouse B cells when combined with surface IgM cross-linking. However, anti-CEACAM1 was not mitogenic when added alone. The proliferation was more pronounced and lasted longer as compared with other activators of B cells, such as anti-IgM in the presence of interleukin-4 or lipopolysaccharide. A similar, costimulatory effect was exerted by CEACAM1-expressing fibroblasts, indicating that homophilic CEACAM1-CEACAM1 cell-mediated binding is the physiological stimulus for CEACAM1-triggered B cell signaling. The anti-CEACAM1/anti-IgM-activated cells aggregated in a lymphocyte function-associated antigen-1-dependent manner. Furthermore, cells that were activated by anti-CEACAM1/anti-IgM secreted Ig but did not go through Ig class-switching. Anti-CEACAM1 induced phosphorylation of c-Jun N-terminal kinase (stress-activated protein kinase) but did not activate the extracellular signal-regulated kinase or p38 mitogen-activated protein kinases.


Asunto(s)
Glicoproteínas/fisiología , Activación de Linfocitos , Receptores de Antígenos de Linfocitos B/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD , Linfocitos B/metabolismo , Antígeno Carcinoembrionario , Moléculas de Adhesión Celular , Fibroblastos , Glicoproteínas/inmunología , Inmunoglobulina M , Proteínas Quinasas JNK Activadas por Mitógenos , Antígeno-1 Asociado a Función de Linfocito , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Transducción de Señal
10.
Mol Cancer Ther ; 9(3): 642-52, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20197387

RESUMEN

Although clinical responses in liquid tumors and certain lymphomas have been reported, the clinical efficacy of histone deacetylase inhibitors in solid tumors has been limited. This may be in part due to the poor pharmacokinetic of these drugs, resulting in inadequate tumor concentrations of the drug. SB939 is a new hydroxamic acid based histone deacetylase inhibitor with improved physicochemical, pharmaceutical, and pharmacokinetic properties. In vitro, SB939 inhibits class I, II, and IV HDACs, with no effects on other zinc binding enzymes, and shows significant antiproliferative activity against a wide variety of tumor cell lines. It has very favorable pharmacokinetic properties after oral dosing in mice, with >4-fold increased bioavailability and 3.3-fold increased half-life over suberoylanilide hydroxamic acid (SAHA). In contrast to SAHA, SB939 accumulates in tumor tissue and induces a sustained inhibition of histone acetylation in tumor tissue. These excellent pharmacokinetic properties translated into a dose-dependent antitumor efficacy in a xenograft model of human colorectal cancer (HCT-116), with a tumor growth inhibition of 94% versus 48% for SAHA (both at maximum tolerated dose), and was also effective when given in different intermittent schedules. Furthermore, in APC(min) mice, a genetic mouse model of early-stage colon cancer, SB939 inhibited adenoma formation, hemocult scores, and increased hematocrit values more effectively than 5-fluorouracil. Emerging clinical data from phase I trials in cancer patients indicate that the pharmacokinetic and pharmacologic advantages of SB939 are translated to the clinic. The efficacy of SB939 reported here in two very different models of colorectal cancer warrants further investigation in patients.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/farmacocinética , Ácidos Hidroxámicos/administración & dosificación , Ácidos Hidroxámicos/farmacocinética , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Disponibilidad Biológica , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Células HCT116 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Resultado del Tratamiento , Células Tumorales Cultivadas , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Curr Opin Gastroenterol ; 22(4): 349-53, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16760748

RESUMEN

PURPOSE OF REVIEW: The open ecosystem of the alimentary tract, harboring approximately 1 kg of bacteria, exhibits a rapid, but tightly controlled turnover. Impaired nuclear receptor function can give rise to perturbation in the gut, leading to inflammation and possibly neoplasia. Intriguingly, bacteria-dependent signaling pathways can modulate, and in turn be modulated by, a subset of nuclear receptors. This review attempts to highlight how microbes and nuclear receptors could jointly regulate gut homeostasis. RECENT FINDINGS: Commensal bacteria can utilize peroxisome proliferator activated receptor-gamma-dependent nuclear export of RelA as a novel mechanism to attenuate inflammatory signals triggered by a pathogen. Other nuclear receptors, such as liver X receptor, vitamin D receptor and farnesoid X receptor were also recently shown to interact with bacteria-induced mammalian inflammatory pathways. Although details of this interplay are still being unraveled, a role for these and other nuclear receptors in gastrointestinal inflammation and possibly neoplasia is beyond dispute. SUMMARY: The commensal microflora is being accorded due importance in regulating homeostasis of the gastrointestinal tract. Recent data suggest that the molecular messengers used by these bacteria include nuclear receptors. Exploiting mechanisms of nuclear receptor activity as drug targets, together with a detailed knowledge of the microbiota, could improve our understanding of gut-related ailments, and aid in mitigating their symptoms.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Homeostasis/fisiología , Enfermedades Inflamatorias del Intestino/metabolismo , Animales , Bacterias/crecimiento & desarrollo , Bacterias/metabolismo , Progresión de la Enfermedad , Tracto Gastrointestinal/microbiología , Humanos , Enfermedades Inflamatorias del Intestino/microbiología , Receptores Citoplasmáticos y Nucleares/metabolismo
12.
Proc Natl Acad Sci U S A ; 102(5): 1460-5, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15665104

RESUMEN

Control of colon cell fate in adenocarcinomas is disrupted, in part, due to aberrant Wnt/beta-catenin signaling. The nuclear receptor peroxisome proliferator-activated receptor-gamma (PPARgamma) has been implicated in the development of colon cancers. In the adenomatous polyposis coli multiple intestinal neoplasia (APCMin) mouse cancer model, PPARgamma expression in the colonic mucosa is markedly altered. In addition, PPARgamma protein levels are elevated, possibly through sequestration by activated beta-catenin in colon cancer cell lines. Induction of the Wnt/beta-catenin pathway by LiCl also elevated PPARgamma levels and induced PPARgamma-dependent reporter and endogenous target genes. Mechanistically, PPARgamma, through interactions with beta-catenin and T cell transcription factor (Tcf)-4, may be a determinant of cell fate and is likely a target of the Wnt pathway in cancer cells.


Asunto(s)
Cadherinas/fisiología , Proteínas del Citoesqueleto/fisiología , PPAR gamma/fisiología , Transactivadores/fisiología , Poliposis Adenomatosa del Colon/genética , Animales , Secuencia de Bases , Línea Celular , Línea Celular Tumoral , Núcleo Celular/fisiología , Neoplasias del Colon , Cartilla de ADN , Genes APC , Genes Reporteros , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Mucosa Intestinal/fisiología , Riñón , Ratones , PPAR gamma/genética , ARN Mensajero/genética , Ratas , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Proteínas Wnt , beta Catenina
13.
Immunology ; 109(3): 384-91, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12807484

RESUMEN

B cells and dendritic cells, lacking functional Wiskott-Aldrich syndrome protein (WASP), have aberrant formation of membrane protrusions. We hypothesized that protrusions may play a role in antigen presentation, and consequently, that impaired antigen presentation may be an underlying factor of the immune deficiency in patients with Wiskott-Aldrich syndrome. In this paper, we investigated the antigen presentation capacity of B cells and dendritic cells from WASP knockout mice, using soluble and particulate antigen, to CD4+ T cells from T-cell receptor transgenic DO11.10 mice. As antigen we used soluble ovalbumin (OVA), a peptide thereof (amino acids 323-339) or bacteria expressing OVA. We found that WASP-deficient B cells and dendritic cells efficiently processed and presented soluble OVA protein as well as its peptide in vitro, inducing proliferation and cytokine production from CD4+ T cells. Antigen presentation of soluble protein was efficient also in vivo, because immunization of WASP-deficient mice with OVA elicited proliferation of transferred, fluorescent-labelled, CD4+ T cells. Although we could detect uptake of bacteria in dendritic cells, processing and presentation of bacterial-expressed OVA was impaired in WASP-deficient dendritic cells. In conclusion, our data suggest that WASP is not needed for processing and presentation of soluble antigen, but that efficient presentation of particulate antigen require WASP.


Asunto(s)
Presentación de Antígeno/inmunología , Proteínas/inmunología , Animales , Antígenos Bacterianos/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Citocinas/biosíntesis , Células Dendríticas/inmunología , Femenino , Inmunidad Celular , Masculino , Ratones , Ratones Transgénicos , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Proteínas/genética , Solubilidad , Síndrome de Wiskott-Aldrich/inmunología , Proteína del Síndrome de Wiskott-Aldrich
14.
Int Immunol ; 16(2): 353-64, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14734621

RESUMEN

Interactive contact between B lymphocytes and T cells is necessary for their expansion during an immune response. It has been shown that B lymphocytes receive signals from T cells, such as IL-4 and cross-linking of CD40, which are crucial for their differentiation. We previously found that these factors induce formation of microvilli on B cells and that this was correlated with increased homotypic adhesion of B lymphocytes. In this study we have investigated if IL-4 induce segregation of proteins to microvilli and lipid rafts. Using immuno-electron microscopy we analyzed cell-surface distribution of molecules involved in B-T cell co-activation. Recruitment to detergent-resistant membrane fractions was analyzed using sucrose gradient centrifugation. We found that microvilli were enriched in ICAM-1 and MHC class II molecules. In contrast, LFA-1 and CD40 were more abundant on the smooth cell surfaces, while B7-2 (CD86) was randomly distributed. We also discovered that depletion of cholesterol, using beta-methyl-cyclodextrin, lowered the number of microvilli, indicating that intact lipid rafts are required for their expression. Moreover, activation of B lymphocytes by lipopolysaccharide (LPS) induced increased expression of GM(1), a marker for lipid rafts. However, although both surface and total levels of GM(1) were similar in B lymphocytes stimulated with either LPS or LPS plus IL-4, GM(1) was mainly expressed on microvilli in LPS plus IL-4-stimulated cells. Taken together, our results indicate that microvilli represent distinct inducible membrane domains that can regulate direct cell-cell interactions via grouping and three-dimensional presentation of cell-surface receptors.


Asunto(s)
Linfocitos B/ultraestructura , Genes MHC Clase II/fisiología , Molécula 1 de Adhesión Intercelular/metabolismo , Microdominios de Membrana/ultraestructura , Microvellosidades/ultraestructura , Animales , Antígenos CD/metabolismo , Linfocitos B/inmunología , Antígeno B7-2 , Antígenos CD40/metabolismo , Comunicación Celular/efectos de los fármacos , Comunicación Celular/fisiología , Toxina del Cólera/farmacología , Ciclodextrinas/farmacología , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Glicoproteínas de Membrana/metabolismo , Microdominios de Membrana/metabolismo , Ratones , Microvellosidades/inmunología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA