Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Mol Gastroenterol Hepatol ; 13(3): 717-737, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34781022

RESUMEN

BACKGROUND & AIMS: We previously showed that histamine suppressed inflammation-associated colonic tumorigenesis through histamine type 2 receptor (H2R) signaling in mice. This study aimed to precisely elucidate the downstream effects of H2R activation in innate immune cells. METHODS: Analyses using online databases of single-cell RNA sequencing of intestinal epithelial cells in mice and RNA sequencing of mouse immune cells were performed to determine the relative abundances of 4 histamine receptors among different cell types. Mouse neutrophils, which expressed greater amounts of H2R, were collected from the peritoneum of wild-type and H2R-deficient mice, of which low-density and high-density neutrophils were extracted by centrifugation and were subjected to RNA sequencing. The effects of H2R activation on neutrophil differentiation and its functions in colitis and inflammation-associated colon tumors were investigated in a mouse model of dextran sulfate sodium-induced colitis. RESULTS: Data analysis of RNA sequencing and quantitative reverse-transcription polymerase chain reaction showed that Hrh2 is highly expressed in neutrophils, but barely detectable in intestinal epithelial cells. In mice, the absence of H2R activation promoted infiltration of neutrophils into both sites of inflammation and colonic tumors. H2R-deficient high-density neutrophils yielded proinflammatory features via nuclear factor-κB and mitogen-activated protein kinase signaling pathways, and suppressed T-cell proliferation. On the other hand, low-density neutrophils, which totally lack H2R activation, showed an immature phenotype compared with wild-type low-density neutrophils, with enhanced MYC pathway signaling and reduced expression of the maturation marker Toll-like receptor 4. CONCLUSIONS: Blocking H2R signaling enhanced proinflammatory responses of mature neutrophils and suppressed neutrophil maturation, leading to accelerated progression of inflammation-associated colonic tumorigenesis.


Asunto(s)
Mucosa Intestinal , Neutrófilos , Animales , Carcinogénesis/patología , Homeostasis , Inflamación/patología , Mucosa Intestinal/metabolismo , Ratones , Neutrófilos/metabolismo
2.
Microbiologyopen ; 8(10): e908, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31369218

RESUMEN

The histamine H2 receptor (H2R) is a G protein-coupled receptor that mediates cyclic AMP production, protein kinase A activation, and MAP kinase signaling. In order to explore the multifaceted effects of histamine signaling on immune cells, phagocytosis was evaluated using primary mouse-derived macrophages. Phagocytosis is initiated by signaling via surface-bound scavenger receptors and can be regulated by autophagy. Absence of H2R signaling resulted in diminished phagocytosis of live bacteria and synthetic microspheres by primary macrophages from histamine H2 receptor gene (Hrh2)-deficient mice. Flow cytometry and immunofluorescence microscopy were used to quantify phagocytosis of phylogenetically diverse bacteria as well as microspheres of defined chemical composition. Autophagy and scavenger receptor gene expression were quantified in macrophages after exposure to Escherichia coli. Expression of the autophagy genes, Becn1 and Atg12, was increased in Hrh2-/- macrophages, indicating upregulation of autophagy pathways. Expression of the Macrophage Scavenger Receptor 1 gene (Msr1) was diminished in Hrh2-deficient macrophages, supporting the possible importance of histamine signaling in scavenger receptor abundance and macrophage function. Flow cytometry confirmed diminished MSR1 surface abundance in Hrh2-/- macrophages. These data suggest that H2R signaling is required for effective phagocytosis by regulating the process of autophagy and scavenger receptor MSR1 abundance in macrophages.


Asunto(s)
Macrófagos/inmunología , Fagocitosis , Receptores Histamínicos H2/metabolismo , Receptores Depuradores de Clase A/metabolismo , Transducción de Señal , Animales , Autofagia , Células Cultivadas , Escherichia coli/inmunología , Citometría de Flujo , Ratones , Microscopía Fluorescente , Microesferas , Receptores Histamínicos H2/deficiencia
3.
Am J Physiol Gastrointest Liver Physiol ; 316(1): G205-G216, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30462522

RESUMEN

Inflammatory bowel disease (IBD) is a well-known risk factor for the development of colorectal cancer. Prior studies have demonstrated that microbial histamine can ameliorate intestinal inflammation in mice. We tested the hypothesis whether microbe-derived luminal histamine suppresses inflammation-associated colon cancer in Apcmin/+ mice. Mice were colonized with the human-derived Lactobacillus reuteri. Chronic inflammation was induced by repeated cycles of low-dose dextran sulfate sodium (DSS). Mice that were given histamine-producing L. reuteri via oral gavage developed fewer colonic tumors, despite the presence of a complex mouse gut microbiome. We further demonstrated that administration of a histamine H1-receptor (H1R) antagonist suppressed tumorigenesis, while administration of histamine H2-receptor (H2R) antagonist significantly increased both tumor number and size. The bimodal functions of histamine include protumorigenic effects through H1R and antitumorigenic effects via H2R, and these results were supported by gene expression profiling studies on tumor specimens of patients with colorectal cancer. Greater ratios of gene expression of H2R ( HRH2) vs. H1R ( HRH1) were correlated with improved overall survival outcomes in patients with colorectal cancer. Additionally, activation of H2R suppressed phosphorylation of mitogen-activated protein kinases (MAPKs) and inhibited chemokine gene expression induced by H1R activation in colorectal cancer cells. Moreover, the combination of a H1R antagonist and a H2R agonist yielded potent suppression of lipopolysaccharide-induced MAPK signaling in macrophages. Given the impact on intestinal epithelial and immune cells, simultaneous modulation of H1R and H2R signaling pathways may be a promising therapeutic target for the prevention and treatment of inflammation-associated colorectal cancer. NEW & NOTEWORTHY Histamine-producing Lactobacillus reuteri can suppress development of inflammation-associated colon cancer in an established mouse model. The net effects of histamine may depend on the relative activity of H1R and H2R signaling pathways in the intestinal mucosa. Our findings suggest that treatment with H1R or H2R antagonists could yield opposite effects. However, by harnessing the ability to block H1R signaling while stimulating H2R signaling, novel strategies for suppression of intestinal inflammation and colorectal neoplasia could be developed.


Asunto(s)
Carcinogénesis/metabolismo , Inflamación/metabolismo , Receptores Histamínicos H1/metabolismo , Receptores Histamínicos H2/metabolismo , Animales , Carcinogénesis/efectos de los fármacos , Colon/efectos de los fármacos , Colon/metabolismo , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Histamina/metabolismo , Antagonistas de los Receptores Histamínicos H1/farmacología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Lipopolisacáridos/farmacología , Ratones Transgénicos , Receptores Histamínicos H1/efectos de los fármacos , Receptores Histamínicos H2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
4.
mBio ; 6(6): e01358-15, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26670383

RESUMEN

UNLABELLED: Probiotics and commensal intestinal microbes suppress mammalian cytokine production and intestinal inflammation in various experimental model systems. Limited information exists regarding potential mechanisms of probiotic-mediated immunomodulation in vivo. In this report, we demonstrate that specific probiotic strains of Lactobacillus reuteri suppress intestinal inflammation in a trinitrobenzene sulfonic acid (TNBS)-induced mouse colitis model. Only strains that possess the hdc gene cluster, including the histidine decarboxylase and histidine-histamine antiporter genes, can suppress colitis and mucosal cytokine (interleukin-6 [IL-6] and IL-1ß in the colon) gene expression. Suppression of acute colitis in mice was documented by diminished weight loss, colonic injury, serum amyloid A (SAA) protein concentrations, and reduced uptake of [(18)F]fluorodeoxyglucose ([(18)F]FDG) in the colon by positron emission tomography (PET). The ability of probiotic L. reuteri to suppress colitis depends on the presence of a bacterial histidine decarboxylase gene(s) in the intestinal microbiome, consumption of a histidine-containing diet, and signaling via the histamine H2 receptor (H2R). Collectively, luminal conversion of l-histidine to histamine by hdc(+) L. reuteri activates H2R, and H2R signaling results in suppression of acute inflammation within the mouse colon. IMPORTANCE: Probiotics are microorganisms that when administered in adequate amounts confer beneficial effects on the host. Supplementation with probiotic strains was shown to suppress intestinal inflammation in patients with inflammatory bowel disease and in rodent colitis models. However, the mechanisms of probiosis are not clear. Our current studies suggest that supplementation with hdc(+) L. reuteri, which can convert l-histidine to histamine in the gut, resulted in suppression of colonic inflammation. These findings link luminal conversion of dietary components (amino acid metabolism) by gut microbes and probiotic-mediated suppression of colonic inflammation. The effective combination of diet, gut bacteria, and host receptor-mediated signaling may result in opportunities for therapeutic microbiology and provide clues for discovery and development of next-generation probiotics.


Asunto(s)
Colitis/microbiología , Colitis/terapia , Microbioma Gastrointestinal/genética , Mucosa Intestinal/microbiología , Limosilactobacillus reuteri/fisiología , Probióticos , Receptores Histamínicos H2/metabolismo , Animales , Colitis/inducido químicamente , Colitis/inmunología , Colon/inmunología , Colon/microbiología , Colon/fisiopatología , Dieta , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/fisiología , Histamina/metabolismo , Histidina/genética , Histidina/metabolismo , Histidina Descarboxilasa/genética , Histidina Descarboxilasa/metabolismo , Inmunomodulación , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Mucosa Intestinal/inmunología , Limosilactobacillus reuteri/enzimología , Ratones , Tomografía de Emisión de Positrones , Probióticos/uso terapéutico , Receptores Histamínicos H2/genética , Proteína Amiloide A Sérica/metabolismo , Transducción de Señal , Ácido Trinitrobencenosulfónico/administración & dosificación
6.
Nucleic Acids Res ; 43(13): 6257-69, 2015 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-26040697

RESUMEN

Roles for SOX9 have been extensively studied in development and particular emphasis has been placed on SOX9 roles in cell lineage determination in a number of discrete tissues. Aberrant expression of SOX9 in many cancers, including colorectal cancer, suggests roles in these diseases as well and recent studies have suggested tissue- and context-specific roles of SOX9. Our genome wide approach by chromatin immunoprecipitation sequencing (ChIP-seq) in human colorectal cancer cells identified a number of physiological targets of SOX9, including ubiquitously expressed cell cycle regulatory genes, such as CCNB1 and CCNB2, CDK1, and TOP2A. These novel high affinity-SOX9 binding peaks precisely overlapped with binding sites for histone-fold NF-Y transcription factor. Furthermore, our data showed that SOX9 is recruited by NF-Y to these promoters of cell cycle regulatory genes and that SOX9 is critical for the full function of NF-Y in activation of the cell cycle genes. Mutagenesis analysis and in vitro binding assays provided additional evidence to show that SOX9 affinity is through NF-Y and that SOX9 DNA binding domain is not necessary for SOX9 affinity to those target genes. Collectively, our results reveal possibly a context-dependent, non-classical regulatory role for SOX9.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción SOX9/metabolismo , Activación Transcripcional , Sitios de Unión , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Genoma Humano , Humanos , Regiones Promotoras Genéticas , Factor de Transcripción SOX9/fisiología
7.
PLoS One ; 9(9): e107577, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25229425

RESUMEN

The transcription factor SOX9 plays an essential role in determining the fate of several cell types and is a master factor in regulation of chondrocyte development. Our aim was to determine which genes in the genome of chondrocytes are either directly or indirectly controlled by SOX9. We used RNA-Seq to identify genes whose expression levels were affected by SOX9 and used SOX9 ChIP-Seq to identify those genes that harbor SOX9-interaction sites. For RNA-Seq, the RNA expression profile of primary Sox9flox/flox mouse chondrocytes infected with Ad-CMV-Cre was compared with that of the same cells infected with a control adenovirus. Analysis of RNA-Seq data indicated that, when the levels of Sox9 mRNA were decreased more than 8-fold by infection with Ad-CMV-Cre, 196 genes showed a decrease in expression of at least 4-fold. These included many cartilage extracellular matrix (ECM) genes and a number of genes for ECM modification enzymes (transferases), membrane receptors, transporters, and others. In ChIP-Seq, 75% of the SOX9-interaction sites had a canonical inverted repeat motif within 100 bp of the top of the peak. SOX9-interaction sites were found in 55% of the genes whose expression was decreased more than 8-fold in SOX9-depleted cells and in somewhat fewer of the genes whose expression was reduced more than 4-fold, suggesting that these are direct targets of SOX9. The combination of RNA-Seq and ChIP-Seq has provided a fuller understanding of the SOX9-controlled genetic program of chondrocytes.


Asunto(s)
Condrocitos/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Factor de Transcripción SOX9/metabolismo , Animales , Sitios de Unión , Inmunoprecipitación de Cromatina , Expresión Génica , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Ratones Noqueados , Motivos de Nucleótidos , Posición Específica de Matrices de Puntuación , Unión Proteica , Transporte de Proteínas , Factor de Transcripción SOX9/genética
8.
Am J Physiol Gastrointest Liver Physiol ; 305(1): G74-83, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23660500

RESUMEN

SOX9 regulates cell lineage specification by directly regulating target genes in a discrete number of tissues, and previous reports have shown cell proliferative and suppressive roles for SOX9. Although SOX9 is expressed in colorectal cancer, only a few direct targets have been identified in intestinal epithelial cells. We previously demonstrated increased proliferation in Sox9-deficient crypts through loss-of-function studies, indicating that SOX9 suppresses cell proliferation. In this study, crypt epithelial cells isolated from Sox9-deficient mice were used to identify potential target genes of SOX9. Insulin-like growth factor (IGF)-binding protein 4 (IGFBP-4), an inhibitor of the IGF/IGF receptor pathway, was significantly downregulated in Sox9-deficient intestinal epithelial cells and adenoma cells of Sox9-deficient ApcMin/+ mice. Immunolocalization experiments revealed that IGFBP-4 colocalized with SOX9 in mouse and human intestinal epithelial cells and in specimens from patients with primary colorectal cancer. Reporter assays and chromatin immunoprecipitation demonstrated direct binding of SOX9 to the IGFBP-4 promoter. Overexpression of SOX9 attenuated cell proliferation, which was restored following treatment with a neutralizing antibody against IGFBP-4. These results suggest that SOX9 regulates cell proliferation, at least in part via IGFBP-4. Furthermore, the antiproliferative effect of SOX9 was confirmed in vivo using Sox9-deficient mice, which showed increased tumor burden when bred with ApcMin/+ mice. Our results demonstrate, for the first time, that SOX9 is a transcriptional regulator of IGFBP-4 and that SOX9-induced activation of IGFBP-4 may be one of the mechanisms by which SOX9 suppresses cell proliferation and progression of colon cancer.


Asunto(s)
Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Factor de Transcripción SOX9/metabolismo , Animales , Secuencia de Bases , Células CACO-2 , Proliferación Celular , Regulación de la Expresión Génica/fisiología , Humanos , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Factor de Transcripción SOX9/genética , Organismos Libres de Patógenos Específicos
9.
Am J Physiol Gastrointest Liver Physiol ; 304(1): G12-25, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23125155

RESUMEN

Intestinal ischemia-reperfusion (I/R) injury causes severe illness frequently complicated by remote multiorgan dysfunction and sepsis. Recent studies implicated interleukin-17A (IL-17A) in regulating inflammation, autoimmunity, and I/R injury. Here, we determined whether IL-17A is critical for generation of intestinal I/R injury and subsequent liver and kidney injury. Mice subjected to 30 min of superior mesenteric artery ischemia not only developed severe small intestinal injury (necrosis, apoptosis, and neutrophil infiltration) but also developed significant renal and hepatic injury. We detected large increases in IL-17A in the small intestine, liver, and plasma. IL-17A is critical for generating these injuries, since genetic deletion of IL-17A- or IL-17A-neutralizing antibody treatment markedly protected against intestinal I/R injury and subsequent liver and kidney dysfunction. Intestinal I/R caused greater increases in portal plasma and small intestine IL-17A, suggesting an intestinal source for IL-17A generation. We also observed that intestinal I/R caused rapid small intestinal Paneth cell degranulation and induced murine α-defensin cryptdin-1 expression. Furthermore, genetic or pharmacological depletion of Paneth cells significantly attenuated the intestinal I/R injury as well as hepatic and renal dysfunction. Finally, Paneth cell depletion significantly decreased small intestinal, hepatic, and plasma IL-17A levels after intestinal I/R. Taken together, we propose that Paneth cell-derived IL-17A may play a critical role in intestinal I/R injury as well as extraintestinal organ dysfunction.


Asunto(s)
Interleucina-17/fisiología , Enfermedades Intestinales/patología , Daño por Reperfusión/patología , Lesión Renal Aguda/genética , Lesión Renal Aguda/fisiopatología , Alanina Transaminasa/sangre , Animales , Apoptosis , Línea Celular , Creatinina/sangre , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Inflamación/genética , Interleucina-17/antagonistas & inhibidores , Interleucina-17/genética , Enfermedades Intestinales/genética , Hepatopatías/genética , Hepatopatías/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/fisiología , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Daño por Reperfusión/genética
10.
J Immunol ; 189(11): 5421-33, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23109723

RESUMEN

Acute kidney injury (AKI) is frequently complicated by extrarenal multiorgan injury, including intestinal and hepatic dysfunction. In this study, we hypothesized that a discrete intestinal source of proinflammatory mediators drives multiorgan injury in response to AKI. After induction of AKI in mice by renal ischemia-reperfusion or bilateral nephrectomy, small intestinal Paneth cells increased the synthesis and release of IL-17A in conjunction with severe intestinal apoptosis and inflammation. We also detected significantly increased IL-17A in portal and systemic circulation after AKI. Intestinal macrophages appear to transport released Paneth cell granule constituents induced by AKI, away from the base of the crypts into the liver. Genetic or pharmacologic depletion of Paneth cells decreased small intestinal IL-17A secretion and plasma IL-17A levels significantly and attenuated intestinal, hepatic, and renal injury after AKI. Similarly, portal delivery of IL-17A in macrophage-depleted mice decreased markedly. In addition, intestinal, hepatic, and renal injury following AKI was attenuated without affecting intestinal IL-17A generation. In conclusion, AKI induces IL-17A synthesis and secretion by Paneth cells to initiate intestinal and hepatic injury by hepatic and systemic delivery of IL-17A by macrophages. Modulation of Paneth cell dysregulation may have therapeutic implications by reducing systemic complications arising from AKI.


Asunto(s)
Lesión Renal Aguda/patología , Inflamación/patología , Riñón/patología , Pulmón/patología , Células de Paneth/patología , Daño por Reperfusión/patología , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/inmunología , Animales , Apoptosis , Inflamación/complicaciones , Inflamación/inmunología , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Riñón/inmunología , Pulmón/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Nefrectomía , Células de Paneth/inmunología , Sistema Porta/inmunología , Daño por Reperfusión/complicaciones , Daño por Reperfusión/inmunología
11.
Cell Signal ; 24(11): 2197-204, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22750291

RESUMEN

The production of type I interferon must be tightly regulated and aberrant production of type I interferon is harmful or even fatal to the host. TBK1 phosphorylation at Ser172 plays an essential role in TBK1-mediated antiviral response. However, how TBK1 activity is negatively regulated remains poorly understood. Using a functional genomics approach, we have identified PPM1B as a TBK1 phosphatase. PPM1B dephosphorylates TBK1 in vivo and in vitro. PPM1B wild-type but not its phosphatase-deficient R179G mutant inhibits TBK1-mediated antiviral response and facilitates VSV replication in the cells. Viral infection induces association of PPM1B with TBK1 in a transient fashion in the cells. Conversely, suppression of PPM1B expression enhances virus-induced IRF3 phosphorylation and IFNß production. Our study identifies a previously unrecognized role for PPM1B in the negative regulation of antiviral response by acting as a TBK1 phosphatase.


Asunto(s)
Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Replicación del ADN , Células HEK293 , Células HeLa , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/genética , Fosforilación , Unión Proteica , Proteína Fosfatasa 2C , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Vesiculovirus/genética , Vesiculovirus/metabolismo
12.
Gastroenterology ; 133(2): 539-46, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17681175

RESUMEN

BACKGROUND AND AIMS: The transcription factor SOX9 has been shown previously to have an essential role in the differentiation of a small number of discrete cell lineages. In the intestine, Sox9 is expressed in the epithelial cells of the crypts and is a target of Wnt signaling. METHODS: To examine the function of SOX9 in the intestine, we inactivated the Sox9 gene in intestinal epithelial cells by generating mice that harbored a conditional Sox9 gene and a Villin-Cre transgene. RESULTS: In the absence of SOX9, Paneth cells were not formed, but the differentiation of other intestinal epithelial cell types was unaffected. The lack of SOX9 also lead to crypt enlargement, to a marked increase in cell proliferation throughout the crypts, and to replacement of the Paneth cells by proliferating epithelial cells. CONCLUSIONS: We conclude that SOX9 is required for the differentiation of Paneth cells. Our results elucidate an essential step in the differentiation of gut epithelium.


Asunto(s)
Diferenciación Celular , Células Epiteliales/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Mucosa Intestinal/metabolismo , Células de Paneth/metabolismo , Factores de Transcripción/metabolismo , Animales , Proliferación Celular , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Mucosa Intestinal/citología , Mucosa Intestinal/crecimiento & desarrollo , Ratones , Ratones Noqueados , Microvellosidades/metabolismo , Fenotipo , Factor de Transcripción SOX9 , Factores de Tiempo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
13.
Matrix Biol ; 23(8): 499-505, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15694126

RESUMEN

Sox9 is a transcription factor that is critical for chondrogenesis, testis determination, and development of several other organs in vertebrates. Thus the levels of Sox9 protein and its activity may be tightly regulated. Here we show that inhibitors of the 26S proteasome increase both the levels of Sox9 protein and its transcriptional activity measured with Col2a1 promoter/enhancer construct in RCS cells and C3H10T1/2 cells. Indeed, in intact cells ubiquitination assays indicate that Sox9 is multiply ubiquitinated. The K398A mutation, which was introduced in a potential ubiquitin-binding site, increases the stability of Sox9 protein and its transcriptional activity of Col2a1, Col11a2, and AMH promoter/enhancer constructs without affecting the subcellular localization and the DNA binding efficiency of Sox9. Pulse-chase experiments show that the increased Sox9 levels resulting from treatment with the MG132 proteasome inhibitor or from the K398A mutation produce stabilization of the protein. Our in vitro studies indicate that the ubiquitin-proteasome proteolytic system degrades Sox9 and regulates its transcriptional activity.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina/química , Animales , Sitios de Unión , Western Blotting , Células COS , Línea Celular Tumoral , Células Cultivadas , Condrocitos/metabolismo , Colágeno Tipo II/genética , Elementos de Facilitación Genéticos , Proteínas del Grupo de Alta Movilidad/química , Humanos , Ratones , Ratones Endogámicos C3H , Microscopía Fluorescente , Mutación , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Estructura Terciaria de Proteína , Ratas , Factor de Transcripción SOX9 , Factores de Tiempo , Factores de Transcripción/química , Transcripción Genética , Transfección , Ubiquitina/metabolismo
14.
Genes Dev ; 18(9): 1072-87, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15132997

RESUMEN

Chondrogenesis is a multistep process that is essential for endochondral bone formation. Previous results have indicated a role for beta-catenin and Wnt signaling in this pathway. Here we show the existence of physical and functional interactions between beta-catenin and Sox9, a transcription factor that is required in successive steps of chondrogenesis. In vivo, either overexpression of Sox9 or inactivation of beta-catenin in chondrocytes of mouse embryos produces a similar phenotype of dwarfism with decreased chondrocyte proliferation, delayed hypertrophic chondrocyte differentiation, and endochondral bone formation. Furthermore, either inactivation of Sox9 or stabilization of beta-catenin in chondrocytes also produces a similar phenotype of severe chondrodysplasia. Sox9 markedly inhibits activation of beta-catenin-dependent promoters and stimulates degradation of beta-catenin by the ubiquitination/proteasome pathway. Likewise, Sox9 inhibits beta-catenin-mediated secondary axis induction in Xenopus embryos. Beta-catenin physically interacts through its Armadillo repeats with the C-terminal transactivation domain of Sox9. We hypothesize that the inhibitory activity of Sox9 is caused by its ability to compete with Tcf/Lef for binding to beta-catenin, followed by degradation of beta-catenin. Our results strongly suggest that chondrogenesis is controlled by interactions between Sox9 and the Wnt/beta-catenin signaling pathway.


Asunto(s)
Condrocitos/citología , Proteínas del Citoesqueleto/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Transactivadores/fisiología , Factores de Transcripción/fisiología , Animales , Sitios de Unión , Diferenciación Celular , División Celular , Condrocitos/fisiología , Ciclina D1/metabolismo , Proteínas del Citoesqueleto/genética , Elementos de Facilitación Genéticos , Expresión Génica , Heterocigoto , Proteínas del Grupo de Alta Movilidad/química , Proteínas del Grupo de Alta Movilidad/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Modelos Biológicos , Fenotipo , Factor de Transcripción SOX9 , Transducción de Señal , Transactivadores/genética , Factores de Transcripción/química , Factores de Transcripción/genética , Transcripción Genética , Proteínas de Xenopus , Xenopus laevis , beta Catenina
15.
Proc Natl Acad Sci U S A ; 100(16): 9360-5, 2003 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-12878728

RESUMEN

Sox9 has essential roles in endochondral bone formation during axial and appendicular skeletogenesis. Sox9 is also expressed in neural crest cells, but its function in neural crest remains largely unknown. Because many craniofacial skeletal elements are derived from cranial neural crest (CNC) cells, we asked whether deletion of Sox9 in CNC cells by using the Cre recombinase/loxP recombination system would affect craniofacial development. Inactivation of Sox9 in neural crest resulted in a complete absence of cartilages and endochondral bones derived from the CNC. In contrast, all of the mesodermal skeletal elements and intramembranous bones were essentially conserved. The migration and the localization of Sox9-null mutant CNC cells were normal. Indeed, the size of branchial arches and the frontonasal mass of mutant embryos was comparable to that of WT embryos, and the pattern of expression of Ap2, a marker of migrating CNC cells, was normal. Moreover, in mouse embryo chimeras Sox9-null mutant cells migrated to their correct location in endochondral skeletal elements; however, Sox9-null CNC cells were unable to contribute chondrogenic mesenchymal condensations. In mutant embryos, ectopic expression of osteoblast marker genes, such as Runx2, Osterix, and Col1a1, was found in the locations where the nasal cartilages exist in WT embryos. These results indicate that inactivation of Sox9 causes CNC cells to lose their chondrogenic potential. We hypothesize that these cells change their cell fate and acquire the ability to differentiate into osteoblasts. We conclude that Sox9 is required for the determination of the chondrogenic lineage in CNC cells.


Asunto(s)
Condrocitos/citología , Proteínas del Grupo de Alta Movilidad/fisiología , Proteínas de Neoplasias , Cresta Neural/citología , Factores de Transcripción/fisiología , Animales , Desarrollo Óseo , Linaje de la Célula , Movimiento Celular , Sistema Nervioso Central/embriología , Condrocitos/metabolismo , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Proteínas del Grupo de Alta Movilidad/metabolismo , Ratones , Ratones Transgénicos , Mutación , Cresta Neural/embriología , Osteoblastos/citología , Osteoblastos/metabolismo , Factor de Transcripción SOX9 , Factores de Tiempo , Factores de Transcripción/metabolismo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...