Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Oncogene ; 37(2): 197-207, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28892049

RESUMEN

Mucin-4 (Muc4) is a large cell surface glycoprotein implicated in the protection and lubrication of epithelial structures. Previous studies suggest that aberrantly expressed Muc4 can influence the adhesiveness, proliferation, viability and invasiveness of cultured tumor cells, as well as the growth rate and metastatic efficiency of xenografted tumors. Although it has been suggested that one of the major mechanisms by which Muc4 potentiates tumor progression is via its engagement of the ErbB2/HER2 receptor tyrosine kinase, other mechanisms exist and remain to be delineated. Moreover, the requirement for endogenous Muc4 for tumor growth progression has not been previously explored in the context of gene ablation. To assess the contribution of endogenous Muc4 to mammary tumor growth properties, we first created a genetically engineered mouse line lacking functional Muc4 (Muc4ko), and then crossed these animals with the NDL (Neu DeLetion mutant) model of ErbB2-induced mammary tumorigenesis. We observed that Muc4ko animals are fertile and develop normally, and adult mice exhibit no overt tissue abnormalities. In tumor studies, we observed that although some markers of tumor growth such as vascularity and cyclin D1 expression are suppressed, primary mammary tumors from Muc4ko/NDL female mice exhibit similar latencies and growth rates as Muc4wt/NDL animals. However, the presence of lung metastases is markedly suppressed in Muc4ko/NDL mice. Interestingly, histological analysis of lung lesions from Muc4ko/NDL mice revealed a reduced association of disseminated cells with platelets and white blood cells. Moreover, isolated cells derived from Muc4ko/NDL tumors interact with fewer blood cells when injected directly into the vasculature or diluted into blood from wild type mice. We further observed that blood cells more efficiently promote the viability of non-adherent Muc4wt/NDL cells than Muc4ko/NDL cells. Together, our observations suggest that Muc4 may facilitate metastasis by promoting the association of circulating tumor cells with blood cells to augment tumor cell survival in circulation.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias Pulmonares/patología , Neoplasias Mamarias Experimentales/patología , Mucina 4/metabolismo , Receptor ErbB-2/metabolismo , Animales , Apoptosis , Células Sanguíneas/patología , Neoplasias de la Mama/sangre , Neoplasias de la Mama/genética , Supervivencia Celular , Progresión de la Enfermedad , Femenino , Humanos , Pulmón/patología , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Masculino , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/sangre , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Noqueados , Mucina 4/genética , Células Neoplásicas Circulantes/patología , Receptor ErbB-2/genética
2.
Oncogene ; 36(36): 5158-5167, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28481871

RESUMEN

The lethality of the aggressive brain tumor glioblastoma multiforme (GBM) results in part from its strong propensity to invade surrounding normal brain tissue. Although oncogenic drivers such as epidermal growth factor receptor activation and Phosphatase and Tensin homolog inactivation are thought to promote the motility and invasiveness of GBM cells via phosphatidylinostitol 3-kinase activation, other unexplored mechanisms may also contribute to malignancy. Here we demonstrate that several components of the planar cell polarity (PCP) arm of non-canonical Wnt signaling including VANGL1, VANGL2 and FZD7 are transcriptionally upregulated in glioma and correlate with poorer patient outcome. Knockdown of the core PCP pathway component VANGL1 suppresses the motility of GBM cell lines, pointing to an important mechanistic role for this pathway in glioblastoma malignancy. We further observe that restoration of Nrdp1, a RING finger type E3 ubiquitin ligase whose suppression in GBM also correlates with poor prognosis, reduces GBM cell migration and invasiveness by suppressing PCP signaling. Our observations indicate that Nrdp1 physically interacts with the Vangl1 and Vangl2 proteins to mediate the K63-linked polyubiquitination of the Dishevelled, Egl-10 and Pleckstrin (DEP) domain of the Wnt pathway protein Dishevelled (Dvl). Ubiquitination hinders Dvl binding to phosphatidic acid, an interaction necessary for efficient Dvl recruitment to the plasma membrane upon Wnt stimulation of Fzd receptor and for the propagation of downstream signals. We conclude that the PCP pathway contributes significantly to the motility and hence the invasiveness of GBM cells, and that Nrdp1 acts as a negative regulator of PCP signaling by inhibiting Dvl through a novel polyubiquitination mechanism. We propose that the upregulation of core PCP components, together with the loss of the key negative regulator Nrdp1, act coordinately to promote GBM invasiveness and malignancy.


Asunto(s)
Polaridad Celular , Proteínas Dishevelled/metabolismo , Glioblastoma/metabolismo , Poliubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Células Cultivadas , Proteínas Dishevelled/genética , Glioblastoma/patología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Proteínas Wnt/metabolismo
3.
Brain Res ; 1363: 159-69, 2010 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-20869350

RESUMEN

Malignant glioma cells maintain an elevated intracellular pH (pH(i)) within hypoxic-ischemic tumor microenvironments through persistent activation of sodium-proton transport (McLean et al., 2000). Amiloride has been reported to selectively kill human malignant glioma cell lines but not primary astrocytes (Hegde et al., 2004). While amiloride reduces pH(i) of malignant gliomas by inhibiting isoform 1 of sodium-proton exchange (NHE1), direct acidification was shown to be cytostatic rather than cytotoxic. At cytotoxic concentrations, amiloride has multiple drug targets including inhibition of NHE1 and sodium-calcium exchange. Amiloride's glioma cytotoxicity can be explained, at least in part, by dual inhibition of NHE1 and of Na(+)-dependent calcium efflux by isoform 1.1 of the sodium-calcium exchanger (NCX1.1), which increases [Ca(2+)](i) and initiates glioma cell demise. As a result of persistent NHE1 activity, cytosolic free levels of sodium ([Na(+)](i)) in U87 and C6 glioma cells are elevated 3-fold, as compared with normal astrocytes. Basal cytosolic free calcium levels ([Ca(2+)](i)) also are increased 5-fold. 2', 4'-dichlorobenzamil (DCB) inhibits the sodium-dependent calcium transporter (NCX1.1) much more potently than NHE1. DCB was employed in a concentration-dependent fashion in glioma cells to selectively inhibit the forward mode of NCX1.1 at ≤1µM, while dually inhibiting both NHE1 and NCX1.1 at ≥20µM. DCB (1µM) was not cytotoxic to glioma cells, while DCB (20µM) further increased basal elevated levels of [Ca(2+)](i) in glioma cells that was followed by cell demise. Cariporide and SEA0400 are more selective inhibitors of NHE1 and NCX1.1 than amiloride or DCB, respectively. Individually, Cariporide and SEA0400 are not cytotoxic, but in combination induced glioma cell death. Like amiloride, the combination of Cariporide and SEA0400 produced glioma cell death in the absence of demonstrable caspase activation.


Asunto(s)
Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Calcio/metabolismo , Muerte Celular/fisiología , Glioma/metabolismo , Sodio/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacología , Compuestos de Anilina/farmacología , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Neoplasias Encefálicas/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Citosol/metabolismo , Glioma/patología , Guanidinas/farmacología , Humanos , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , Ionomicina/farmacología , Ionóforos/farmacología , Espacio Personal , Éteres Fenílicos/farmacología , Protones , Ratas , Ratas Sprague-Dawley , Intercambiador de Sodio-Calcio/antagonistas & inhibidores , Intercambiador de Sodio-Calcio/metabolismo , Intercambiador 1 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/metabolismo , Sulfonas/farmacología , Microambiente Tumoral/fisiología
4.
Oncogene ; 27(43): 5741-52, 2008 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-18542056

RESUMEN

Epidermal growth factor receptor (EGFR) mutation is frequently observed in human cancer and contributes to the growth, survival and therapeutic resistance of tumors. EGFRvIII is an oncogenic EGFR mutant resulting from the deletion of exons 2-7 and is the most common EGFR mutant observed in glioblastoma multiforme, an aggressive brain tumor. EGFRvIII is constitutively active but poorly ubiquitinated, leading to inefficient receptor trafficking to lysosomes and unattenuated oncogenic signaling. The mechanism by which EGFRvIII evades downregulation is not fully understood although recent studies suggest that its interaction with the ubiquitin ligase Cbl may be compromised. In this study, we examine the regulation of EGFRvIII by the recently identified negative regulator, LRIG1, which targets EGFR through recognition of its extracellular domain. Here, we determine whether the extracellular domain deletion in EGFRvIII renders it refractory to LRIG1 regulation. We find that EGFRvIII retains interaction with LRIG1 and is in fact more sensitive to LRIG1 action than wild-type receptor. We demonstrate that LRIG1 regulation of EGFRvIII is distinct from the only other known mechanism of EGFR regulation, Cbl-mediated degradation. Ectopic expression of LRIG1 in EGFRvIII(+) glioblastoma cells opposes EGFRvIII-driven tumor cell proliferation, survival, motility and invasion. Finally, RNAi-mediated silencing of LRIG1 alters EGFRvIII intracellular trafficking and leads to enhanced EGFRvIII expression, suggesting that loss of LRIG1 in tumors may contribute to a permissive environment for EGFRvIII overexpression, contributing to EGFRvIII oncogenesis.


Asunto(s)
Receptores ErbB/fisiología , Glioblastoma/genética , Glicoproteínas de Membrana/fisiología , Apoptosis , Glioblastoma/patología , Humanos , Glicoproteínas de Membrana/química , Mutación , Fosforilación , Proteínas Proto-Oncogénicas c-cbl/fisiología
5.
Br J Cancer ; 90(2): 289-93, 2004 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-14735165

RESUMEN

Receptors of the EGF receptor or ErbB family of growth factor receptor tyrosine kinases are frequently overexpressed in a variety of solid tumours, and the aberrant activation of their tyrosine kinase activities is thought to contribute to tumour growth and progression. Much effort has been put into developing inhibitors of ErbB receptors, and both antibody and small-molecule approaches have exhibited clinical success. Recently, a number of endogenous negative regulatory proteins have been identified that suppress the signalling activity of ErbB receptors in cells. These include intracellular RING finger E3 ubiquitin ligases such as cbl and Nrdp1 that mediate ErbB receptor degradation, and may include a wide variety of secreted and transmembrane proteins that suppress receptor activation by growth factor ligands. It will be of interest to determine the extent to which tumour cells suppress these pathways to promote their progression, and whether restoration of endogenous receptor-negative regulatory pathways may be exploited for therapeutic benefit.


Asunto(s)
Receptores ErbB/biosíntesis , Regulación Neoplásica de la Expresión Génica , Genes erbB/fisiología , Neoplasias/fisiopatología , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Progresión de la Enfermedad , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias/genética , Proteína Oncogénica v-cbl , Proteínas/farmacología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Proteínas Oncogénicas de Retroviridae/farmacología , Transducción de Señal , Ubiquitina-Proteína Ligasas
6.
Reprod Domest Anim ; 38(1): 63-5, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12535332

RESUMEN

The non-invasive type of implantation in the pig is characterized by the maintenance of a thick glycocalyx coating on the uterine epithelial surface microvilli. Present study investigated the alteration in the sialomucin complex (Muc4) expression during the oestrous cycle and early pregnancy in the pig. Endometrial tissue samples were immunostained with the primary antibody to the Muc4 transmembrane subunit ASGP-2. Muc4 immunostaining increased in the surface and glandular epithelia between days 5 and 10 of oestrous cycle. Immunostaining continued to increase on day 12 with the greatest intensity of uterine Muc4 immunostaining detected on day 15 of the oestrous cycle and early pregnancy. Endometrial Muc4 expression in cyclic gilts decreased dramatically during early proestrous but continued to remain abundant in the surface and glandular epithelium of pregnant gilts during the period of conceptus attachment to the uterine surface.


Asunto(s)
Endometrio/metabolismo , Ciclo Estral/metabolismo , Mucinas/metabolismo , Preñez/metabolismo , Porcinos/metabolismo , Animales , Ciclo Estral/fisiología , Femenino , Inmunohistoquímica/veterinaria , Mucina 4 , Mucinas/inmunología , Mucinas/fisiología , Embarazo , Preñez/fisiología , Porcinos/fisiología
7.
Invest Ophthalmol Vis Sci ; 42(12): 2749-56, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11687512

RESUMEN

PURPOSE: To show the presence and forms of sialomucin complex (rat Muc4) and receptor tyrosine kinase ErbBs in the rat lacrimal gland and analyze for complexes of ErbB2 and its ligand Muc4. METHODS: Northern blot analyses were used to identify sialomucin complex/Muc4 (SMC/Muc4) mRNA in rat lacrimal gland. Immunoblot analyses were performed to detect SMC/Muc4 and ErbBs. Sequential immunoprecipitation and immunoblot analyses were used to differentiate membrane and soluble forms of the SMC/Muc4 transmembrane subunit ASGP-2. Methacarn-fixed, paraffin-embedded sections of lacrimal glands from female adult rats were immunocytochemically stained using antisera to SMC/Muc4 and ErbBs to determine their relative locations in the gland. Colocalization of SMC/Muc4 and ErbB2 was confirmed by confocal immunofluorescence. Sequential immunoprecipitation and immunoblot were performed to analyze complexes of the SMC/Muc4 and ErbB2 in the lacrimal tissue. RESULTS: Northern blot analyses of rat lacrimal glands, with a probe for SMC/Muc4, demonstrated the presence of a approximately 9-kb transcript, consistent with observations in other tissues. Similarly, immunoblot analyses with antibodies against both the transmembrane (ASGP-2) and mucin (ASGP-1) subunits showed the presence of the two SMC/Muc4 subunits in lysates from rat lacrimal gland. Significantly, two different forms of ASGP-2 were observed, a high-molecular-weight ( approximately 200-kDa) form and the more common 120- to 140-kDa form. Sequential immunoprecipitation and immunoblot analyses to differentiate membrane and soluble forms of SMC/Muc4 indicated that the high-molecular-weight form of ASGP-2 was predominantly associated with membranes, whereas the 120- to 140-kDa form was both membrane-associated and soluble. The lacrimal gland consists of acini connected by intercalated and interlobular ducts. Both acini and some intercalated ducts were stained by anti-ASGP-2 monoclonal antisera. Two patterns of acinar staining were observed: membrane staining at the borders of the epithelial cells and a granular staining within the cells. Staining of ductal surfaces with antibody to the cytoplasmic domain of ASGP-2 suggests that membrane SMC/Muc4 is being produced by the ductal cells and is not simply an adsorbed soluble product from the acinar cells. Immunoblot and immunocytochemical analyses demonstrated the presence of all four ErbBs, with ErbB2 showing the most widespread distribution, similar to that of SMC/Muc4. Immunofluorescence colocalization of membrane SMC/Muc4 and ErbB2 and coimmunoprecipitation of a complex of the two provided evidence of their association in membranes of lacrimal gland acinar cells. CONCLUSIONS: SMC/Muc4 is produced by the rat lacrimal gland as both membrane and soluble forms, specifically associated with both acinar and ductal cells. Because sialomucin complex is also present in the ocular tear film, the rat lacrimal gland represents a second source of this mucin for the tear film, in addition to the corneal and conjunctival epithelia. Moreover, the presence of a complex of SMC/Muc4 and the receptor tyrosine kinase ErbB2 in lacrimal tissue suggests that SMC/Muc4 acts as a ligand for the receptor and has functions in the lacrimal gland other than that of a mucin.


Asunto(s)
Aparato Lagrimal/metabolismo , Mucinas/biosíntesis , Receptor ErbB-2/metabolismo , Animales , Northern Blotting , Electroforesis en Gel de Poliacrilamida , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Immunoblotting , Técnicas para Inmunoenzimas , Microscopía Confocal , Peso Molecular , Mucina 4 , Mucinas/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Sialoglicoproteínas/metabolismo
8.
J Cell Physiol ; 189(2): 162-70, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11598901

RESUMEN

ErbB2 has been implicated in numerous functions, including normal and aberrant development of a variety of tissues. Although no soluble ligand has been identified for ErbB2, we have recently shown that ASGP-2, the transmembrane subunit of the cell surface glycoprotein Muc4 (also called sialomucin complex, SMC), can act as an intramembrane ligand for ErbB2 and modulate its activity. Muc4/SMC is abundantly expressed at the apical surface of most epithelia of the rat female reproductive tract. Since Muc4/SMC can interact with ErbB2 when they are expressed in the same cell and membrane, we investigated whether these two proteins are co-expressed and co-localized in tissues of the female reproductive tract. Using an anti-ErbB2 antibody from Dako, we found moderate staining at the basolateral surface of the oviduct and also around the cell membrane of the most superficial and medial layers of the stratified epithelia of the vagina. In contrast, Neomarkers neu Ab1 antibody intensely stained the apical surface of the epithelium of the oviduct and the medial and basal layers of the stratified epithelia of the vagina, substantially overlapping the distribution of Muc4/SMC. Furthermore, Muc4/SMC and ErbB2 association in different tissues of the female reproductive tract was demonstrated by co-immunoprecipitation analysis. Interestingly, phosphorylated ErbB2 detected by anti-phospho-ErbB2 is primarily present at the apical surface of the oviduct. Thus, our results show that differentially localized forms of ErbB2 are recognized by different antibodies and raise interesting questions about the nature of the different forms of ErbB2, the mechanism for differential localization, and possible functions of ErbB2 in the female reproductive tract. They also raise a cautionary note about the use of different ErbB2 antibodies for expression and localization studies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Genitales Femeninos/química , Receptor ErbB-2/análisis , Animales , Epitelio/química , Femenino , Genitales Femeninos/anatomía & histología , Técnicas para Inmunoenzimas , Mucina 4 , Mucinas/metabolismo , Oviductos/anatomía & histología , Oviductos/química , Fosforilación , Ratas , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Vagina/anatomía & histología , Vagina/química
9.
J Mammary Gland Biol Neoplasia ; 6(3): 323-37, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11547901

RESUMEN

MUC4 is a one of the membrane mucins of the mucin gene (MUC) family, characterized by mucin tandem repeat domains and a transmembrane domain which associates it with the cell plasma membrane. Although MUC4 is encoded by a single gene, it is produced by epithelial cells as a heterodimer through a proteolytic cleavage mechanism. This heterodimer is found in both membrane and soluble forms associated with epithelia. Functionally, MUC4 is proposed to provide a protective mechanism for vulnerable epithelia, such as those of the airway, eye, female reproductive tract and mammary gland. The protective mechanism(s) may be highjacked by some carcinomas, such as those of the breast, to increase tumor progression. Two mechanisms are proposed to contribute to the MUC4 functions. First, MUC4 acts as an anti-adhesive or anti-recognition barrier at epithelial or tumor cell surfaces. Second, MUC4 can bind the receptor tyrosine kinase ErbB2 and alter its cellular signaling. Expression of MUC4 in mammary gland is repressed by posttranscriptional mechanisms involving basement membrane and TGF-beta, which are relieved during pregnancy to permit secretion of MUC4 into milk. These mechanisms are also abrogated in some breast cancers, providing a scenario for promotion of tumor progression. These observations imply important functions for MUC4 in both normal mammary function and in breast cancer.


Asunto(s)
Antígenos de Superficie/metabolismo , Neoplasias de la Mama/metabolismo , Mucinas/metabolismo , Animales , Antígenos de Superficie/genética , Biomarcadores de Tumor/metabolismo , Mama/metabolismo , Neoplasias de la Mama/genética , Adhesión Celular/fisiología , Progresión de la Enfermedad , Femenino , Humanos , Mucina 4 , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
10.
J Biol Chem ; 276(38): 35239-42, 2001 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-11483589

RESUMEN

The DF3/MUC1 mucin-like, transmembrane glycoprotein is aberrantly overexpressed in most human carcinomas. The MUC1 cytoplasmic domain interacts with the c-Src tyrosine kinase and thereby increases binding of MUC1 and beta-catenin. In the present work, coimmunoprecipitation studies demonstrate that MUC1 associates constitutively with the epidermal growth factor receptor (EGF-R) in human ZR-75-1 breast carcinoma cells. Immunofluorescence studies show that EGF-R and MUC1 associate at the cell membrane. We also show that the activated EGF-R phosphorylates the MUC1 cytoplasmic tail on tyrosine at a YEKV motif that functions as a binding site for the c-Src SH2 domain. The results demonstrate that EGF-R-mediated phosphorylation of MUC1 induces binding of MUC1 to c-Src in cells. Moreover, in vitro and in vivo studies demonstrate that EGF-R increases binding of MUC1 and beta-catenin. These findings support a novel role for EGF-R in regulating interactions of MUC1 with c-Src and beta-catenin.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas del Citoesqueleto/metabolismo , Receptores ErbB/metabolismo , Mucina-1/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transactivadores , Humanos , Microscopía Fluorescente , Fosforilación , Unión Proteica , Células Tumorales Cultivadas , beta Catenina
11.
Biochem Soc Trans ; 29(Pt 2): 162-6, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11356146

RESUMEN

Sialomucin complex (SMC/rat Muc4) is a heterodimeric glycoprotein complex composed of an anti-adhesive mucin subunit ascites sialoglycoprotein (ASGP)-1 and a transmembrane subunit ASGP-2. SMC expression is tightly regulated in the uterus, and its expression appears to block blastocyst implantation. Expression is controlled by steroid hormone levels in the uterine luminal epithelium, but not the uterine glandular epithelium, oviduct, cervix or vagina. Increased progesterone levels lead to downregulation of SMC in the uterine luminal epithelium at the time of receptivity for implantation. Transforming growth factor beta (TGF-beta) has been implicated as a factor in uterine progesterone responses. Studies on primary rat uterine luminal epithelial cells showed that both SMC protein and transcript are downregulated by TGF-beta1, although SMC expression is not altered by treatments with oestrogen or progesterone. SMC is also downregulated when epithelial cells are co-cultured with isolated uterine stromal cells. Oestradiol and anti-TGF-beta block the stromal cell effect. These data indicate that uterine epithelial cells respond to hormones to downregulate SMC via an indirect effect on stromal cells involving paracrine action of TGF-beta.


Asunto(s)
Mucinas/metabolismo , Útero/metabolismo , Animales , Células Cultivadas , Implantación del Embrión , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Estrógenos/farmacología , Estro/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Mucina 4 , Embarazo , Ratas , Factor de Crecimiento Transformador beta/farmacología , Útero/citología , Útero/efectos de los fármacos
12.
Oncogene ; 20(4): 461-70, 2001 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-11313977

RESUMEN

Overexpression of the membrane mucin MUC4/Sialomucin complex (SMC) has been observed during malignant progression of mammary tumors in both humans and rats, suggesting that deregulation of MUC4/SMC expression might facilitate development of these malignancies. As previously reported, overexpression of SMC results in suppression of both cell adhesion and immune killing of tumor cells. SMC also acts as a ligand for ErbB2/Neu, modulating phosphorylation of the receptor tyrosine kinase in the presence and absence of heregulin. The present studies investigated the effect of Muc4/SMC up-regulation on primary tumor growth using a tetracycline-inducible SMC expression system in a xenotransplanted tumor model. SMC up-regulation provoked rapid growth of transfected A375 melanoma in nude mice. Up-regulation of SMC, however, did not significantly increase proliferation of A375 cells in vitro. Instead, a strong suppression of apoptosis was observed in situ in SMC-overexpressing tumors. These data suggest that Muc4/SMC expression promotes tumor growth in vivo at least in part via suppression of tumor cell apoptosis. Importantly, reduction of apoptosis was also observed in vitro, indicating that anti-apoptotic effect of SMC is independent of tumor-host interactions. These findings strongly suggest that SMC up-regulation alters intracellular signaling to favor cell survival, providing for the first time evidence for the regulation of programmed cell death by a gene of the MUC family.


Asunto(s)
Apoptosis , Melanoma Experimental/metabolismo , Mucinas/metabolismo , Receptor ErbB-2/metabolismo , Melanoma Experimental/etiología , Mucina 4 , Poli Adenosina Difosfato Ribosa/metabolismo , Sialomucinas , Trasplante Heterólogo
13.
Tissue Cell ; 33(1): 111-8, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11292166

RESUMEN

Sialomucin Complex (SMC; Muc4) is a heterodimeric glycoprotein consisting of two subunits, the mucin component ASGP-1 and the transmembrane subunit ASGP-2. Northern blot and immunoblot analyses demonstrated the presence of SMC/Muc4 in submaxillary, sublingual and parotid salivary glands of the rat. Immunocytochemical staining of SMC using monoclonal antisera raised against ASGP-2 and glycosylated ASGP-1 on paraffin-embedded sections of parotid, submaxillary and sublingual tissues was performed to examine the localization of the mucin in the major rat salivary glands. Histological and immunocytochemical staining of cell markers showed that the salivary glands consisted of varying numbers of serous and mucous acini which are drained by ducts. Parotid glands were composed almost entirely of serous acini, sublingual glands were mainly mucous in composition and a mixture of serous and mucous acini were present in submaxillary glands. Since immunoreactive (ir)-SMC was specifically localized to the serous cells, staining was most abundant in parotid glands, intermediate levels in submaxillary glands and least in sublingual glands. Ir-SMC in sublingual glands was localized to caps of cells around mucous acini, known as serous demilunes, which are also present in submaxillary glands. Immunocytochemical staining of SMC in human parotid glands was localized to epithelial cells of serous acini and ducts. However, the staining pattern of epithelial cells was heterogeneous, with ir-SMC present in some acinar and ductal epithelial cells but not in others. This report provides a map of normal ir-SMC/Muc4 distribution in parotid, submaxillary and sublingual glands which can be used for the study of SMC/Muc4 expression in salivary gland tumors.


Asunto(s)
Mucinas/análisis , Glándula Parótida/anatomía & histología , Glándula Parótida/metabolismo , Glándula Sublingual/metabolismo , Glándula Submandibular/metabolismo , Animales , Northern Blotting , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Mucina 4 , Mucinas/genética , Mucinas/inmunología , Pruebas de Precipitina , ARN Mensajero/análisis , Ratas , Ratas Endogámicas F344 , Sialoglicoproteínas/inmunología , Glándula Sublingual/anatomía & histología , Glándula Submandibular/anatomía & histología
14.
J Biol Chem ; 276(25): 22685-98, 2001 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-11297548

RESUMEN

The mechanisms by which receptor tyrosine kinases (RTKs) utilize intracellular signaling pathways to direct gene expression and cellular response remain unclear. A current question is whether different RTKs within a single cell target similar or different sets of genes. In this study we have used the ErbB receptor network to explore the relationship between RTK activation and gene expression. We profiled growth factor-stimulated signaling pathway usage and broad gene expression patterns in two human mammary tumor cell lines expressing different complements of ErbB receptors. Although the growth factors epidermal growth factor (EGF) and neuregulin (NRG) 1 similarly stimulated Erk1/2 in MDA-MB-361 cells, EGF acting through an EGF receptor/ErbB2 heterodimer preferentially stimulated protein kinase C, and NRG1beta acting through an ErbB2/ErbB3 heterodimer preferentially stimulated Akt. The two growth factors regulated partially overlapping yet distinct sets of genes in these cells. In MDA-MB-453 cells, NRG1beta acting through an ErbB2/ErbB3 heterodimer stimulated prolonged signaling of all pathways examined relative to NRG2beta acting through the same heterodimeric receptor species. Surprisingly, NRG1beta and NRG2beta also regulated partially overlapping but distinct sets of genes in these cells. These results demonstrate that the activation of different RTKs, or activation of the same RTKs with different ligands, can lead to distinct profiles of gene regulation within a single cell type. Our observations also suggest that the identity and kinetics of signaling pathway usage by RTKs may play a role in the selection of regulated genes.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Expresión Génica/fisiología , Neurregulina-1/metabolismo , Proteínas Tirosina Quinasas Receptoras/fisiología , Transducción de Señal/fisiología , Animales , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Células Tumorales Cultivadas
15.
Curr Opin Cell Biol ; 13(2): 125-30, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11248544

RESUMEN

The ErbB family of receptor tyrosine kinases serves as a model for understanding the propagation of growth factor signals across the plasma membrane and the interpretation of those signals into a cellular response. Recent studies point to a critical role for the accumulation of ErbBs at specific cell-surface locations in the fidelity of ErbB signaling. The past year has witnessed significant advances in our understanding of the molecular mechanisms of ErbB localization and the role of PDZ-domain-containing proteins and cell-surface glycoproteins in directly modulating signaling through ErbBs.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Membrana Celular/metabolismo , Receptores ErbB/metabolismo , Humanos , Modelos Biológicos , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Receptor ErbB-4 , Transducción de Señal
16.
Otolaryngol Head Neck Surg ; 124(2): 127-41, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11226945

RESUMEN

OBJECTIVES: This study investigates MUC4 expression in normal squamous epithelia and squamous cell carcinoma (SCC) of the upper aerodigestive tract (UADT), and in salivary gland neoplasms. STUDY DESIGN: MUC4 antigens in tumor and adjacent normal tissue are localized by immunocytochemical studies. Fresh frozen tissues from surgical resection specimens are further analyzed by Western blot. RESULTS: MUC4 is identified by immunocytochemical staining throughout the normal UADT mucosa, in 34 of 40 primary UADT SCC, and in 11 of 12 metastatic cervical lymph nodes. A trend toward decreased MUC4 staining in moderately and poorly differentiated tumors is noted. Immunoblots show MUC4 in 4 of 5 SCC analyzed. Immunocytochemical staining of MUC4 in 13 major and minor salivary gland neoplasms reveal variable staining of normal and neoplastic tissue. MUC4 is demonstrated in immunoblots of normal parotid tissue and in the single parotid malignancy analyzed, but is not demonstrated in one minor salivary gland malignancy. These findings characterize normal UADT mucosal and salivary MUC4 expression, and MUC4 expression in SCC of the UADT and in salivary gland tumors. SIGNIFICANCE: Correlation of MUC4 expression with clinical outcomes may establish MUC4 as a potential molecular prognostic marker for these tumors.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Mucinas/metabolismo , Neoplasias de Oído, Nariz y Garganta/metabolismo , Neoplasias de las Glándulas Salivales/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/metabolismo , Western Blotting , Carcinoma de Células Escamosas/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Mucina 4 , Estadificación de Neoplasias , Neoplasias de Oído, Nariz y Garganta/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Neoplasias de las Glándulas Salivales/patología , Glándulas Salivales/metabolismo , Glándulas Salivales/patología
17.
J Cell Physiol ; 186(3): 397-405, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11169979

RESUMEN

Sialomucin complex (SMC, rat Muc4) is a heterodimeric glycoprotein composed of two subunits, the mucin component ASGP-1 and the transmembrane subunit ASGP-2. SMC/Muc4 is highly expressed on the surface of 13762 rat mammary adenocarcinoma cells at approximately 100 times the level found in the lactating mammary gland. Immunocytochemical staining of SMC/Muc4 in the developing rat mammary gland is localized to the apical membrane of the ductal epithelium. This staining pattern is similar to that for peanut lectin, a differentiation marker, which binds to cells expressing the disaccharide Thomsen-Friedenreich or TF antigen. Blotting of glycoproteins expressing the TF antigen from mammary tissues with peanut lectin detects a protein matching the migration of ASGP-2. Analysis of immunoprecipitated SMC/Muc4 by peanut lectin blotting shows that the TF antigen is abundantly present on the ASGP-2 subunit, hence the similarity of staining pattern with SMC/Muc4 antisera and peroxidase-conjugated lectin in mammary tissues. The TF antigen is also present on ASGP-2 of SMC/Muc4 produced by confluent cultures of Rama 37 rat mammary epithelial stem cells after their induction to an alveolar-like phenotype with prolactin. These results indicate that the TF antigen is present on the ASGP-2 transmembrane subunit of SMC/Muc4 from phenotypically normal tissues and cells, in contrast to malignant cells whose peanut lectin-binding disaccharide is located on ASGP-1.


Asunto(s)
Glándulas Mamarias Animales/citología , Mucinas/análisis , Aglutinina de Mani/química , Sialoglicoproteínas/análisis , Animales , Sitios de Unión , Membrana Celular/ultraestructura , Dimerización , Femenino , Inmunohistoquímica , Lactancia/fisiología , Glándulas Mamarias Animales/fisiología , Mucina 4 , Mucinas/química , Periodo Posparto/fisiología , Embarazo , Subunidades de Proteína , Ratas , Ratas Endogámicas F344
18.
Oncogene ; 19(49): 5568-73, 2000 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-11114736

RESUMEN

The four members of the ErbB family of receptor tyrosine kinases (RTKs) mediate a variety of cellular responses to epidermal growth factor (EGF)-like growth factors, and serve as a model for the generation of both diversity and specificity in RTK signaling. Previous studies indicate that receptor-receptor interactions figure prominently in signaling through ErbB receptors. In addition to a role in receptor kinase activation, ligand-induced ErbB receptor homo- and heterodimerization is thought to account for the diversity of biological responses stimulated by EGF-like growth factors. Since each receptor has the potential to couple to different complements of signaling pathways, EGF-like ligands specify cellular response by dictating which pairs of receptors become activated. More recently evidence has been uncovered for ligand discrimination by individual ErbB receptor dimers; receptors appear to realize which ligand is binding and differentially respond through autophosphorylation site usage. These observations indicate that ligand stimulation of RTKs is not generic, and point to another layer in the ErbB signal diversification mechanism. The mechanistic implications of ligand discrimination are discussed.


Asunto(s)
Receptor ErbB-2/metabolismo , Transducción de Señal , Animales , Ligandos , Fosforilación
19.
J Cell Physiol ; 185(2): 310-6, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11025453

RESUMEN

Blastocyst implantation is arguably the most critical stage of mammalian embryogenesis and requires that the uterus be in a receptive state. Initiation of receptivity involves loss of anti-adhesive molecules from the apical surface of the uterine luminal epithelium, one of which is sialomucin complex (SMC/Muc4), a highly O-glycosylated, anti-adhesive glycoprotein composed of mucin ascites sialoglycoprotein-1 (ASGP-1) and transmembrane (ASGP-2) subunits. SMC expression at the uterine luminal surface, but not in glandular epithelium, is hormonally regulated and varies with the estrous cycle. SMC is lost from the luminal uterine surface at the period of receptivity. However, the mechanism by which SMC is hormonally regulated is not understood. Analyses of SMC regulation in hormone-responsive primary cultures of rat uterine luminal epithelial cells (RULEC) demonstrated robust SMC expression by the RULEC, which is not altered by treatments with estrogen or progesterone. However, both SMC protein and transcript are downregulated by transforming growth factor-beta (TGF-beta1). SMC is also downregulated when RULEC are co-cultured with isolated uterine stromal cells. Estradiol and anti-TGF-beta block the stromal cell effect. These results suggest an indirect hormonal regulation of RULEC SMC, in which TGF-beta acts as a hormonally regulated, mesenchymal paracrine factor to repress SMC production by the epithelial cells and permit implantation.


Asunto(s)
Implantación del Embrión/fisiología , Mucinas/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Útero/metabolismo , Animales , Técnicas de Cocultivo , Regulación hacia Abajo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Estrógenos/farmacología , Femenino , Mucina 4 , Progesterona/farmacología , Ratas , Ratas Endogámicas F344 , Factor de Crecimiento Transformador beta/farmacología , Útero/citología , Útero/efectos de los fármacos
20.
Oncogene ; 19(38): 4354-61, 2000 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-10980611

RESUMEN

Sialomucin complex (SMC, rat Muc4) is a membrane mucin implicated in the protection of epithelia and the metastasis of some tumors. It is a heterodimeric complex, containing a mucin subunit with anti-adhesive activity and a transmembrane subunit with epidermal growth factor-like domains, one of which acts as an intramembrane ligand for ErbB2. Serum, insulin and insulin-like growth factor, but not epidermal growth factor, induce the expression of sialomucin complex in mammary epithelial cells. Induction correlates with sustained, but not transient, activation of extracellular-regulated protein kinase (ERK). MEK inhibitor U0126 blocked the induction, while activated MEK-1 transfected into a rat mammary adenocarcinoma cell line induced a sustained activation of ERK and up-regulated SMC/Muc4 expression. Northern and Western blotting indicated that up-regulation occurred concomitantly at the transcript and protein levels, both of which could be blocked by U0126. These results suggest that expression of SMC/Muc4 in mammary epithelial cells is regulated by selected growth factors through an ERK-dependent pathway at the transcript level.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mucinas/metabolismo , Animales , Butadienos/farmacología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Sustancias de Crecimiento/farmacología , Hidrocortisona/farmacología , Insulina/metabolismo , Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , MAP Quinasa Quinasa 1 , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Mucina 4 , Mucinas/efectos de los fármacos , Mucinas/genética , Nitrilos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Ratas Endogámicas F344 , Receptor ErbB-2/metabolismo , Sialomucinas , Transducción de Señal , Selenito de Sodio/farmacología , Transcripción Genética , Transferrina/farmacología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA