Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 289(9): 5499-509, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24403068

RESUMEN

Recently, it was discovered that serglycin, a hematopoietic cell proteoglycan, is the major proteoglycan expressed and constitutively secreted by multiple myeloma (MM) cells. High levels of serglycin are present in the bone marrow aspirates of at least 30% of newly diagnosed MM patients. However, its contribution to the pathophysiology of MM is unknown. Here, we show that serglycin knockdown (by ∼85% compared with normal levels), using lentiviral shRNA, dramatically attenuated MM tumor growth in mice with severe combined immunodeficiency. Tumors formed from cells deficient in serglycin exhibited diminished levels of hepatocyte growth factor expression and impaired development of blood vessels, indicating that serglycin may affect tumor angiogenesis. Furthermore, knockdown of serglycin significantly decreased MM cell adhesion to bone marrow stromal cells and collagen I. Even though serglycin proteoglycan does not have a transmembrane domain, flow cytometry showed that serglycin is present on the MM cell surface, and attachment to the cell surface is, at least in part, dependent on its chondroitin sulfate side chains. Co-precipitation of serglycin from conditioned medium of MM cells using a CD44-Fc chimera suggests that CD44 is the cell surface-binding partner for serglycin, which therefore may serve as a major ligand for CD44 at various stages during myeloma progression. Finally, we demonstrate that serglycin mRNA expression in MM cells is up-regulated by activin, a predominant cytokine among those increased in MM patients with osteolytic lesions. These studies provide direct evidence for a critical role for serglycin in MM pathogenesis and show that targeting serglycin may provide a novel therapeutic approach for MM.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple/metabolismo , Proteínas de Neoplasias/biosíntesis , Neovascularización Patológica/metabolismo , Proteoglicanos/biosíntesis , Proteínas de Transporte Vesicular/biosíntesis , Activinas/genética , Activinas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Adhesión Celular/genética , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Ratones , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Mieloma Múltiple/terapia , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Proteoglicanos/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Células del Estroma/metabolismo , Células del Estroma/patología , Proteínas de Transporte Vesicular/genética
2.
Int J Cancer ; 136(9): 2001-11, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24828886

RESUMEN

A number of proposals have been made in the past century regarding what may drive sporadic cancers to initiate and develop. Yet the problem remains largely unsolved as none of the proposals have been widely accepted as cancer-initiation drivers. We propose here a driver model for the initiation and early development of solid cancers associated with inflammation-induced chronic hypoxia and reactive oxygen species (ROS) accumulation. The model consists of five key elements: (i)human cells tend to have a substantial gap between ATP demand and supply during chronic hypoxia, which would inevitably lead to increased uptake of glucose and accumulation of its metabolites; (ii) the accumulation of these metabolites will cast mounting pressure on the cells and ultimately result in the production and export of hyaluronic acid; (iii) the exported hyaluronic acid will be degraded into fragments of various sizes, serving as tissue-repair signals, including signals for cell proliferation, cell survival and angiogenesis, which lead to the initial proliferation of the underlying cells; (iv) cell division provides an exit for the accumulated glucose metabolites using them towards macromolecular synthesis for the new cell, and hence alleviate the pressure from the metabolite accumulation; and (v) this process continues as long as the hypoxic condition persists. In tandem, genetic mutations may be selected to make cell divisions and hence survival more sustainable and efficient, also increasingly more uncontrollable. This model also applies to some hereditary cancers as their key mutations, such as BRCA for breast cancer, generally lead to increased ROS and ultimately to repression of mitochondrial activities and up-regulation of glycolysis, as well as hypoxia; hence the energy gap, glucose-metabolite accumulation, hyaluronic acid production and continuous cell division for survival.


Asunto(s)
Carcinogénesis/patología , Supervivencia Celular/fisiología , Hipoxia/patología , Neoplasias/metabolismo , Neoplasias/patología , Especies Reactivas de Oxígeno/metabolismo , Animales , Carcinogénesis/metabolismo , Proliferación Celular/fisiología , Humanos , Hipoxia/metabolismo
3.
J Biol Chem ; 288(36): 26089-26104, 2013 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-23888049

