Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Leukoc Biol ; 112(2): 257-271, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34826345

RESUMEN

Macrophages use an array of innate immune sensors to detect intracellular pathogens and to tailor effective antimicrobial responses. In addition, extrinsic activation with the cytokine IFN-γ is often required as well to tip the scales of the host-pathogen balance toward pathogen restriction. However, little is known about how host-pathogen sensing impacts the antimicrobial IFN-γ-activated state. It was observed that in the absence of IRF3, a key downstream component of pathogen sensing pathways, IFN-γ-primed macrophages more efficiently restricted the intracellular bacterium Legionella pneumophila and the intracellular protozoan parasite Trypanosoma cruzi. This effect did not require IFNAR, the receptor for Type I IFNs known to be induced by IRF3, nor the sensing adaptors MyD88/TRIF, MAVS, or STING. This effect also did not involve differential activation of STAT1, the major signaling protein downstream of both Type 1 and Type 2 IFN receptors. IRF3-deficient macrophages displayed a significantly altered IFN-γ-induced gene expression program, with up-regulation of microbial restriction factors such as Nos2. Finally, we found that IFN-γ-primed but not unprimed macrophages largely excluded the activated form of IRF3 from the nucleus following bacterial infection. These data are consistent with a relationship of mutual inhibition between IRF3 and IFN-γ-activated programs, possibly as a component of a partially reversible mechanism for modulating the activity of potent innate immune effectors (such as Nos2) in the context of intracellular infection.


Asunto(s)
Factor 3 Regulador del Interferón , Interferón gamma , Legionella pneumophila , Macrófagos , Trypanosoma cruzi , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón gamma/metabolismo , Legionella pneumophila/patogenicidad , Macrófagos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Trypanosoma cruzi/patogenicidad
2.
Immunohorizons ; 5(10): 818-829, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34667099

RESUMEN

In this study, we report that the TLR4 ligand, LPS, and TLR3 ligand polyinosinic:polycytidylic acid failed to activate IRF3 or STAT1 in bone marrow-derived macrophages (BMMs) isolated from two independently generated lines of Rosa26-integrated Cas9-expressing C57BL/6J (B6) mice. RNA-sequencing analysis reveals that hundreds to thousands of genes including IFN-stimulated genes were differentially expressed in BMMs from these Cas9 strains compared with B6 upon LPS stimulation. Furthermore, the NF-κB signaling axis and TRIF-mediated necroptosis were also strongly reduced in response to LPS and polyinosinic:polycytidylic acid. In contrast, there were no defects in the responses of BMMs to ligands of the RIG-I, STING, TLR2, TLR9, and IFN receptors. Defects in TLR3 and TLR4 signaling were observed in mice with the B6 but not 129 background, and when Cas9 was integrated at the Rosa26 but not H11 locus. However, integration at the Rosa26 site, CAG promoter-driven Cas9 or eGFP were not individually sufficient to cause the defect. Taken together, the results of this study suggest a putative TRIF-mediated defect in TLR-3/4 signaling in BMMs from commercially available and widely used B6-Cas9-expressing mice.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Macrófagos/inmunología , Animales , Proteína 9 Asociada a CRISPR/genética , Células Cultivadas , Femenino , Lipopolisacáridos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Necroptosis/inmunología , Poli I-C/inmunología , Cultivo Primario de Células , ARN no Traducido/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo
3.
Nature ; 551(7680): 333-339, 2017 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-29144463

RESUMEN

Intestinal epithelial cells absorb nutrients, respond to microbes, function as a barrier and help to coordinate immune responses. Here we report profiling of 53,193 individual epithelial cells from the small intestine and organoids of mice, which enabled the identification and characterization of previously unknown subtypes of intestinal epithelial cell and their gene signatures. We found unexpected diversity in hormone-secreting enteroendocrine cells and constructed the taxonomy of newly identified subtypes, and distinguished between two subtypes of tuft cell, one of which expresses the epithelial cytokine Tslp and the pan-immune marker CD45, which was not previously associated with non-haematopoietic cells. We also characterized the ways in which cell-intrinsic states and the proportions of different cell types respond to bacterial and helminth infections: Salmonella infection caused an increase in the abundance of Paneth cells and enterocytes, and broad activation of an antimicrobial program; Heligmosomoides polygyrus caused an increase in the abundance of goblet and tuft cells. Our survey highlights previously unidentified markers and programs, associates sensory molecules with cell types, and uncovers principles of gut homeostasis and response to pathogens.


Asunto(s)
Células Epiteliales/citología , Epitelio/metabolismo , Intestino Delgado/citología , Análisis de la Célula Individual , Animales , Diferenciación Celular , Citocinas/metabolismo , Enterocitos/metabolismo , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Homeostasis , Antígenos Comunes de Leucocito/metabolismo , Masculino , Ratones , Organoides/citología , Organoides/metabolismo , Células de Paneth/metabolismo , Transcripción Genética , Linfopoyetina del Estroma Tímico
4.
Cell ; 162(3): 675-86, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26189680

RESUMEN

Finding the components of cellular circuits and determining their functions systematically remains a major challenge in mammalian cells. Here, we introduced genome-wide pooled CRISPR-Cas9 libraries into dendritic cells (DCs) to identify genes that control the induction of tumor necrosis factor (Tnf) by bacterial lipopolysaccharide (LPS), a key process in the host response to pathogens, mediated by the Tlr4 pathway. We found many of the known regulators of Tlr4 signaling, as well as dozens of previously unknown candidates that we validated. By measuring protein markers and mRNA profiles in DCs that are deficient in known or candidate genes, we classified the genes into three functional modules with distinct effects on the canonical responses to LPS and highlighted functions for the PAF complex and oligosaccharyltransferase (OST) complex. Our findings uncover new facets of innate immune circuits in primary cells and provide a genetic approach for dissection of mammalian cell circuits.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas Genéticas , Inmunidad Innata , Animales , Células de la Médula Ósea/inmunología , Diferenciación Celular , Supervivencia Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Técnicas de Inactivación de Genes , Redes Reguladoras de Genes , Hexosiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/inmunología
5.
Science ; 347(6226): 1259038, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25745177

RESUMEN

Protein expression is regulated by the production and degradation of messenger RNAs (mRNAs) and proteins, but their specific relationships remain unknown. We combine measurements of protein production and degradation and mRNA dynamics so as to build a quantitative genomic model of the differential regulation of gene expression in lipopolysaccharide-stimulated mouse dendritic cells. Changes in mRNA abundance play a dominant role in determining most dynamic fold changes in protein levels. Conversely, the preexisting proteome of proteins performing basic cellular functions is remodeled primarily through changes in protein production or degradation, accounting for more than half of the absolute change in protein molecules in the cell. Thus, the proteome is regulated by transcriptional induction for newly activated cellular functions and by protein life-cycle changes for remodeling of preexisting functions.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Interacciones Huésped-Patógeno/inmunología , Simulación de Dinámica Molecular , Biosíntesis de Proteínas , Proteolisis , Aminoácidos/química , Aminoácidos/metabolismo , Animales , Técnicas de Cultivo de Célula , Marcaje Isotópico/métodos , Lipopolisacáridos/inmunología , Ratones , Proteínas Mitocondriales/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Análisis de Secuencia de ARN
6.
J Immunol ; 192(10): 4655-65, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24729611

RESUMEN

In murine schistosomiasis, immunopathology and cytokine production in response to parasite eggs are uneven and strain dependent. CBA/J (CBA) mice develop severe hepatic granulomatous inflammation associated with prominent Th17 cell responses driven by dendritic cell (DC)-derived IL-1ß and IL-23. Such Th17 cells fail to develop in low-pathology C57BL/6 (BL/6) mice, and the reasons for these strain-specific differences in APC reactivity to eggs remain unclear. We show by gene profiling that CBA DCs display an 18-fold higher expression of the C-type lectin receptor CD209a, a murine homolog of human DC-specific ICAM-3-grabbing nonintegrin, compared with BL/6 DCs. Higher CD209a expression was observed in CBA splenic and granuloma APC subpopulations, but only DCs induced Th17 cell differentiation in response to schistosome eggs. Gene silencing in CBA DCs and overexpression in BL/6 DCs demonstrated that CD209a is essential for egg-elicited IL-1ß and IL-23 production and subsequent Th17 cell development, which is associated with SRC, RAF-1, and ERK1/2 activation. These findings reveal a novel mechanism controlling the development of Th17 cell-mediated severe immunopathology in helminthic disease.


Asunto(s)
Moléculas de Adhesión Celular/inmunología , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Lectinas Tipo C/inmunología , Receptores de Superficie Celular/inmunología , Schistosoma/inmunología , Esquistosomiasis/inmunología , Células Th17/inmunología , Animales , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Línea Celular , Células Dendríticas/metabolismo , Células Dendríticas/patología , Activación Enzimática/genética , Activación Enzimática/inmunología , Femenino , Regulación de la Expresión Génica/genética , Silenciador del Gen/inmunología , Granuloma/genética , Granuloma/inmunología , Granuloma/patología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Interleucina-23/genética , Interleucina-23/inmunología , Interleucina-23/metabolismo , Lectinas Tipo C/biosíntesis , Lectinas Tipo C/genética , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Ratones , Ratones Endogámicos CBA , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/inmunología , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Proto-Oncogénicas pp60(c-src)/inmunología , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Schistosoma/genética , Schistosoma/metabolismo , Esquistosomiasis/genética , Esquistosomiasis/metabolismo , Esquistosomiasis/patología , Bazo/inmunología , Bazo/metabolismo , Bazo/patología , Células Th17/metabolismo , Células Th17/patología
7.
Nat Commun ; 4: 2672, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24157732

RESUMEN

The recent development of a semiconductor-based, non-optical DNA sequencing technology promises scalable, low-cost and rapid sequence data production. The technology has previously been applied mainly to genomic sequencing and targeted re-sequencing. Here we demonstrate the utility of Ion Torrent semiconductor-based sequencing for sensitive, efficient and rapid chromatin immunoprecipitation followed by sequencing (ChIP-seq) through the application of sample preparation methods that are optimized for ChIP-seq on the Ion Torrent platform. We leverage this method for epigenetic profiling of tumour tissues.


Asunto(s)
Genoma Humano , Histonas/metabolismo , Melanoma/química , Procesamiento Proteico-Postraduccional , Análisis de Secuencia de ADN/instrumentación , Neoplasias Cutáneas/química , Animales , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Células Dendríticas/citología , Células Dendríticas/metabolismo , Epigénesis Genética , Femenino , Histonas/genética , Humanos , Melanoma/genética , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Semiconductores , Análisis de Secuencia de ADN/métodos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo
8.
Nature ; 498(7453): 236-40, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23685454

RESUMEN

Recent molecular studies have shown that, even when derived from a seemingly homogenous population, individual cells can exhibit substantial differences in gene expression, protein levels and phenotypic output, with important functional consequences. Existing studies of cellular heterogeneity, however, have typically measured only a few pre-selected RNAs or proteins simultaneously, because genomic profiling methods could not be applied to single cells until very recently. Here we use single-cell RNA sequencing to investigate heterogeneity in the response of mouse bone-marrow-derived dendritic cells (BMDCs) to lipopolysaccharide. We find extensive, and previously unobserved, bimodal variation in messenger RNA abundance and splicing patterns, which we validate by RNA-fluorescence in situ hybridization for select transcripts. In particular, hundreds of key immune genes are bimodally expressed across cells, surprisingly even for genes that are very highly expressed at the population average. Moreover, splicing patterns demonstrate previously unobserved levels of heterogeneity between cells. Some of the observed bimodality can be attributed to closely related, yet distinct, known maturity states of BMDCs; other portions reflect differences in the usage of key regulatory circuits. For example, we identify a module of 137 highly variable, yet co-regulated, antiviral response genes. Using cells from knockout mice, we show that variability in this module may be propagated through an interferon feedback circuit, involving the transcriptional regulators Stat2 and Irf7. Our study demonstrates the power and promise of single-cell genomics in uncovering functional diversity between cells and in deciphering cell states and circuits.


Asunto(s)
Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Empalme del ARN/inmunología , Análisis de la Célula Individual , Transcriptoma/genética , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Hibridación Fluorescente in Situ , Factor 7 Regulador del Interferón , Interferones/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Reproducibilidad de los Resultados , Factor de Transcripción STAT2 , Análisis de Secuencia de ARN , Virus/inmunología
9.
Cancer Res ; 69(3): 836-44, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19176381

RESUMEN

Tumor dormancy has important implications for early detection and treatment of cancer. Lack of experimental models and limited clinical accessibility constitute major obstacles to the molecular characterization of dormant tumors. We have developed models in which human tumors remain dormant for a prolonged period of time (>120 days) until they switch to rapid growth and become strongly angiogenic. These angiogenic tumors retain their ability to grow fast once injected in new mice. We hypothesized that dormant tumors undergo a stable genetic reprogramming during their switch to the fast-growing phenotype. Genome-wide transcriptional analysis was done to dissect the molecular mechanisms underlying the switch of dormant breast carcinoma, glioblastoma, osteosarcoma, and liposarcoma tumors. A consensus expression signature distinguishing all four dormant versus switched fast-growing tumors was generated. In alignment with our phenotypic observation, the angiogenesis process was the most significantly affected functional gene category. The switch of dormant tumors was associated with down-regulation of angiogenesis inhibitor thrombospondin and decreased sensitivity of angiogenic tumors to angiostatin. The conversion of dormant tumors to exponentially growing tumors was also correlated with regulation and activation of pathways not hitherto linked to tumor dormancy process, such as endothelial cell-specific molecule-1, 5'-ecto-nucleotidase, tissue inhibitor of metalloproteinase-3, epidermal growth factor receptor, insulin-like growth factor receptor, and phosphatidylinositol 3-kinase signaling. Further, novel dormancy-specific biomarkers such as H2BK and Eph receptor A5 (EphA5) were discovered. EphA5 plasma levels in mice and mRNA levels in tumor specimens of glioma patients correlated with diseases stage. These data will be instrumental in identifying novel early cancer biomarkers and could provide a rationale for development of dormancy-promoting tumor therapy strategies.


Asunto(s)
Neoplasias/irrigación sanguínea , Neoplasias/genética , Angiomotinas , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones SCID , Proteínas de Microfilamentos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Fenotipo , Reacción en Cadena de la Polimerasa , Receptor EphA5/biosíntesis , Receptor EphA5/sangre , Receptor EphA5/genética , Receptor EphA5/metabolismo , Reproducibilidad de los Resultados , Transcripción Genética , Tropomiosina/metabolismo
10.
J Immunol ; 172(6): 3535-43, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15004154

RESUMEN

Carcinoembryonic Ag-related cellular adhesion molecule 1 (CEACAM1) represents a group of transmembrane protein isoforms that consist of variable numbers of extracellular Ig-like domains together with either a long cytoplasmic (cyt) tail containing two immunoreceptor tyrosine-based inhibitory motifs or a unique short cyt tail. Although CEACAM1 has been reported to be expressed on the surface of T lymphocytes upon activation, its roles in T cell regulation are controversial due to the lack of functional characterization of each individual CEACAM1 isoform. We thus cotransfected Jurkat T cells with CEACAM1 isoform-encoding constructs and an IL-2 promoter-bearing plasmid or a small interference RNA targeting src homology domain 2 containing phosphatase 1. In a luciferase reporter assay and through measurements of cytokine secretion (IL-2, IL-4, and IFN-gamma), CEACAM1 containing either a long or a short cyt tail inhibited or costimulated, respectively, TCR/CD3 complex plus CD28 mediated activation with the inhibitory functions of the long cyt tail dominating. The inhibitory function of CEACAM1, was dependent upon src homology domain 2 containing phosphatase 1 activity, required both tyrosine residues within the immunoreceptor tyrosine-based inhibitory motif domains of the cyt tail and was mediated through the mitogen-activated protein kinase pathway. CEACAM1-mediated inhibition could be functionally reconstituted by incubation of PBMC with either a CEACAM1-specific mAb or CEACAM1-Fc fusion protein in the presence of an allogeneic or mitogenic stimulus, respectively. These studies indicate that the long and short cyt tails of CEACAM1 serve as inhibitory and costimulatory receptors, respectively, in T cell regulation.


Asunto(s)
Antígenos CD/fisiología , Antígenos de Diferenciación/fisiología , Antígenos de Neoplasias/fisiología , Moléculas de Adhesión Celular/fisiología , Regulación hacia Abajo/inmunología , Inmunosupresores/farmacología , Activación de Linfocitos/inmunología , Proteínas Nucleares , Linfocitos T/inmunología , Linfocitos T/metabolismo , Regulación hacia Arriba/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Células Cultivadas , Citoplasma/inmunología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Genes Reporteros , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas Quinasas JNK Activadas por Mitógenos , Células Jurkat , Ligandos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factores de Transcripción NFATC , Fosforilación , Isoformas de Proteínas/fisiología , Proteína Fosfatasa 1 , Estructura Terciaria de Proteína , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/fisiología , Receptores Inmunológicos/fisiología , Linfocitos T/enzimología , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
11.
Am J Physiol Gastrointest Liver Physiol ; 286(6): G992-9, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-14764442

RESUMEN

Gastrin is a known growth/differentiation factor for the gastric mucosa. Its effects are likely mediated by the induction of heparin-binding epidermal-like growth factor (HB-EGF), a member of the EGF family of growth factors that is expressed by gastric parietal cells. In this study, we investigated the regulation of the HB-EGF promoter by gastrin in a human gastric cancer cell line. Serial human HB-EGF promoter-luciferase reporter deletion constructs and heterologous promoter constructs were transfected into AGS-E cells and stimulated with gastrin (10(-7) M) with or without various signal transduction inhibitors. EMSA were also performed. Gastrin stimulation resulted in a fivefold increase in HB-EGF-luciferase activity. The cis-acting element mediating gastrin responsiveness was mapped to the -69 to -58 region of the HB-EGF promoter. Gastrin stimulation was PKC dependent and at least partially mediated by activation of the EGF receptor.


Asunto(s)
Factor de Crecimiento Epidérmico/genética , Receptores ErbB/fisiología , Gastrinas/fisiología , Regiones Promotoras Genéticas/fisiología , Proteína Quinasa C/fisiología , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Gastrinas/genética , Gastrinas/farmacología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Elementos de Respuesta/genética , Elementos de Respuesta/fisiología , Transducción de Señal , Factor de Transcripción Sp1/fisiología , Estereoisomerismo , Factores de Transcripción/metabolismo , Rayos Ultravioleta , Zinc/metabolismo
12.
Artículo en Inglés | MEDLINE | ID: mdl-12816760

RESUMEN

The enterochromaffin-like (ECL) cell controls gastric acid secretion via histamine, generated by l-histidine decarboxylase (HDC). HDC expression is regulated by gastrin. However, gastrin is not alone in controlling ECL cell function. For example, the neural peptide pituitary adenylate cyclase-activating polypeptide (PACAP) also increases ECL cell proliferation. To investigate a potential role of PACAP in regulating HDC expression, we generated a series of HDC promoter-luciferase reporter constructs and transiently transfected them into PC12 cells (stably expressing the gastrin-CCK-2 receptor). We found that PACAP regulates HDC promoter activity. This is temporally biphasic, involving both adenyl cyclase and phospholipase C-dependent pathways. Deletional analysis, block mutation, and EMSA demonstrated a PACAP-response element at -177 to -170, wholly necessary for the effects of PACAP and discrete from known gastrin-responsive elements. Discrete neural and endocrine pathways regulate ECL cells through different patterns of postreceptor signaling and promoter activation, which may be appropriate to their functions in vivo.


Asunto(s)
Gastrinas/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Histidina Descarboxilasa/genética , Neuropéptidos/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Adenilil Ciclasas/metabolismo , Animales , Colforsina/farmacología , Ensayo de Cambio de Movilidad Electroforética , Humanos , Luciferasas/genética , Células PC12 , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa , Ratas , Receptor de Colecistoquinina B/efectos de los fármacos , Receptor de Colecistoquinina B/genética , Receptores de Colecistoquinina/efectos de los fármacos , Receptores de Colecistoquinina/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección
13.
Mol Endocrinol ; 16(12): 2802-18, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12456801

RESUMEN

Recently, binding of specific protein 1 (Sp1) and cAMP response element binding protein (CREB) to a GC-rich element at -92/-62 has been identified as a critical step in gastrin-dependent regulation of the chromogranin A (CgA) gene in gastric epithelial cells. Here we demonstrate that binding of early growth response protein 1 (Egr-1) to the distal part of the -92/-62 site is also required for gastrin-dependent CgA transactivation. Gastrin elevated cellular and nuclear Egr-1 levels in a time-dependent manner and also increased Egr-1 binding to the CgA -92/-73 region. Disruption of this site reduced gastrin responsiveness without influencing basal promoter activity, while loss of Sp1 and/or CREB binding sites diminished basal and gastrin-stimulated CgA promoter activity. Ectopic Egr-1 overexpression potently stimulated the CgA promoter, whereas coexpression of Egr-1 with Sp1 and/or CREB resulted in additive effects. Functional analysis of Sp1-, Egr-1-, or CREB-specific promoter mutations in transfection studies confirmed the tripartite organization of the CgA -92/-62 element. Signaling studies revealed that MAPK kinase 1 (MEK1)/ERK1/2 cascades are critical for gastrin-dependent Egr-1 protein accumulation as well as Egr-1 binding to the CgA promoter. Our studies for the first time identify Egr-1 as a nuclear target of gastrin and show that functional interplay of Egr-1, Sp1, and CREB is indispensable for gastrin-dependent CgA transactivation in gastric epithelial cells.


Asunto(s)
Cromograninas/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/metabolismo , Gastrinas/farmacología , Proteínas Inmediatas-Precoces , Factores de Transcripción/metabolismo , Sitios de Unión , Núcleo Celular/metabolismo , Cromogranina A , ADN/química , ADN/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz , Inhibidores Enzimáticos/farmacología , Células Epiteliales/metabolismo , Flavonoides/farmacología , Humanos , Immunoblotting , MAP Quinasa Quinasa 1 , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Elementos de Respuesta , Transducción de Señal , Neoplasias Gástricas , Acetato de Tetradecanoilforbol/farmacología , Activación Transcripcional , Transfección , Células Tumorales Cultivadas
14.
Biochem Biophys Res Commun ; 297(5): 1089-95, 2002 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-12372397

RESUMEN

In human gastric cancer cells the human histidine decarboxylase gene is regulated by gastrin through two overlapping cis-acting elements known as gastrin response elements 1&2 (GAS-RE1, GAS-RE2) [J. Biol. Chem. 274 (1999) 20961]. Here, we report the identification and characterization of a third element GAS-RE3 that was localized to a region +28 to +48 downstream of the transcriptional start site (+1). Gastrin stimulation induced a rapid increase in binding to the element of a novel nuclear factor named gastrin response element-binding protein 3 (GAS-REBP3). Block mutations in the GAS-RE3 sequence (+38GTGCG(+42) to +38TAAGT(+42)) led to reduced promoter activity and decreased binding in EMSA. UV cross-linking studies and Southwestern blot analysis with wildtype and mutant GAS-RE3 showed that GAS-REBP3 was a approximately 110kDa protein. Thus, gastrin-mediated regulation of HDC gene expression appears to be mediated by a complex cis-acting element, which binds at least three distinct nuclear factors.


Asunto(s)
Gastrinas/genética , Histidina Descarboxilasa/genética , Regiones Promotoras Genéticas , Elementos de Respuesta , Secuencia de Bases , Sitios de Unión , Southern Blotting , Western Blotting , Reactivos de Enlaces Cruzados/farmacología , Gastrinas/metabolismo , Genes Reporteros , Humanos , Datos de Secuencia Molecular , Mutación , Oligonucleótidos/genética , Unión Proteica , Transfección , Rayos Ultravioleta
15.
Biochem J ; 367(Pt 3): 703-14, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12162790

RESUMEN

The heavy chain (HC) of the neonatal Fc receptor (FcRn) for IgG is non-convalently associated with beta(2)-microglobulin (beta(2)m). In beta(2)m(-/-) mice, FcRn functions are greatly impaired. We sought to determine how FcRn HC, particularly its structure and biogenesis, is affected by the absence of beta(2)m. Human FcRn HC, expressed from the beta(2)m-null cell line FO-1(FcRn), was present as a monomeric 45-kDa protein under reducing conditions but primarily as a 92-kDa oligomeric protein under non-reducing conditions. Two-dimensional electrophoresis and MS analysis showed that the 92-kDa protein was a dimer of the 45-kDa HC. Immunostaining showed that FcRn HC in FO-1(FcRn) was co-localized with the endoplasmic reticulum (ER) protein Bip/GRP78 but not with an endosome protein, EEA1. In contrast, FcRn HC in FO-1(FcRn+beta2m) was detected in both the ER and endosome. The dimeric HC in FcRn oligomers was free of beta(2)m association in FO-1(FcRn+beta2m). Mutation of non-paired cysteine residues at positions 48 and 251 within the human FcRn cDNA failed to eliminate the oligomers. The FcRn HC oligomers could be reduced by reconstitution of FO-1(FcRn) with beta(2)m or by balanced expression of FcRn HC with beta(2)m, or beta(2)m fused with a KDEL retention sequence. Similarly, the majority of FcRn HC isolated from neonatal beta(2)m(-/-) mice was in a dimeric form under non-reducing conditions. The amount of FcRn HC was significantly decreased in beta(2)m(-/-) mice and FO-1(FcRn). Furthermore, beta(2)m-free FcRn HC was sensitive to endoglycosidase digestion. These results indicate that FcRn HC alone can form disulphide-bonded oligomers in the ER, which may represent a misfolded protein. The beta(2)m association with FcRn HC is critical for correct folding of FcRn and exiting the ER for routing to endosomes and the cell surface.


Asunto(s)
Retículo Endoplásmico/metabolismo , Inmunoglobulina G/metabolismo , Receptores Fc/metabolismo , Microglobulina beta-2/metabolismo , Animales , Secuencia de Bases , Cisteína/metabolismo , Cartilla de ADN , Dimerización , Chaperón BiP del Retículo Endoplásmico , Humanos , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Receptores Fc/química , Receptores Fc/genética
16.
Biochem Biophys Res Commun ; 293(1): 366-74, 2002 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-12054609

RESUMEN

Trefoil factor 2 (TFF2)/spasmolytic polypeptide (SP) is a highly stable peptide which is abundantly expressed and secreted by mucous cells of the stomach and which functions in gastric cytoprotection. Previous studies from our group have shown that TFF2 is an immediate early gene capable of regulating its own expression through activation of the TFF2 promoter. We therefore aimed to investigate the cis-acting elements mediating this response in AGS cells transfected with TFF2 promoter-reporter gene constructs, using a TFF2-expression system resembling physiologic paracrine conditions. TFF2 peptide expression was achieved through stable transfection of AGS cells with a TFF2-expression construct. Stimulation of transiently transfected cells with this TFF2-containing conditioned media resulted in a significant increase in TFF2 promoter activity. Promoter stimulation was blocked by an anti-TFF2 antibody, indicating that it was mediated specifically by TFF2. Deletion analysis of the TFF2 promoter led to the identification of a specific response element located between -191 and -174 upstream of the transcriptional initiation site. This region of the promoter, which was designated SPRE (for spasmolytic polypeptide response element), was sufficient to confer responsiveness in a heterologous promoter system. Mutational analysis and electrophoretic mobility shift assays (EMSA) showed that a GAG motif was responsible for mediating promoter activation in response to TFF2 stimulation. Since auto- and cross-induction of TFF2 promoter is likely to be a means of rapid amplification of TFF2 expression in the critical first minutes following mucosal injury, these results should lead to insight into the molecular events initiating epithelial restitution and healing.


Asunto(s)
Regulación de la Expresión Génica , Sustancias de Crecimiento/genética , Mucinas , Proteínas Musculares , Neuropéptidos , Péptidos/genética , Regiones Promotoras Genéticas , Secuencia de Bases , Línea Celular , Cartilla de ADN , Sustancias de Crecimiento/análisis , Homeostasis , Humanos , Mucosa Intestinal/fisiología , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Péptidos/análisis , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/biosíntesis , Eliminación de Secuencia , Transfección , Factor Trefoil-2 , Factor Trefoil-3
17.
J Immunol ; 168(3): 1028-35, 2002 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-11801635

RESUMEN

Carcinoembryonic Ag cell adhesion molecule 1 (CEACAM1) consists of highly related homologs in humans and rodents that are characterized by significant alternate splicing generating isoforms capable of negative intracellular signaling by virtue of two immunoreceptor tyrosine-based inhibition motifs in its cytoplasmic (cyt) tail. Although human T cells have been recently observed to express CEACAM1, the expression and function of CEACAM1 in mouse T cells have not been defined. Although resting mouse spleen T cells exhibited no evidence of CEACAM1 on the cell surface, CEACAM1 was rapidly up-regulated on CD4+ and CD8+ T cells after activation with either Con A or anti-CD3 without a requirement for either de novo transcription or translation due to the fact that CEACAM1 was present intracellularly before activation. Using a GST-CEACAM1-cytoplasmic tail fusion protein, it was shown that the cytoplasmic tail of CEACAM1 bound the src homology domain-containing phosphatase 1 and adaptor protein 1 complex in its phosphorylated and nonphosphorylated states, respectively. CEACAM1 ligation with an anti-CEACAM1 mAb resulted in inhibition of an allogeneic MLR and anti-CD3 plus anti-CD28 Ab-induced proliferation of spleen T cells in vitro and inhibition of a delayed-type hypersensitivity response to oxazolone in vivo. Inhibition of the delayed-type hypersensitivity response required that the anti-CEACAM1-specific mAb be present at the time of T cell sensitization. These studies support a role for CEACAM1 as a novel class of immunoreceptor tyrosine-based inhibition motif-bearing regulatory molecules on T cells that are active during early phases of the immune response in mice.


Asunto(s)
Antígenos CD/biosíntesis , Antígenos de Diferenciación/biosíntesis , Antígeno Carcinoembrionario/biosíntesis , Moléculas de Adhesión Celular/biosíntesis , Activación de Linfocitos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos CD/fisiología , Antígenos de Diferenciación/inmunología , Antígenos de Diferenciación/metabolismo , Antígenos de Diferenciación/fisiología , Antígeno Carcinoembrionario/inmunología , Antígeno Carcinoembrionario/metabolismo , Antígeno Carcinoembrionario/fisiología , Proteínas Portadoras/metabolismo , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Moléculas de Adhesión Celular/fisiología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Células Cultivadas , Inyecciones Intraperitoneales , Interfase/inmunología , Péptidos y Proteínas de Señalización Intracelular , Ligandos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína Fosfatasa 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas con Dominio SH2 , Linfocitos T/citología , Factores de Tiempo , Dominios Homologos src/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA