Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
Stem Cells Dev ; 25(21): 1681-1690, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27554431

RESUMEN

Lefty is a member of transforming growth factor-beta (TGF-ß) superfamily and a potent antagonist of the TGF-ß/Nodal/Activin signaling pathway. Lefty is critical in sustaining self-renewal/pluripotency status, and implicated in the differentiation of embryonic stem cells (ESCs). However, emerging studies depict Lefty as a multifaceted protein involved in myriad cellular events. Lefty proteins (human Lefty A and B) are secreted glycoproteins, but their mode of secretion and the significance of their "glycan" moiety remain mostly unexplored. By employing an in vitro system of human ESCs (hESCs), we observed that Lefty protein(s) are encased in exosomes for extracellular release. The exosomal- and cell-associated Lefty diverge in their proteolytic processing, and possess N-glycan structures of high mannose and complex nature. Differentiation of hESCs to mesenchymal cells (MSCs) or neuronal progenitor cells (NPCs) entails distinct changes in the Lefty A/Lefty B gene(s), and protein expression. Specifically, the proteolytic cleavage and N-glycan composition of the cell-associated and exosomal Lefty differ in the differentiated progenies. These modifications affected Lefty's inhibitory effect on Nodal signaling in aggressive melanoma cells. The microheterogeneity in the processing and glycosylation of Lefty protein(s) between hESCs, MSCs, and NPCs could present efficient means of diversifying the endogenous functions of Lefty. Whether Lefty's diverse functions in embryonic patterning, as well as its diffusion range in the extracellular environment, are similarly affected remains to be determined. Our studies underscore the potential relevance of Lefty-packaged exosomes for combating debilitating diseases such as cancer.

3.
Oncotarget ; 6(33): 34071-86, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26460952

RESUMEN

Nodal is highly expressed in various human malignancies, thus supporting the rationale for exploring Nodal as a therapeutic target. Here, we describe the effects of a novel monoclonal antibody (mAb), 3D1, raised against human Nodal. In vitro treatment of C8161 human melanoma cells with 3D1 mAb shows reductions in anchorage-independent growth and vasculogenic network formation. 3D1 treated cells also show decreases of Nodal and downstream signaling molecules, P-Smad2 and P-ERK and of P-H3 and CyclinB1, with an increase in p27. Similar effects were previously reported in human breast cancer cells where Nodal expression was generally down-regulated; following 3D1 mAb treatment, both Nodal and P-H3 levels are reduced. Noteworthy is the reduced growth of human melanoma xenografts in Nude mice treated with 3D1 mAb, where immunostaining of representative tumor sections show diminished P-Smad2 expression. Similar effects both in vitro and in vivo were observed in 3D1 treated A375SM melanoma cells harboring the active BRAF(V600E) mutation compared to treatments with IgG control or a BRAF inhibitor, dabrafenib. Finally, we describe a 3D1-based ELISA for the detection of Nodal in serum samples from cancer patients. These data suggest the potential of 3D1 mAb for selecting and targeting Nodal expressing cancers.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Neoplasias de la Mama/patología , Melanoma/patología , Proteína Nodal/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Ciclina B1/biosíntesis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Ratones , Proteína Nodal/sangre , Proteína Nodal/inmunología , Oximas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Proteína Smad2/biosíntesis , Resonancia por Plasmón de Superficie
4.
Int J Cancer ; 136(5): E242-51, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25204799

RESUMEN

The significant role of the embryonic morphogen Nodal in maintaining the pluripotency of embryonic stem cells is well documented. Interestingly, the recent discovery of Nodal's re-expression in several aggressive and metastatic cancers has highlighted its critical role in self renewal and maintenance of the stem cell-like characteristics of tumor cells, such as melanoma. However, the key TGFß/Nodal signaling component(s) governing Nodal's effects in metastatic melanoma remain mostly unknown. By employing receptor profiling at the mRNA and protein level(s), we made the novel discovery that embryonic stem cells and metastatic melanoma cells share a similar repertoire of Type I serine/threonine kinase receptors, but diverge in their Type II receptor expression. Ligand:receptor crosslinking and native gel binding assays indicate that metastatic melanoma cells employ the heterodimeric TGFß receptor I/TGFß receptor II (TGFßRI/TGFßRII) for signal transduction, whereas embryonic stem cells use the Activin receptors I and II (ACTRI/ACTRII). This unexpected receptor usage by tumor cells was tested by: neutralizing antibody to block its function; and transfecting the dominant negative receptor to compete with the endogenous receptor for ligand binding. Furthermore, a direct biological role for TGFßRII was found to underlie vasculogenic mimicry (VM), an endothelial phenotype contributing to vascular perfusion and associated with the functional plasticity of aggressive melanoma. Collectively, these findings reveal the divergence in Nodal signaling between embryonic stem cells and metastatic melanoma that can impact new therapeutic strategies targeting the re-emergence of embryonic pathways.


Asunto(s)
Células Madre Embrionarias/metabolismo , Melanoma/metabolismo , Proteína Nodal/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Activinas/genética , Activinas/metabolismo , Western Blotting , Células Cultivadas , Células Madre Embrionarias/citología , Humanos , Melanoma/genética , Melanoma/patología , Proteína Nodal/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/secundario , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
5.
Mol Cancer Res ; 12(10): 1480-91, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25256709

RESUMEN

UNLABELLED: Patients with metastatic disease face high rates of mortality with a paucity of therapeutic options. Protein-based therapeutics provide advantages over traditional chemotherapy through increased specificity, decreased immune impairment, and more direct means of delivery. However, development is often hindered because of insufficient knowledge about protein processing by cells when exogenously applied. This study focuses on recombinant Maspin (rMaspin), a serine protease inhibitor (SERPINB5), which alters invasive properties when directly applied to cancer cells. Previous evidence suggests differences in the effects of rMaspin treatment when compared with endogenous reexpression, with little explanation for these discrepancies. A leading hypothesis is that exogenously applied rMaspin is subject to different regulatory and/or processing mechanisms in cancer cells when compared with endogenous expression. Therefore, a more detailed understanding of the mechanisms of internalization and subcellular trafficking of rMaspin is needed to guide future translational development. We describe the molecular trafficking of rMaspin in cytoplasmic vesicles of the endosomal/lysosomal pathway and characterize its uptake by multiple endocytic mechanisms. Time-lapse laser scanning confocal microscopy shows the uptake, in real time, of dye-labeled rMaspin in cancer cells. This study indicates that cellular processing of rMaspin plays a key role by affecting its biologic activity and highlights the need for new approaches aimed at increasing the availability of rMaspin when used to treat cancer. IMPLICATIONS: Novel characterization of internalization and subcellular trafficking of rMaspin provides new insights for future therapeutic development.


Asunto(s)
Endocitosis/efectos de los fármacos , Lisosomas/metabolismo , Serpinas/farmacología , Biomarcadores/metabolismo , Línea Celular Tumoral , Clatrina/metabolismo , Vesículas Citoplasmáticas/efectos de los fármacos , Vesículas Citoplasmáticas/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Lisosomas/efectos de los fármacos , Invasividad Neoplásica , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
6.
PLoS One ; 9(7): e103230, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25054204

RESUMEN

The post-lactational regression of mammary gland is a complex multi-step process designed to conserve the biological function of the gland for next pregnancy. This developmental stage is a biological intrigue with great relevance to breast cancer research, and thus has been the subject of intensive scrutiny. Multipronged studies (microarray, proteomics profiling, animal knock-out models) have provided a repertoire of genes critical to involution. However, the caveat of these approaches remains in their failure to reveal post-translational modification(s), an emerging and critical aspect of gene regulation in developmental processes and mammary gland remodeling. The massive surge in the lysosomal enzymes concurrent with the onset of involution has been known for decades, and considered essential for "clearance" purposes. However, functional significance of these enzymes in diverse biological processes distinct from their proteolytic activity is just emerging. Studies from our laboratory had indicated specific post-translational modifications of the aspartyl endopeptidase Cathepsin D (CatD) at distinct stages mammary gland development. This study addresses the biological significance of these modifications in the involution process, and reveals that post-translational modifications drive CatD into the nucleus to cleave Histone 3. The cleavage of Histone 3 has been associated with cellular differentiation and could be critical instigator of involution process. From functional perspective, deregulated expression and increased secretion of CatD are associated with aggressive and metastatic phenotype of breast cancer. Thus unraveling CatD's physiological functions in mammary gland development will bridge the present gap in understanding its pro-tumorigenic/metastatic functions, and assist in the generation of tailored therapeutic approaches.


Asunto(s)
Catepsina D/metabolismo , Histonas/metabolismo , Glándulas Mamarias Animales/fisiología , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Catepsina D/análisis , Femenino , Histonas/análisis , Lactancia , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Embarazo , Procesamiento Proteico-Postraduccional , Proteolisis
7.
Biol Med (Aligarh) ; 6(2)2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25663755

RESUMEN

Since its discovery as a lysosomal hydrolase, Cathepsin D (CatD) has been the subject of intensive scrutiny by numerous scientists. Those accumulated efforts have defined its biosynthetic pathway, structure, and companion proteins in the context of its perceived "house keeping" function. However, in the past two decades CatD has emerged as a multifunctional enzyme, involved in myriad biological processes beyond its original "housekeeping" role. CatD is responsible for selective and limited cleavage (quite distinct from non-specific protein degradation) of particular substrates vital to proper cellular function. These proteolytic events are critical in the control of biological processes, including cell cycle progression, differentiation and migration, morphogenesis and tissue remodeling, immunological processes, ovulation, fertilization, neuronal outgrowth, angiogenesis, and apoptosis. Consistent with the biological relevance of CatD, its deficiency, altered regulation or post-translational modification underlie important pathological conditions such as cancer, atherosclerosis, neurological and skin disorders. Specifically, deregulated synthesis, post-translational modifications and hyper-secretion of CatD, along with its mitogenic effects, are established hallmarks of cancer. More importantly, but less studied, is its significance in regulating the sensitivity to anticancer drugs. This review outlines CatD's post-translational modifications, cellular trafficking, secretion and protein binding partners in normal mammary gland, and restates the "site-specific" function of CatD which is most probably dictated by its post-translational modifications and binding partners. Noteworthy, CatD's association with one of its binding partners in the context of drug sensitivity is highlighted, with the optimism that it could contribute to the development of more effective chemotherapeutic agent(s) tailored for individual patients.

8.
Cancer Metastasis Rev ; 31(3-4): 529-51, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22752408

RESUMEN

Maspin, a non-inhibitory member of the serine protease inhibitor superfamily, has been characterized as a tumor suppressor gene in multiple cancer types. Among the established anti-tumor effects of Maspin are the inhibition of cancer cell invasion, attachment to extracellular matrices, increased sensitivity to apoptosis, and inhibition of angiogenesis. However, while significant experimental data support the role of Maspin as a tumor suppressor, clinical data regarding the prognostic implications of Maspin expression have led to conflicting results. This highlights the need for a better understanding of the context dependencies of Maspin in normal biology and how these are perturbed in the context of cancer. In this review, we outline the regulation and roles of Maspin in normal and developmental biology while discussing novel evidence and emerging theories related to its functions in cancer. We provide insight into the immense therapeutic potential of Maspin and the challenges related to its successful clinical translation.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Serpinas/fisiología , Serpinas/uso terapéutico , Animales , Apoptosis , Resistencia a Antineoplásicos , Epigénesis Genética , Humanos , Integrinas/fisiología , Neovascularización Fisiológica , Óxido Nítrico/fisiología , Unión Proteica , Proteínas Recombinantes/uso terapéutico , Serpinas/química , Serpinas/genética , Tamoxifeno/farmacología , Proteína p53 Supresora de Tumor/fisiología
9.
Cancer Biol Ther ; 10(5): 457-66, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20592493

RESUMEN

Cathepsin D (CatD) is a lysosomal aspartyl endopeptidase originally considered a "house keeping enzyme" involved in the clearance of unwanted proteins. However, recent studies have revealed CatD's involvement in apoptosis and autophagy, thus signifying an important function in the proper development and maintenance of multi-cellular organs. In the mammary gland, matrix degradation and the remodeling process are orchestrated by proteolytic enzymes, but the role of CatD at distinct developmental stages has remained mostly unexplored. Based on our previous studies we sought to address the role of this endopeptidase in mammary gland development and remodeling. By employing a mouse model, we report a previously unidentified participation of CatD in different stages of mammary gland development. Our findings reveal that CatD undergoes distinct protein processing at different stages of mammary gland development, and this customized processing results in differential enzymatic activity (constitutive and low pH activatable) best fitting particular stage(s) of development. In addition, at the onset of involution the N-glycan structure of this endopeptidase switches from a mixed high mannose and hybrid structure to an almost exclusively high mannose type, but reverts back to the original N-glycan composition by day 4 of involution. Our findings illuminate (at least in part) the "raison d'être" for CatD's numerous and highly regulated proteolytic processing steps from the pro-form to the mature enzyme. In the mammary gland, specific cleavage product(s) perform specialized function(s) befitting each stage of remodeling. It is noteworthy that deregulated synthesis, secretion and glycosylation of CatD are hallmarks of cancer progression. Thus, identifying the role of CatD in a dynamic normal tissue undergoing highly regulated cycles of remodeling could provide valuable information illuminating the deregulation of CatD associated with cancer development and metastasis.


Asunto(s)
Catepsina D/metabolismo , Glándulas Mamarias Animales/enzimología , Glándulas Mamarias Animales/fisiología , Aciltransferasas/genética , Aciltransferasas/metabolismo , Animales , Apoptosis , Autofagia , Membrana Basal/enzimología , Membrana Basal/fisiología , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Matriz Extracelular/enzimología , Matriz Extracelular/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Enzimológica de la Expresión Génica , Glicosilación , Humanos , Lactancia , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , N-Acetilglucosaminiltransferasas , Reacción en Cadena de la Polimerasa , Embarazo , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo
10.
Cancer Biol Ther ; 9(1): 23-30, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19923884

RESUMEN

Cathepsin D is a lysosomal hydrolase involved in intra- and extracellular proteolysis. This enzyme is aberrantly produced and processed in malignancy, and most notably is over-secreted into the tumor cell microenvironment. This hyper-secretion may lead to excessive degradation of the extracellular matrix, and contribute to tumor progression and metastases. These phenomena have been established in vitro, and there is evidence that Cathepsin D is similarly dysregulated in human breast cancer patients. Because breast cancer lacks an effective screening or surveillance biomarker, here we address the hypothesis that serum Cathepsin D activity may be useful to assess the presence or progression of breast cancer in females. While representative histologic sections from various disease-specific cohorts confirm previous findings that increased Cathepsin D production and secretion correlate with tumor progression, we report no difference in serum Cathepsin D activity between patients who are disease free, patients with pre-invasive or limited invasive disease, and patients with metastatic disease. Furthermore, in patients with known metastatic disease, there were no clinical variables associated with significantly different serum Cathepsin D activity. However, the immunohistochemical localization of Cathepsin D expression in histopathologic sections from breast cancer patients correlates with disease progression. Based on the serum results, and in contradistinction to Cathepsin D localization in breast cancer tissues, our findings support using Cathepsin D as a reliable histopathology biomarker for disease progression, but not for serum screening.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Catepsina D/metabolismo , Metástasis Linfática/patología , Neoplasias de la Mama/metabolismo , Estudios de Casos y Controles , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Femenino , Humanos , Lisosomas/enzimología , Persona de Mediana Edad
11.
Dev Growth Differ ; 51(5): 473-81, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19527266

RESUMEN

Interferon regulatory factor 6 (IRF6) is a non-canonical member of the interferon regulatory factor family of transcription factors. We recently identified IRF6 as a novel Maspin-interacting protein in mammary epithelial cells. Maspin is a tumor suppressor in the breast and has also been implicated in mammary gland morphogenesis. To explore a possible role for IRF6 in conjunction with Maspin during mammary gland growth and differentiation, we examined the expression of IRF6 and Maspin during post-utero mammary gland development using a combination of in vitro and in vivo approaches. The data revealed that the expression of IRF6 and Maspin is temporally and spatially regulated throughout mammary gland development, with maximal expression of both proteins occurring in fully differentiated, lactating lobuloalveolar cells. We further show that IRF6 adopts a lumenal localization pattern following complete epithelial cell polarization and present new evidence for the secretion of IRF6 into the milk. These results support the hypothesis that IRF6 and Maspin are important for mammary epithelial cell differentiation, and advance our understanding of the Maspin-IRF6 partnership during normal mammary gland development.


Asunto(s)
Factores Reguladores del Interferón/metabolismo , Serpinas/metabolismo , Animales , Western Blotting , Técnicas de Cultivo de Célula , Línea Celular , Células Epiteliales/metabolismo , Femenino , Humanos , Inmunohistoquímica , Glándulas Mamarias Humanas/metabolismo , Ratones
12.
J Cell Biochem ; 105(1): 208-18, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18494001

RESUMEN

In this study we examined the ability of interferon-gamma (IFN-gamma) to regulate mammary epithelial cell growth and gene expression, with particular emphasis on two genes: Maspin (a member of serine protease inhibitor superfamily), and the lysosomal aspartyl endopeptidase cathepsin D (CatD). The protein products of these genes are critically involved in regulation of multitude of biological functions in different stages of mammary tissue development and remodeling. In addition, the expression of Maspin is down-regulated in primary breast cancer and is lost in metastatic disease, while CatD is excessively produced and aberrantly secreted by breast cancer cells. We report that IFN-gamma receptors are expressed in mammary epithelial cells, and receptor engagement by IFN-gamma transduces the IFN-gamma signal via Stat-1 resulting in decreased vacuolar pH. This change in vacuolar pH alters CatD protein processing and secretion concurrent with increased Maspin secretion. In addition, IFN-gamma exerts a suppressive effect on cell growth and proliferation, and induces morphological changes in mammary epithelial cells. Our studies also reveal that breast cancer cells, which are devoid of Maspin, are refractory to IFN-gamma with respect to changes in vacuolar pH and CatD. However, Maspin transfection of breast cancer cells partially sensitizes the cells to IFN-gamma's effect, thus providing new therapeutic implications.


Asunto(s)
Autofagia/efectos de los fármacos , Catepsina D/metabolismo , Células Epiteliales/enzimología , Interferón gamma/farmacología , Glándulas Mamarias Humanas/efectos de los fármacos , Glándulas Mamarias Humanas/enzimología , Vacuolas/enzimología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Neoplasias de la Mama/patología , Línea Celular , Células Epiteliales/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Electrónica de Transmisión , Fenotipo , ARN Mensajero/genética , Vacuolas/efectos de los fármacos
13.
Mol Cell Biol ; 28(7): 2235-43, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18212048

RESUMEN

Interferon regulatory factor 6 (IRF6) is a novel and unique member of the IRF family of transcription factors. IRF6 has not been linked to the regulatory pathways or functions associated with other IRF family members, and the regulation and function of IRF6 remain unknown. We recently identified a protein interaction between IRF6 and the tumor suppressor maspin. To gain insight into the biological significance of the maspin-IRF6 interaction, we examined the regulation and function of IRF6 in relation to maspin in normal mammary epithelial cells. Our results demonstrate that in quiescent cells, IRF6 exists primarily in a nonphosphorylated state. However, cellular proliferation leads to rapid IRF6 phosphorylation, resulting in proteasome-dependent IRF6 degradation. These data are supported in situ by the increased expression of IRF6 in quiescent, differentiated lobuloalveolar cells of the lactating mammary gland compared to its expression in proliferating ductal and glandular epithelial cells during pregnancy. Furthermore, the reexpression of IRF6 in breast cancer cells results in cell cycle arrest, and the presence of maspin augments this response. These data support a model in which IRF6, in collaboration with maspin, promotes mammary epithelial cell differentiation by facilitating entry into the G(0) phase of the cell cycle.


Asunto(s)
Adenocarcinoma/patología , Neoplasias de la Mama/patología , Factores Reguladores del Interferón/fisiología , Glándulas Mamarias Animales/citología , Fase de Descanso del Ciclo Celular/fisiología , Adenocarcinoma/metabolismo , Animales , Neoplasias de la Mama/metabolismo , División Celular , Línea Celular Tumoral/citología , Línea Celular Tumoral/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Factores Reguladores del Interferón/genética , Lactancia/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación , Embarazo , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes de Fusión/fisiología , Serpinas/fisiología
14.
Cancer Res ; 67(8): 3535-9, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17440060

RESUMEN

Cellular interaction with the extracellular milieu plays a significant role in normal biological and pathologic processes. Excessive degradation of basement membrane matrix by proteolytic enzymes is a hallmark of tumor invasion and metastasis, and aspartyl proteinase cathepsin D is implicated as a major contributor to this process. Maspin, a non-inhibitory serpin, plays an important role in mammary gland development and remodeling. Expression of Maspin is decreased in primary tumors and lost in metastatic lesions. Maspin is mostly cytoplasmic and is partially secreted; however, the fate and function of secreted Maspin has remained mostly unexplored. We hypothesized that secreted Maspin is incorporated into the matrix deposited by normal mammary epithelial cells and thus could play a critical role in cathepsin D-mediated matrix degradation and remodeling of mammary tissue. In the absence of Maspin, as is the case with most cancer cells, matrix degradation proceeds unrestricted, thus facilitating the progression to metastasis. To test this, we employed an in vitro model where gels containing both types I and IV collagen were preconditioned with normal mammary epithelial cells to allow the incorporation of secreted Maspin. This conditioned matrix was used to examine cathepsin D-mediated collagen degradation by human breast cancer cell lines. Our results indicate that secretion of Maspin and its deposition into the extracellular milieu play an important role in matrix degradation. In this capacity, Maspin could potentially regulate mammary tissue remodeling occurring under normal and pathologic conditions. In addition, these findings could have a potential effect on future therapeutic intervention strategies for breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Catepsina D/antagonistas & inhibidores , Matriz Extracelular/metabolismo , Genes Supresores de Tumor/fisiología , Glándulas Mamarias Humanas/metabolismo , Serpinas/fisiología , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Catepsina D/genética , Catepsina D/metabolismo , Línea Celular , Línea Celular Tumoral , Colágeno/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Matriz Extracelular/enzimología , Humanos , Glándulas Mamarias Humanas/citología , Mutación , Serpinas/metabolismo , Relación Estructura-Actividad
15.
Clin Cancer Res ; 12(24): 7279-83, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17189399

RESUMEN

Maspin (mammary serine protease inhibitor) was identified in 1994 by subtractive hybridization analysis of normal mammary tissue and breast cancer cell lines. Subsequently, emerging evidence portrays maspin as a multifaceted protein, interacting with diverse group of intercellular and extracellular proteins, regulating cell adhesion, motility, apoptosis, and angiogenesis and critically involved in mammary gland development. The tissue-specific expression of maspin is epigenetically controlled, and aberrant methylation of maspin promoter is closely associated with maspin gene silencing. Identification of new tissue sites expressing maspin and novel maspin-binding partners has expanded the horizon for maspin research and promises maspin-based therapeutic approaches for combating cancer. This perspective briefly outlines the past and present strides in deciphering this unique molecule and speculates on new frontiers in maspin research and prospects of maspin as a diagnostic/prognostic indicator in cancer.


Asunto(s)
Genes Supresores de Tumor/fisiología , Serpinas/fisiología , Animales , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/análisis , Metilación de ADN , Regulación de la Expresión Génica , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Técnicas de Diagnóstico Molecular/métodos , Neoplasias/tratamiento farmacológico , Unión Proteica , Serpinas/análisis , Serpinas/metabolismo , Serpinas/uso terapéutico , Distribución Tisular
16.
J Cell Physiol ; 209(3): 617-24, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17001697

RESUMEN

Maspin (Mammary Serine Protease Inhibitor) was first reported in 1994 as a serpin with tumor suppressive properties. Maspin was initially isolated through subtractive hybridization and differential display analysis as a 42-kDa protein that is expressed in normal mammary epithelial cells but reduced or absent in breast carcinomas (Zou et al., 1994). Further research led to maspin's characterization as a class II tumor suppressor based on its ability to inhibit cell invasion, promote apoptosis, and inhibit angiogenesis (Sheng et al., 1996; Zhang et al., 2000b; Jiang et al., 2002). Since then, efforts have been made to characterize maspin's tumor suppressive mechanisms. In particular, researchers have studied maspin localization, the regulation of maspin expression, and more recently, maspin protein interactions. By elucidating these mechanisms, researchers are beginning to understand the complex, pleiotropic nature of maspin and the pathways through which maspin exerts its tumor suppressive properties. These new findings not only further enhance our understanding of cancer biology but also provide an avenue to develop maspin's potential as a diagnostic marker for cancer progression, and as a potentially powerful therapeutic agent in the fight against breast cancer.


Asunto(s)
Genes Supresores de Tumor , Neoplasias , Serpinas , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Factores Reguladores del Interferón/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica , Análisis de Secuencia por Matrices de Oligonucleótidos , Serpinas/genética , Serpinas/metabolismo , Serpinas/uso terapéutico , Distribución Tisular , Técnicas del Sistema de Dos Híbridos
17.
J Biol Chem ; 280(40): 34210-7, 2005 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-16049006

RESUMEN

Since its reported discovery in 1994, maspin (mammary serine protease inhibitor) has been characterized as a class II tumor suppressor by its ability to promote apoptosis and inhibit cell invasion. Maspin is highly expressed in normal mammary epithelial cells but reduced or absent in aggressive breast carcinomas. However, despite efforts to characterize the mechanism(s) by which maspin functions as a tumor suppressor, its molecular characterization has remained somewhat elusive. Therefore, in an attempt to identify maspin-interacting proteins and thereby gain insight into the functional pathways of maspin, we employed a maspin-baited yeast two-hybrid system and subsequently identified Interferon Regulatory Factor 6 (IRF6) as a maspin-binding protein. IRF6 belongs to the IRF family of transcription factors, which is best known for its regulation of interferon and interferon-inducible genes following a pathogenic stimulus. Although many of the IRF family members have been well characterized, IRF6 remains poorly understood. We report that IRF6 is expressed in normal mammary epithelial cells and that it directly associates with maspin in a yeast two-hybrid system and in vitro. The interaction occurs via the conserved IRF protein association domain and is regulated by phosphorylation of IRF6. We have shown that, similar to maspin, IRF6 expression is inversely correlated with breast cancer invasiveness. We further demonstrated that the transient re-expression of IRF6 in breast cancer cells results in an increase of N-cadherin and a redistribution of vimentin commensurate with changes in cell morphology, suggestive of an epithelial-to-mesenchymal transition event. Concomitantly, we showed that maspin acts as a negative regulator of this process. These findings help to elucidate the molecular mechanisms of maspin and suggest an interactive role between maspin and IRF6 in regulating cellular phenotype, the loss of which can lead to neoplastic transformation.


Asunto(s)
Neoplasias de la Mama/patología , Mama/fisiología , Genes Supresores de Tumor/fisiología , Factores Reguladores del Interferón/metabolismo , Inhibidores de Serina Proteinasa/fisiología , Serpinas/fisiología , Secuencia de Aminoácidos , Mama/citología , Cadherinas/biosíntesis , Transformación Celular Neoplásica , Células Epiteliales/fisiología , Femenino , Perfilación de la Expresión Génica , Humanos , Factores Reguladores del Interferón/biosíntesis , Datos de Secuencia Molecular , Fenotipo , Fosforilación , Reacción en Cadena de la Polimerasa , Células Tumorales Cultivadas , Técnicas del Sistema de Dos Híbridos , Vimentina/metabolismo , Levaduras/genética
18.
Cancer Biol Ther ; 4(4): 400-6, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15846059

RESUMEN

Maspin, a unique serine proteinase inhibitor (serpin), plays a key role in mammary gland development and is silenced during breast cancer progression. Maspin has been shown to inhibit tumor cell motility and invasion in cell culture, as well as growth and metastasis in animal models. In this study, we investigated the effect of maspin on the regulation of hypoxia-induced expression of urokinase-type plasminogen activator (uPA) and its receptor (uPAR), with respect to invasive potential in metastatic breast cells MDA-MB-231. We hypothesized that maspin can neutralize or mitigate hypoxia-induced expression of uPA/uPAR in metastatic breast cancer cells, resulting in suppression of their invasive potential. To test our hypothesis, we employed the highly invasive MDA-MB-231 breast cancer cells that are devoid of maspin, and transfected them with the maspin gene, and then determined the effect of hypoxia on uPA/uPAR expression. Normal mammary epithelial cells 1436N1 were used as a control. Our findings demonstrate that maspin downregulated the basal and hypoxia-induced uPA/uPAR expression and reduced the stimulatory effect of hypoxia on the in vitro invasive ability of MDA-MB-231-cells. In addition, maspin also inhibited the enzymatic activity of secreted and cell associated uPA in MDA-MB-231 cells. These results indicate that maspin inhibits hypoxia-induced invasion of metastatic breast cancer cells by blocking the uPA system, thus illuminating an important molecular pathway for therapeutic consideration.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Receptores de Superficie Celular/antagonistas & inhibidores , Serpinas/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Antineoplásicos/metabolismo , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Femenino , Genes Supresores de Tumor , Humanos , Modelos Biológicos , Invasividad Neoplásica , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Serpinas/metabolismo , Factores de Tiempo
19.
J Cell Biochem ; 91(4): 649-61, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14991757

RESUMEN

The role(s) of E-cadherin in tumor progression, invasion, and metastasis remains somewhat enigmatic. In order to investigate various aspects of E-cadherin biological activity, particularly in prostate cancer progression, our laboratory cloned unique subpopulations of the heterogeneous DU145 human prostatic carcinoma cell line and characterized their distinct biological functions. The data revealed that the highly invasive, fibroblastic-like subpopulation of DU145 cells (designated DU145-F) expressed less than 0.1-fold of E-cadherin protein when compared to the parental DU145 or the poorly invasive DU145 cells (designated DU145-E). Experimental disruption of E-cadherin function stimulated migration and invasion of DU145-E and other E-cadherin-positive prostate cancer cell lines, but did not affect the fibroblastic-like DU145-F subpopulation. Within the medium of parental DU145 cells, the presence of an 80 kDa E-cadherin fragment was detected. Subsequent functional analyses revealed the stimulatory effect of this fragment on the migratory and invasive capability of E-cadherin-positive cells. These results suggest that E-cadherin plays an important role in regulating the invasive potential of prostate cancer cells through an unique paracrine mechanism.


Asunto(s)
Cadherinas/metabolismo , Invasividad Neoplásica , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Anticuerpos/inmunología , Anticuerpos/farmacología , Cadherinas/inmunología , Cadherinas/farmacología , Cateninas , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Proteínas del Citoesqueleto/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/farmacología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Fosfoproteínas/metabolismo , Neoplasias de la Próstata/enzimología , Transactivadores/metabolismo , beta Catenina , Catenina delta
20.
Clin Cancer Res ; 10(2): 449-54, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14760064

RESUMEN

PURPOSE: Mammary epithelial cells and the majority of breast cancer tumors require estrogen for continued growth. Antiestrogen therapy alone, or in combination with other drugs, has long been a common procedure for breast cancer treatment and prophylaxis. Thus, there is a critical need to elucidate the mechanism(s) of action of antiestrogen treatment, especially for patients who are at risk of breast cancer development or who are currently receiving hormone therapy. In this study, we examined the ability of hormones to regulate the expression of a tumor suppressor gene, maspin, which is a serine protease inhibitor (serpin) that plays an important role in mammary gland development and is silenced during breast cancer progression. Specifically, our hypothesis tested the clinical efficacy of tamoxifen to regulate maspin expression. EXPERIMENTAL DESIGN: We used maspin promoter luciferase reporter plasmids that were transfected into normal human mammary epithelial (HMEC1331) and MCF-7 breast cancer cells, followed by determination of the effect of hormones and their antagonists on maspin promoter activity. At the protein level, cytosolic fractions from both cell types before and after hormone treatment were subjected to Western blot analysis to determine maspin level. RESULTS AND CONCLUSIONS: Our studies revealed that the antiestrogen tamoxifen induces maspin promoter activity. Interestingly, antiandrogen flutamide could also induce maspin in both cell lines tested. These observations were further confirmed in patient tissues. These novel findings provide a new mechanism of action for tamoxifen under normal and pathological conditions. More significantly, these findings could have a potential impact on future therapeutic intervention strategies for breast cancer.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Biosíntesis de Proteínas , Proteínas , Serpinas/biosíntesis , Tamoxifeno/farmacología , Actinas/metabolismo , Adulto , Western Blotting , Línea Celular Tumoral , Citoplasma/metabolismo , Citosol/metabolismo , Electroforesis en Gel de Poliacrilamida , Flutamida/farmacología , Genes Supresores de Tumor , Hormonas/metabolismo , Humanos , Inmunohistoquímica , Luciferasas/metabolismo , Persona de Mediana Edad , Modelos Genéticos , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...