RESUMEN

The immunoglobulin superfamily glycoprotein CD147 (emmprin; basigin) is associated with an invasive phenotype in various types of cancers, including malignant breast cancer. We showed recently that up-regulation of CD147 in non-transformed, non-invasive breast epithelial cells is sufficient to induce an invasive phenotype characterized by membrane type-1 matrix metalloproteinase (MT1-MMP)-dependent invadopodia activity (Grass, G. D., Bratoeva, M., and Toole, B. P. (2012) Regulation of invadopodia formation and activity by CD147. J. Cell Sci. 125, 777-788). Here we found that CD147 induces breast epithelial cell invasiveness by promoting epidermal growth factor receptor (EGFR)-Ras-ERK signaling in a manner dependent on hyaluronan-CD44 interaction. Furthermore, CD147 promotes assembly of signaling complexes containing CD147, CD44, and EGFR in lipid raftlike domains. We also found that oncogenic Ras regulates CD147 expression, hyaluronan synthesis, and formation of CD147-CD44-EGFR complexes, thus forming a positive feedback loop that may amplify invasiveness. Last, we showed that malignant breast cancer cells are heterogeneous in their expression of surface-associated CD147 and that high levels of membrane CD147 correlate with cell surface EGFR and CD44 levels, activated EGFR and ERK1, and activated invadopodia. Future studies should evaluate CD147 as a potential therapeutic target and disease stratification marker in breast cancer.


Asunto(s)
Basigina/metabolismo , Neoplasias de la Mama/metabolismo , Receptores ErbB/metabolismo , Receptores de Hialuranos/metabolismo , Sistema de Señalización de MAP Quinasas , Complejos Multiproteicos/metabolismo , Proteínas de Neoplasias/metabolismo , Basigina/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Receptores ErbB/genética , Femenino , Humanos , Receptores de Hialuranos/genética , Microdominios de Membrana/genética , Microdominios de Membrana/metabolismo , Microdominios de Membrana/patología , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Complejos Multiproteicos/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas de Neoplasias/genética
4.
J Cell Sci ; 125(Pt 3): 777-88, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22389410

RESUMEN

A defining feature of malignant tumor progression is cellular penetration through the basement membrane and interstitial matrices that separate various cellular compartments. Accumulating evidence supports the notion that invasive cells employ specialized structures termed invadopodia to breach these structural barriers. Invadopodia are actin-based, lipid-raft-enriched membrane protrusions containing membrane-type-1 matrix metalloproteinase (MT1-MMP; also known as matrix metalloproteinase 14; MMP14) and several signaling proteins. CD147 (emmprin, basigin), an immunoglobulin superfamily protein that is associated with tumor invasion and metastasis, induces the synthesis of various matrix metalloproteinases in many systems. In this study we show that upregulation of CD147 is sufficient to induce MT1-MMP expression, invasiveness and formation of invadopodia-like structures in non-transformed, non-invasive, breast epithelial cells. We also demonstrate that CD147 and MT1-MMP are in close proximity within these invadopodia-like structures and co-fractionate in membrane compartments with the properties of lipid rafts. Moreover, manipulation of CD147 levels in invasive breast carcinoma cells causes corresponding changes in MT1-MMP expression, invasiveness and invadopodia formation and activity. These findings indicate that CD147 regulates invadopodia formation and activity, probably through assembly of MT1-MMP-containing complexes within lipid-raft domains of the invadopodia.


Asunto(s)
Basigina/metabolismo , Extensiones de la Superficie Celular/metabolismo , Invasividad Neoplásica/fisiopatología , Secuencia de Bases , Basigina/genética , Mama/citología , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Extensiones de la Superficie Celular/patología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Humanos , Metaloproteinasa 14 de la Matriz/metabolismo , Microdominios de Membrana/metabolismo , Microdominios de Membrana/patología , ARN Interferente Pequeño/genética , Regulación hacia Arriba
5.
J Virol ; 87(1): 621-35, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097457

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS), and KSHV activation of mitogen-activated protein kinases (MAPKs) initiates a number of key pathogenic determinants of KS. Direct inhibition of signal transduction as a therapeutic approach presents several challenges, and a better understanding of KSHV-induced mechanisms regulating MAPK activation may facilitate the development of new treatment or prevention strategies for KS. MAPK phosphatases, including dual-specificity phosphatase-1 (DUSP1), negatively regulate signal transduction and cytokine activation through MAPK dephosphorylation or interference with effector molecule binding to MAPKs, including the extracellular signal-regulated kinase (ERK). We found that ERK-dependent latent viral gene expression, the induction of promigratory factors, and cell invasiveness following de novo infection of primary human endothelial cells are in part dependent on KSHV suppression of DUSP1 expression during de novo infection. KSHV-encoded miR-K12-11 upregulates the expression of xCT (an amino acid transporter and KSHV fusion/entry receptor), and existing data indicate a role for xCT in the regulation of 14-3-3ß, a transcriptional repressor of DUSP1. We found that miR-K12-11 induces endothelial cell secretion of promigratory factors and cell invasiveness through upregulation of xCT-dependent, 14-3-3ß-mediated suppression of DUSP1. Finally, proof-of-principle experiments revealed that pharmacologic upregulation of DUSP1 inhibits the induction of promigratory factors and cell invasiveness during de novo KSHV infection. These data reveal an indirect role for miR-K12-11 in the regulation of DUSP1 and downstream pathogenesis.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/antagonistas & inhibidores , Herpesvirus Humano 8/patogenicidad , Interacciones Huésped-Patógeno , MicroARNs/metabolismo , Línea Celular , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , ARN Viral/metabolismo
6.
Am J Pathol ; 182(2): 577-85, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23178078

RESUMEN

CD147 (alias emmprin or basigin), an integral plasma membrane glycoprotein and a member of the Ig superfamily, is widespread in normal tissues, but highly up-regulated in many types of malignant cancer cells. CD147 is multifunctional, with numerous binding partners. Recent studies suggest that complexes of CD147 with the hyaluronan receptor CD44 and associated transporters and receptor tyrosine kinases are enriched in the plasma membrane of cancer stem-like cells. Here, we show that subpopulations of tumor cell lines constitutively expressing high levels of cell-surface CD147 exhibit cancer stem-like cell properties; that is, they exhibit much greater invasiveness, anchorage-independent growth, spheroid formation, and drug resistance in vitro and higher tumorigenicity in vivo than those constitutively expressing low levels of cell-surface CD147. Primary CD147-rich cell subpopulations derived from mouse mammary adenocarcinomas also exhibit high levels of invasiveness and spheroid-forming capacity, whereas CD147-low cells do not. Moreover, localization at the plasma membrane of CD44, the EGF receptor, the ABCB1 and ABCG2 drug transporters, and the MCT4 monocarboxylate transporter is elevated in cells constitutively expressing high levels of cell-surface CD147. These results show that CD147 is associated with assembly of numerous pro-oncogenic proteins in the plasma membrane and may play a fundamental role in properties characteristic of cancer stem-like cells.


Asunto(s)
Basigina/metabolismo , Resistencia a Antineoplásicos , Heterogeneidad Genética , Neoplasias/metabolismo , Neoplasias/patología , Animales , Adhesión Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/patología , Receptores ErbB/metabolismo , Femenino , Citometría de Flujo , Humanos , Receptores de Hialuranos/metabolismo , Neoplasias Mamarias Animales , Virus del Tumor Mamario del Ratón , Ratones , Invasividad Neoplásica , Esferoides Celulares/metabolismo , Esferoides Celulares/patología
7.
Int J Cancer ; 131(4): 834-43, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21918972

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS)-one of the most common tumors arising in the setting of immune suppression. Hallmarks of KS lesions include KSHV-infected cells of endothelial lineage and neoangiogenesis. Promigratory factors secreted in the tumor microenvironment by KSHV-infected cells promote endothelial cell (EC) migration and angiogenesis but existing therapies targeting these pathways are not widely utilized. This underscores the need for additional characterization of KSHV-host interactions relevant to EC pathogenesis to identify new therapeutic targets. We recently demonstrated that de novo infection by KSHV promotes EC invasion through upregulation of extracellular matrix metalloproteinase inducer (emmprin)-a multifunctional glycoprotein previously shown to induce tumor cell invasion and regional angiogenesis through upregulation of signal transduction and promotion of tumor-stroma interactions. This study was undertaken to determine whether EC invasion for KSHV-infected cells is induced through activation of specific signal transduction pathways and proangiogenic factors by emmprin. We found that KSHV activation of emmprin induces PI3K/Akt- and mitogen-activated protein kinase (MAPK)-dependent secretion of vascular endothelial growth factor (VEGF). Functionally, EC invasion following de novo infection is induced by emmprin-dependent PI3K/Akt and MAPK activation of VEGF. These findings support the potential utility of targeting emmprin for reducing VEGF secretion and EC migration in the KS microenvironment.


Asunto(s)
Basigina/fisiología , Endotelio Vascular/virología , Herpesvirus Humano 8/fisiología , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Línea Celular , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Activación Enzimática/fisiología , Ensayo de Inmunoadsorción Enzimática , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Development ; 136(16): 2825-35, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19633173

RESUMEN

The glycosaminoglycan hyaluronan (HA) is a structural component of extracellular matrices and also interacts with cell surface receptors to directly influence cell behavior. To explore functions of HA in limb skeletal development, we conditionally inactivated the gene for HA synthase 2, Has2, in limb bud mesoderm using mice that harbor a floxed allele of Has2 and mice carrying a limb mesoderm-specific Prx1-Cre transgene. The skeletal elements of Has2-deficient limbs are severely shortened, indicating that HA is essential for normal longitudinal growth of all limb skeletal elements. Proximal phalanges are duplicated in Has2 mutant limbs indicating an involvement of HA in patterning specific portions of the digits. The growth plates of Has2-deficient skeletal elements are severely abnormal and disorganized, with a decrease in the deposition of aggrecan in the matrix and a disruption in normal columnar cellular relationships. Furthermore, there is a striking reduction in the number of hypertrophic chondrocytes and in the expression domains of markers of hypertrophic differentiation in the mutant growth plates, indicating that HA is necessary for the normal progression of chondrocyte maturation. In addition, secondary ossification centers do not form in the central regions of Has2 mutant growth plates owing to a failure of hypertrophic differentiation. In addition to skeletal defects, the formation of synovial joint cavities is defective in Has2-deficient limbs. Taken together, our results demonstrate that HA has a crucial role in skeletal growth, patterning, chondrocyte maturation and synovial joint formation in the developing limb.


Asunto(s)
Desarrollo Óseo/fisiología , Condrocitos/fisiología , Extremidades , Silenciador del Gen , Glucuronosiltransferasa , Ácido Hialurónico/metabolismo , Articulaciones , Agrecanos/metabolismo , Animales , Tipificación del Cuerpo/fisiología , Proliferación Celular , Condrocitos/citología , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Extremidades/embriología , Extremidades/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Hialuronano Sintasas , Ácido Hialurónico/genética , Articulaciones/anomalías , Articulaciones/embriología , Articulaciones/crecimiento & desarrollo , Deformidades Congénitas de las Extremidades/genética , Mesodermo/fisiología , Ratones , Ratones Noqueados
9.
J Virol ; 85(7): 3596-606, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21270158

RESUMEN

The Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS), and the induction of an invasive cellular phenotype by KSHV following de novo infection is an important pathogenic component mediating tumor progression. The metastasis suppressor gene known as Nm23-H1 regulates tumor cell invasiveness, but whether KSHV itself regulates Nm23-H1 expression or subcellular localization, and whether this impacts cell invasiveness, has not been established. We found that KSHV increases expression and nuclear translocation of Nm23-H1 and that nuclear translocation of Nm23-H1 is regulated by the KSHV-encoded latency-associated nuclear antigen (LANA). Moreover, activation of the Ras-BRaf-MAPK (mitogen-activated protein kinase) signal transduction pathway, secretion of promigratory factors associated with this pathway, and cell invasiveness are dependent on KSHV regulation of Nm23-H1. Finally, induction of cytoplasmic overexpression of Nm23-H1 using a pharmacologic inhibitor of DNA methylation reduced KSHV-associated Ras-BRaf-MAPK pathway activation and suppressed KSHV-induced invasiveness. These data provide the first evidence for KSHV regulation of Nm23-H1 as a mechanism for KSHV induction of an invasive cellular phenotype and support the potential utility of targeting Nm23-H1 as a therapeutic approach for the treatment of KS.


Asunto(s)
Antígenos Virales/metabolismo , Regulación de la Expresión Génica , Herpesvirus Humano 8/patogenicidad , Interacciones Huésped-Patógeno , Nucleósido Difosfato Quinasas NM23/biosíntesis , Proteínas Nucleares/metabolismo , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Humanos
10.
BMC Cancer ; 11: 300, 2011 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-21762534

RESUMEN

BACKGROUND: Emmprin, a glycoprotein containing two Ig domains, is enriched on tumor cell surfaces and stimulates matrix metalloproteinase (MMP) production by adjacent stromal cells. Its first Ig domain (ECI) contains the biologically active site. The dependence of emmprin activity on N-glycosylation is controversial. We investigated whether synthetic ECI with the shortest sugar is functionally active. METHODS: The whole ECI peptides carrying sugar chains, a chitobiose unit or N-linked core pentasaccharide, were synthesized by the thioester method and added to fibroblasts to examine whether they stimulate MMP-2 production. RESULTS: ECI carrying a chitobiose unit, ECI-(GlcNAc) 2, but not ECI without a chitobiose unit or the chitobiose unit alone, dose-dependently stimulated MMP-2 production by fibroblasts. ECI with longer chitobiose units, ECI-[(Man)3(GlcNAc)2], also stimulated MMP-2 production, but the extent of its stimulation was lower than that of ECI-(GlcNAc)2. CONCLUSIONS: Our results indicate that ECI can mimic emmprin activity when substituted with chitobiose, the disaccharide with which N-glycosylation starts.


Asunto(s)
Basigina/farmacología , Disacáridos/química , Fibroblastos/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/biosíntesis , Fragmentos de Péptidos/farmacología , Procesamiento Proteico-Postraduccional , Acetilglucosamina/química , Adenocarcinoma/patología , Adenocarcinoma/secundario , Adulto , Basigina/química , Secuencia de Carbohidratos , Células Cultivadas , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/secundario , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Inducción Enzimática/efectos de los fármacos , Fascitis/patología , Fibroblastos/enzimología , Glicosilación , Humanos , Neoplasias Pulmonares , Mananos/química , Metaloproteinasa 2 de la Matriz/genética , Datos de Secuencia Molecular , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Estructura Terciaria de Proteína , Sarcoma/patología , Relación Estructura-Actividad , Células Tumorales Cultivadas
11.
Semin Cancer Biol ; 18(4): 244-50, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18534864

RESUMEN

Hyaluronan not only is an important structural component of extracellular matrices but also interacts instructively with cells during embryonic development, healing processes, inflammation, and cancer. It binds to several different types of cell surface receptors, including CD44, thus leading to co-regulation of important signaling pathways, notably those induced by activation of receptor tyrosine kinases. Consequently, interactions of both stromal and tumor cell-derived hyaluronan with tumor cells play important cooperative roles in several aspects of malignancy. This review focuses on cell autonomous hyaluronan-tumor cell interactions that lead to activation of receptor tyrosine kinases and enhanced drug resistance. Particular emphasis is placed on the role of hyaluronan-CD44 interactions in drug transporter expression and activity, especially in cancer stem-like cells that are highly malignant and resistant to chemotherapy. Antagonists of hyaluronan-CD44 interaction, especially small hyaluronan oligomers, may be useful in therapeutic strategies aimed at preventing tumor recurrence from these therapy-resistant sub-populations within malignant cancers.


Asunto(s)
Resistencia a Antineoplásicos/fisiología , Ácido Hialurónico/fisiología , Neoplasias/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Resistencia a Múltiples Medicamentos , Humanos , Receptores de Hialuranos/metabolismo , Neoplasias/tratamiento farmacológico , Transducción de Señal
12.
Anat Rec (Hoboken) ; 303(6): 1573-1583, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31090215

RESUMEN

CD147 (basigin; EMMPRIN), hyaluronan, and hyaluronan receptors (e.g., CD44) are intimately involved in several phenomena that underlie malignancy. A major avenue whereby they influence tumor progression is most likely their role in the characteristics of cancer stem cells (CSCs), subpopulations of tumor cells that exhibit chemoresistance, invasiveness, and potent tumorigenicity. Both CD147 and hyaluronan have been strongly implicated in chemoresistance and invasiveness, and may be drivers of CSC characteristics, since current evidence indicates that both are involved in epithelial-mesenchymal transition, a crucial process in the acquisition of CSC properties. Hyaluronan is a prominent constituent of the tumor microenvironment whose interactions with cell surface receptors influence several signaling pathways that lead to chemoresistance and invasiveness. CD147 is an integral plasma membrane glycoprotein of the Ig superfamily and cofactor in assembly and activity of monocarboxylate transporters (MCTs). CD147 stimulates hyaluronan synthesis and interaction of hyaluronan with its receptors, in particular CD44 and LYVE-1, which in turn result in activation of multiprotein complexes containing members of the membrane-type matrix metalloproteinase, receptor tyrosine kinase, ABC drug transporter, or MCT families within lipid raft domains. Multivalent hyaluronan-receptor interactions are essential for formation or stabilization of these lipid raft complexes and for downstream signaling pathways or transporter activities. We conclude that stimulation of hyaluronan-receptor interactions by CD147 and the consequent activities of these complexes may be critical to the properties of CSCs and their role in malignancy. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.


Asunto(s)
Basigina/metabolismo , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Membrana Celular/metabolismo , Humanos , Microdominios de Membrana/metabolismo
13.
J Clin Invest ; 116(11): 3026-34, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17080198

RESUMEN

PGE, a potent vasodilator, plays a primary role in maintaining the patency of the ductus arteriosus (DA). Genetic disruption of the PGE-specific receptor EP4, however, paradoxically results in fatal patent DA (PDA) in mice. Here we demonstrate that EP4-mediated signals promote DA closure by hyaluronic acid-mediated (HA-mediated) intimal cushion formation (ICF). Chronic EP4 stimulation by ONO-AE1-329, a selective EP4 agonist, significantly enhanced migration and HA production in rat DA smooth muscle cells. When HA production was inhibited, EP4-mediated migration was negated. Activation of EP4, adenylyl cyclase, and PKA all increased HA production and the level of HA synthase 2 (HAS2) transcripts. In immature rat DA explants, ICF was promoted by EP4/PKA stimuli. Furthermore, adenovirus-mediated Has2 gene transfer was sufficient to induce ICF in EP4-disrupted DA explants in which the intimal cushion had not formed. Accordingly, signals through EP4 have 2 essential roles in DA development, namely, vascular dilation and ICF. The latter would lead to luminal narrowing, helping adhesive occlusion and permanent closure of the vascular lumen. Our results imply that HA induction serves as an alternative therapeutic strategy for the treatment of PDA to the current one, i.e., inhibition of PGE signaling by cyclooxygenase inhibitors, which might delay PGE-mediated ICF in immature infants.


Asunto(s)
Conducto Arterial/embriología , Conducto Arterial/metabolismo , Ácido Hialurónico/metabolismo , Receptores de Prostaglandina E/metabolismo , Animales , Movimiento Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Conducto Arterial/citología , Regulación del Desarrollo de la Expresión Génica , Glucuronosiltransferasa/genética , Hialuronano Sintasas , Ratones , Ratones Noqueados , Músculo Liso/citología , Músculo Liso/embriología , Músculo Liso/metabolismo , Ratas , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E/deficiencia , Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E , Transducción de Señal , Técnicas de Cultivo de Tejidos , Transcripción Genética/genética
14.
Clin Cancer Res ; 14(6): 1804-13, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18347183

RESUMEN

PURPOSE: To determine if hyaluronan oligomers (o-HA) antagonize the malignant properties of glioma cells and treatment-resistant glioma side population (SP) cells in vitro and in vivo. EXPERIMENTAL DESIGN: A single intratumoral injection of o-HA was given to rats bearing spinal cord gliomas 7 days after engraftment of C6 glioma cells. At 14 days, spinal cords were evaluated for tumor size, invasive patterns, proliferation, apoptosis, activation of Akt, and BCRP expression. C6SP were isolated by fluorescence-activated cell sorting and tested for the effects of o-HA on BCRP expression, activation of Akt and epidermal growth factor receptor, drug resistance, and glioma growth in vivo. RESULTS: o-HA treatment decreased tumor cell proliferation, increased apoptosis, and down-regulated activation of Akt and the expression of BCRP. o-HA treatment of C6SP inhibited activation of epidermal growth factor receptor and Akt, decreased BCRP expression, and increased methotrexate cytotoxicity. In vivo, o-HA also suppressed the growth of gliomas that formed after engraftment of C6 or BCRP+ C6SP cells, although most C6SP cells lost their expression of BCRP when grown in vivo. Interestingly, the spinal cord gliomas contained many BCRP+ cells that were not C6 or C6SP cells but that expressed nestin and/or CD45; o-HA treatment significantly decreased the recruitment of these BCRP+ progenitor cells into the engrafted gliomas. CONCLUSIONS: o-HA suppress glioma growth in vivo by enhancing apoptosis, down-regulating key cell survival mechanisms, and possibly by decreasing recruitment of host-derived BCRP+ progenitor cells. Thus, o-HA hold promise as a new biological therapy to inhibit HA-mediated malignant mechanisms in glioma cells and treatment-resistant glioma stem cells.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Glioma/tratamiento farmacológico , Ácido Hialurónico/antagonistas & inhibidores , Ácido Hialurónico/uso terapéutico , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Médula Espinal/tratamiento farmacológico , Neoplasias de la Médula Espinal/metabolismo , Animales , Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Glioma/metabolismo , Receptores de Hialuranos/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Polímeros/uso terapéutico , Unión Proteica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Células Tumorales Cultivadas
15.
Drug Resist Updat ; 11(3): 110-21, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18490190

RESUMEN

Hyaluronan not only is an important structural component of extracellular matrices but also interacts with cells during dynamic cell processes such as those occurring in cancer. Consequently, interactions of hyaluronan with tumor cells play important cooperative roles in various aspects of malignancy. Hyaluronan binds to several cell surface receptors, including CD44, thus leading to co-regulation of signaling pathways that are important in regulation of multidrug resistance to anticancer drugs, in particular anti-apoptotic pathways induced by activation of receptor tyrosine kinases. Emmprin, a cell surface glycoprotein of the Ig superfamily, stimulates hyaluronan production and downstream signaling consequences. Emmprin and CD44 also interact with various multidrug transporters of the ABC family and monocarboxylate transporters associated with resistance to cancer therapies. Moreover, hyaluronan-CD44 interactions are critical to these properties in the highly malignant, chemotherapy-resistant cancer stem-like cells. Perturbations of the hyaluronan-CD44 interaction at the plasma membrane by various antagonists result in attenuation of receptor tyrosine kinase and transporter activities and inhibition of tumor progression in vivo. These antagonists, especially small hyaluronan oligomers, may be useful in therapeutic strategies aimed at preventing tumor refractoriness or recurrence due to drug-resistant sub-populations within malignant cancers.


Asunto(s)
Antineoplásicos/farmacología , Basigina/fisiología , Resistencia a Antineoplásicos/fisiología , Receptores de Hialuranos/fisiología , Ácido Hialurónico/fisiología , Neoplasias/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Apoptosis/fisiología , Membrana Celular/metabolismo , Supervivencia Celular/fisiología , Humanos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Neoplasias/genética , Fenotipo , Transducción de Señal
17.
Circulation ; 113(6): 834-41, 2006 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-16461815

RESUMEN

BACKGROUND: Matrix metalloproteinases (MMPs) are thought to promote progression of atherosclerosis and cardiovascular complications such as plaque rupture. It has been suggested that, on tumor cells, the extracellular MMP inducer (EMMPRIN) is involved in MMP synthesis by as yet unknown mechanisms. On cardiovascular cells, regulation of EMMPRIN in vivo or any functional relevance for MMP induction in vitro has not yet been studied. Thus, we studied EMMPRIN expression on monocytes in acute myocardial infarction (MI) and its potential relevance for MMP activation. METHODS AND RESULTS: In 20 patients with acute MI, surface expression of EMMPRIN was significantly enhanced on monocytes compared with in 20 patients with chronic stable angina. EMMPRIN upregulation was associated with increased expression of the membrane type 1 MMP (MT1-MMP) on monocytes (flow cytometry) as well as MMP-9 activity (gelatin zymography) in the plasma. At 6 months after successful revascularization, EMMPRIN, MT1-MMP, and MMP-9 had normalized. The secretion of MMP-9 by monocytes was induced by monocyte adhesion to immobilized recombinant EMMPRIN or to EMMPRIN-transfected Chinese hamster ovary cells. Moreover, adherent EMMPRIN-transfected monocytic cells stimulated MMP-2 activity of human vascular smooth muscle cells. Gene silencing of EMMPRIN by small-interfering RNA hindered lipopolysaccharide-induced monocyte secretion of MMP-9, indicating a predominant role of EMMPRIN in MMP-9 induction. CONCLUSIONS: EMMPRIN and MT1-MMP are upregulated on monocytes in acute MI. During cellular interactions, EMMPRIN stimulates MMP-9 in monocytes and MMP-2 in smooth muscle cells, indicating that EMMPRIN may display a key regulatory role for MMP activity in cardiovascular pathologies.


Asunto(s)
Basigina/genética , Metaloproteinasas de la Matriz/metabolismo , Monocitos/metabolismo , Infarto del Miocardio/etiología , Adulto , Anciano , Anciano de 80 o más Años , Aterosclerosis , Basigina/metabolismo , Basigina/fisiología , Adhesión Celular , Activación Enzimática , Proteínas de la Matriz Extracelular , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasas de la Matriz Asociadas a la Membrana , Persona de Mediana Edad , Músculo Liso Vascular/citología , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Regulación hacia Arriba
18.
Mech Ageing Dev ; 127(8): 660-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16650460

RESUMEN

Hutchinson-Gilford Progeria syndrome (HGPS) is a rare genetic disorder that displays features of segmental aging. It is manifested predominantly in connective tissue, with most prominent histological changes occurring in the skin, cartilage, bone and cardiovascular tissues. Detailed quantitative real time reverse-transcription polymerase chain reaction studies confirmed the previous observation that platelet-derived growth factor A-chain transcripts are consistently elevated 11+/-2- to 13+/-2-fold in two HGPS dermal fibroblast lines compared with age-matched controls. Furthermore, we identified two additional genes with substantially altered transcript levels. Nucleotide pyrophosphatase transcription was virtually shut down with decreased expression of 13+/-3- to 59+/-3-fold in HGPS, whereas aggrecan mRNA was elevated to 24+/-5 times to 41+/-4 times that of chronologically age-matched controls. Aggrecan, normally a component of cartilage and not always detectable in normal fibroblasts cultures, was secreted by HGPS fibroblast lines and was produced as a proteoglycan. This demonstrates that elevated aggrecan expression and its secretion are aberrant features of HGPS. We conclude that HGPS cells can display massively altered transcript levels leading to the secretion of inappropriate protein species.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/genética , Proteínas de la Matriz Extracelular/genética , Fibroblastos/fisiología , Lectinas Tipo C/genética , Progeria/fisiopatología , Piel/citología , Actinas/genética , Agrecanos , Línea Celular , Niño , Preescolar , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Análisis por Apareamiento , Factor de Crecimiento Derivado de Plaquetas/genética , Progeria/genética , Pirofosfatasas/genética , ARN Mensajero/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
19.
J Carcinog ; 5: 2, 2006 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-16401353

RESUMEN

BACKGROUND: The development and progression of human tumors is accompanied by various cellular, biochemical and genetic alterations. These events include tumor cells interaction with extracellular matrix molecules including hyaluronan (HA). Hyaluronan is a large polysaccharide associated with pericellular matrix of proliferating, migrating cells. Its implication in malignant transformation, tumor progression and with the degree of differentiation in various invasive tumors has well accepted. It has been well known the role HA receptors in tumor growth and metastasis in various cancer tissues. Previously we have observed the unified over expression of Hyaluronic Acid Binding Protein (HABP), H11B2C2 antigen by the tumor cells in various types progressing tumor tissues with different grades. However, the poor understanding of relation between HA and HA-binding protein expression on tumor cells during tumor progression as well as the asymmetric observations of the role of HA expression in tumor progression prompted us to examine the degree of HA expression on tumor cells vs. stroma in various types of human tumors with different grades. METHODS: In the present study clinically diagnosed tumor tissue samples of different grades were used to screen the histopathological expression of hyaluronan by using b-PG (biotinylated proteoglycan) as a probe and we compared the relative HA expression on tumor cells vs. stroma in well differentiated and poorly differentiated tumors. Specificity of the reaction was confirmed either by pre-digesting the tissue sections with hyaluronidase enzyme or by staining the sections with pre-absorbed complex of the probe and HA-oligomers. RESULTS: We show here the down regulation of HA expression in tumor cells is associated with progression of tumor from well differentiated through poorly differentiated stage, despite the constant HA expression in the tumor associated stroma. CONCLUSION: The present finding enlighten the relative roles of HA expression on tumor vs. stroma during the progression of tumors.

20.
Cancer Res ; 64(4): 1229-32, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14983875

RESUMEN

Emmprin (CD147; basigin) is a plasma membrane glycoprotein, enriched on the surface of many cancer cells, which induces matrix metalloproteinase synthesis via cell-cell interactions. Elevated emmprin production causes increased growth in vivo of human mammary carcinoma cells. In this study, we show that elevation of emmprin expression in less aggressive human carcinoma cells, which normally express low emmprin levels, induces the ability to grow under anchorage-independent conditions. We also found that elevated emmprin expression stimulates hyaluronan production and that the effect of emmprin on anchorage-independent growth is dependent on hyaluronan. Furthermore, emmprin stimulates cell survival pathway signaling in a hyaluronan-dependent manner. From these and other studies we conclude that emmprin enhances several malignant properties of cancer cells, including anchorage-independent growth, invasiveness, and chemoresistance.


Asunto(s)
Antígenos CD , Antígenos de Neoplasias , Neoplasias de la Mama/patología , Ácido Hialurónico/biosíntesis , Glicoproteínas de Membrana/fisiología , Basigina , Neoplasias de la Mama/metabolismo , División Celular , Línea Celular Tumoral , Supervivencia Celular , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA