Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Med (Lausanne) ; 9: 879500, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572966

RESUMEN

Basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (cSCC), referred to as keratinocyte carcinomas, are skin cancer with the highest incidence. BCCs, rarely metastasize; whereas, though generally not characterized by high lethality, approximately 2-4% of primary cSCCs metastasize with patients exhibiting poor prognosis. The extracellular matrix (ECM) serves as a scaffold that provides structural and biological support to cells in all human tissues. The main components of the ECM, including fibrillar proteins, proteoglycans (PGs), glycosaminoglycans (GAGs), and adhesion proteins such as fibronectin, are secreted by the cells in a tissue-specific manner, critical for the proper function of each organ. The skin compartmentalization to the epidermis and dermis compartments is based on a basement membrane (BM), a highly specialized network of ECM proteins that separate and unify the two compartments. The stiffness and assembly of BM and tensile forces affect tumor progenitors' invasion at the stratified epithelium's stromal border. Likewise, the mechanical properties of the stroma, e.g., stiffness, are directly correlated to the pathogenesis of the keratinocyte carcinomas. Since the ECM is a pool for various growth factors, cytokines, and chemokines, its' intense remodeling in the aberrant cancer tissue milieu affects biological functions, such as angiogenesis, adhesion, proliferation, or cell motility by regulating specific signaling pathways. This review discusses the structural and functional modulations of the keratinocyte carcinoma microenvironment. Furthermore, we debate how ECM remodeling affects the pathogenesis of these skin cancers.

2.
Cancers (Basel) ; 14(5)2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35267503

RESUMEN

Osteosarcoma (OS) is a mesenchymally derived, aggressive bone cancer. OS cells produce an aberrant nonmineralized or partly mineralized extracellular matrix (ECM) whose components participate in signaling pathways connected to specific pathogenic phenotypes of this bone cancer. The expression of biglycan (BGN), a secreted small leucine-rich proteoglycan (SLRP), is correlated to aggressive OS phenotype and resistance to chemotherapy. A constitutive signaling of IGF-IR signaling input in sarcoma progression has been established. Here, we show that biglycan activates the IGF-IR signaling pathway to promote MG63 biglycan-secreting OS cell growth by forming a complex with the receptor. Computational models of IGF-IR and biglycan docking suggest that biglycan binds IGF-IR dimer via its concave surface. Our binding free energy calculations indicate the formation of a stable complex. Biglycan binding results in prolonged IGF-IR activation leading to protracted IGF-IR-dependent cell growth response of the poorly-differentiated MG63 cells. Moreover, biglycan facilitates the internalization (p ≤ 0.01, p ≤ 0.001) and sumoylation-enhanced nuclear translocation of IGF-IR (p ≤ 0.05) and its DNA binding in MG63 cells (p ≤ 0.001). The tyrosine kinase activity of the receptor mediates this mechanism. Furthermore, biglycan downregulates the expression of the tumor-suppressor gene, PTEN (p ≤ 0.01), and increases the expression of endothelial-mesenchymal transition (EMT) and aggressiveness markers vimentin (p ≤ 0.01) and fibronectin (p ≤ 0.01) in MG63 cells. Interestingly, this mechanism is not valid in moderately and well-differentiated, biglycan non-expressing U-2OS and Saos-2 OS cells. Furthermore, biglycan exhibits protective effects against the chemotherapeutic drug, doxorubicin, in MG63 OS cells (p ≤ 0.01). In conclusion, these data indicate a potential direct and adjunct therapeutical role of biglycan in osteosarcoma.

3.
Biomolecules ; 11(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34572532

RESUMEN

Carcinogenesis is a multifactorial process with the input and interactions of environmental, genetic, and metabolic factors. During cancer development, a significant remodeling of the extracellular matrix (ECM) is evident. Proteoglycans (PGs), such as lumican, are glycosylated proteins that participate in the formation of the ECM and are established biological mediators. Notably, lumican is involved in cellular processes associated with tumorigeneses, such as EMT (epithelial-to-mesenchymal transition), cellular proliferation, migration, invasion, and adhesion. Furthermore, lumican is expressed in various cancer tissues and is reported to have a positive or negative correlation with tumor progression. This review focuses on significant advances achieved regardingthe role of lumican in the tumor biology. Here, the effects of lumican on cancer cell growth, invasion, motility, and metastasis are discussed, as well as the repercussions on autophagy and apoptosis. Finally, in light of the available data, novel roles for lumican as a cancer prognosis marker, chemoresistance regulator, and cancer therapy target are proposed.


Asunto(s)
Carcinogénesis/metabolismo , Lumican/metabolismo , Animales , Apoptosis , Autofagia , Carcinogénesis/patología , Matriz Extracelular/metabolismo , Humanos , Lumican/química , Modelos Biológicos
4.
Cancers (Basel) ; 13(10)2021 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-34069554

RESUMEN

Bone sarcomas, mesenchymal origin tumors, represent a substantial group of varying neoplasms of a distinct entity. Bone sarcoma patients show a limited response or do not respond to chemotherapy. Notably, developing efficient chemotherapy approaches, dealing with chemoresistance, and preventing metastasis pose unmet challenges in sarcoma therapy. Insulin-like growth factors 1 and 2 (IGF-1 and -2) and their respective receptors are a multifactorial system that significantly contributes to bone sarcoma pathogenesis. Whereas failures have been registered in creating novel targeted therapeutics aiming at the IGF pathway, new agent development should continue, evaluating combinatorial strategies for enhancing antitumor responses and better classifying the patients that could best benefit from these therapies. A plausible approach for developing a combinatorial strategy is to focus on the tumor microenvironment (TME) and processes executed therein. Herewith, we will discuss how the interplay between IGF-signaling and the TME constituents affects sarcomas' basal functions and their response to therapy. This review highlights key studies focusing on IGF signaling in bone sarcomas, specifically studies underscoring novel properties that make this system an attractive therapeutic target and identifies new relationships that may be exploited. Potential direct and adjunct therapeutical implications of the extracellular matrix (ECM) effectors will also be summarized.

5.
Biomolecules ; 11(3)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33800172

RESUMEN

The tumor microenvironment (TME) is composed of cancerous, non-cancerous, stromal, and immune cells that are surrounded by the components of the extracellular matrix (ECM). Glycosaminoglycans (GAGs), natural biomacromolecules, essential ECM, and cell membrane components are extensively altered in cancer tissues. During disease progression, the GAG fine structure changes in a manner associated with disease evolution. Thus, changes in the GAG sulfation pattern are immediately correlated to malignant transformation. Their molecular weight, distribution, composition, and fine modifications, including sulfation, exhibit distinct alterations during cancer development. GAGs and GAG-based molecules, due to their unique properties, are suggested as promising effectors for anticancer therapy. Considering their participation in tumorigenesis, their utilization in drug development has been the focus of both industry and academic research efforts. These efforts have been developing in two main directions; (i) utilizing GAGs as targets of therapeutic strategies and (ii) employing GAGs specificity and excellent physicochemical properties for targeted delivery of cancer therapeutics. This review will comprehensively discuss recent developments and the broad potential of GAG utilization for cancer therapy.


Asunto(s)
Glicosaminoglicanos/metabolismo , Animales , Protocolos Antineoplásicos , Sulfatos de Condroitina/química , Sulfatos de Condroitina/metabolismo , Matriz Extracelular/metabolismo , Heparina/metabolismo , Humanos , Ácido Hialurónico/metabolismo , Nanoestructuras/química
6.
Mol Pharm ; 17(11): 4212-4225, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32986447

RESUMEN

Nanoparticles (NPs) produced from amphiphilic derivatives of poly-N-vinylpyrrolidone (Amph-PVP), composed of various molecular weight polymeric hydrophilic fragments linked into hydrophobic n-alkyl chains of varying lengths, were previously shown to exert excellent biocompatibility. Although routes of administration can be different, finally, most nanosystems enter the blood circulation or lymphatic vessels, and by this, they establish direct contact with endothelial cells. In this study, Amph-PVP NPs and fluorescently labeled Amph-PVP-based NPs, namely "PVP" NPs (Amph-PVP-NPs (6000 Da) unloaded) and "F"-NPs (Amph-PVP-NPs (6000 Da) loaded with fluorescent FITC), were synthesized to study Amph-PVP NPs interactions with HMEC-1 endothelial cells. PVP NPs were readily uptaken by HMEC-1 cells in a concentration-dependent manner, as demonstrated by immunofluorescence imaging. Upon uptake, the FITC dye was localized to the perinuclear region and cytoplasm of treated cells. The generation of lipopolysaccharide (LPS)-induced activated endothelium model revealed an increased uptake of PVPNPs, as shown by confocal microscopy. Both unloaded PVP NPs and F-NPs did not affect EC viability in the 0.01 to 0.066 mg/mL range. Furthermore, we focused on the potential immunological activation of HMEC-1 endothelial cells upon PVPNPs treatment by assessing the expression of their E-Selectin, ICAM-1, and VCAM-1 adhesion receptors. None of the adhesion molecules were affected by NP treatments of both activated by LPS and nonactivated HMEC-1 cells, at the utilized concentrations (p = NS). In this study, PVP (6000 Da) NPs were used to encapsulate indomethacin, a widely used anti-inflammatory drug. The synthesized drug carrier complex did not affect HMEC-1 cell growth and expression of E-selectin, ICAM-1, and VCAM-1 adhesion receptors. In summary, PVP-based NPs are safe for use on both basal and activated endothelium, which more accurately mimics pathological conditions. Amph-PVP NPs are a promising drug delivery system.


Asunto(s)
Antiinflamatorios/administración & dosificación , Materiales Biocompatibles/química , Portadores de Fármacos/química , Células Endoteliales/efectos de los fármacos , Indometacina/administración & dosificación , Nanopartículas/química , Polímeros/química , Pirrolidinonas/química , Antiinflamatorios/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Fluoresceína-5-Isotiocianato/administración & dosificación , Colorantes Fluorescentes/administración & dosificación , Colorantes Fluorescentes/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Indometacina/metabolismo , Peso Molecular , Tamaño de la Partícula
7.
Int J Oncol ; 57(3): 791-803, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32705211

RESUMEN

Chondrosarcoma is a malignant bone tumor characterized by the production of a modified cartilage­type extracellular matrix (ECM). In the present study, the expression levels of the small leucine­rich proteoglycans (SLRPs), decorin, biglycan and lumican, were examined in the HTB94 human chondrosarcoma cell line. HTB94 cells were found to express and secrete the 3 SLRP members. RT­qPCR and western blot analysis demonstrated that lumican was the most abundantly secreted SLRP, whereas decorin and biglycan expression levels were low. The utilization of short interfering RNA specific for the decorin, biglycan, and lumican genes resulted in the efficient downregulation of the respective mRNA levels (P≤0.001). The growth of the HTB94 cells was stimulated by lumican (P≤0.001), whereas their migration and adhesion were not affected (P=NS). By contrast, these cellular functions were not sensitive to a decrease in low endogenous levels of decorin and biglycan. Lumicandeficiency significantly inhibited both basal and insulin­like growth factor I (IGF­I)­induced HTB94 cell growth (P≤0.001 andP≤0.01, respectively). These effects were executed through the insulin­like growth factor I receptor (IGF­IR), whose activation was markedly attenuated (P≤0.01) in lumican­deficient HTB94 cells. The downregulation of lumican induced the substantial inhibition of extracellular regulated kinase (ERK1/2) activation (P≤ 0.01), indicating that ERK1/2 is a necessary component of lumican/IGF­IR­mediated HTB94 cell proliferation. Moreover, the lumican­deficient cells exhibit increased mRNA levels of p53 (P≤0.05), suggesting that lumican facilitates HTB94 cell growth through an IGF­IR/ERK1/2/p53 signaling cascade. On the whole, the findings of the present study demonstrate that endogenous lumican is a novel regulator of HTB94 cell growth.


Asunto(s)
Neoplasias Óseas/patología , Condrosarcoma/patología , Lumican/metabolismo , Línea Celular Tumoral , Proliferación Celular , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lumican/genética , Sistema de Señalización de MAP Quinasas , ARN Interferente Pequeño/metabolismo , Receptor IGF Tipo 1/metabolismo
8.
Toxicol Appl Pharmacol ; 377: 114632, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31226360

RESUMEN

Allergic contact dermatitis (ACD) is caused by topical exposure to chemical allergens. Keratinocytes play a key role in innate immunity, as well as in ACD progression. The transmembrane Toll-like receptor 4 (TLR4), strongly implicated in skin inflammation, has the ability to bind Damage Associated Molecular Patterns (DAMPs), like Low Molecular Weight Hyaluronan (LMWHA). Previously, we had determined that p-phenylenediamine (PPD) and 2,4-dinitrochlorobenzene (DNCB) modulate keratinocyte HA deposition in a manner correlated to their sensitization. In the present study, we aimed to investigate putative co-operation of HA and TLR4 in the process of PPD and DNCB-induced keratinocyte activation. Contact sensitizers were shown to significantly increase the expression of Hyaluronan Synthases (HAS) and TLR4 in NCTC2544 human keratinocytes, as demonstrated by western blot and Real-Time PCR. These data, in correlation to earlier shown enhanced HA degradation suggest that the contact sensitizers facilitate HA turnover of keratinocytes and increase the release of pro-inflammatory, LMWHA fragments. Treatment with exogenous LMWHA enhanced TLR4, HAS levels and Nuclear factor-kappa beta (NF-κΒ) activation. PPD, DNCB and LMWHA-effects were shown to be partly executed through TLR4 downstream signaling as shown by Real-Time, western blot, siRNA and confocal microscopy approaches. Specifically, PPD and DNCB stimulated the activation of the TLR4 downstream mediator NF-κB. Therefore, the shown upregulation of TLR4 expression is suggested to further facilitate the release of endogenous, bioactive HA fragments and sustain keratinocyte activation. In conclusion, keratinocyte contact allergen-dependent sensitization is partly mediated through a LMWHA/TLR4/ NF-κB signaling axis.


Asunto(s)
Alérgenos/toxicidad , Dermatitis Alérgica por Contacto/patología , Ácido Hialurónico/metabolismo , Queratinocitos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/efectos de los fármacos , Línea Celular , Dinitroclorobenceno/toxicidad , Humanos , Hialuronano Sintasas/metabolismo , Ácido Hialurónico/química , Irritantes/toxicidad , Peso Molecular , FN-kappa B/biosíntesis , FN-kappa B/genética , Fenilendiaminas/toxicidad , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética
9.
Front Oncol ; 8: 470, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30406034

RESUMEN

Biglycan, a small leucine rich proteoglycan (SLRP), is an important participant in bone homeostasis and development as well as in bone pathology. In the present study biglycan was identified as a positive regulator of MG63 osteosarcoma cell growth (p ≤ 0.001). IGF-I was shown to increase biglycan expression (p ≤ 0.01), whereas biglycan-deficiency attenuated significantly both basal and IGF-I induced cell proliferation of MG63 cells (p ≤ 0.001; p ≤ 0.01, respectively). These effects were executed through the IGF-IR receptor whose activation was strongly attenuated (p ≤ 0.01) in biglycan-deficient MG63 cells. Biglycan, previously shown to regulate Wnt/ß-catenin pathway, was demonstrated to induce a significant increase in ß-catenin protein expression evident at cytoplasmic (p ≤ 0.01), membrane (p ≤ 0.01), and nucleus fractions in MG63 cells (p ≤ 0.05). As demonstrated by immunofluorescence, increase in ß-catenin expression is attributed to co-localization of biglycan with the Wnt co-receptor low-density lipoprotein receptor-related protein 6 (LRP6) resulting in attenuated ß-catenin degradation. Furthermore, applying anti-ß-catenin and anti-pIGF-IR antibodies to MG-63 cells demonstrated a cytoplasmic and to the membrane interaction between these molecules that increased upon exogenous biglycan treatment. In parallel, the downregulation of biglycan significantly inhibited both basal and IGF-I-dependent ERK1/2 activation, (p ≤ 0.001). In summary, we report a novel mechanism where biglycan through a LRP6/ß-catenin/IGF-IR signaling axis enhances osteosarcoma cell growth.

10.
Lab Invest ; 98(12): 1516-1526, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30206314

RESUMEN

During the past few years, considerable evidence has uncovered a strong relationship between fat and bone metabolism. Consequently, alterations in plasma lipid metabolic pathways strongly affect bone mass and quality. We recently showed that the deficiency of apolipoprotein A-1 (APOA1), a central regulator of high-density lipoprotein cholesterol (HDL-C) metabolism, results in reduced bone mass in C57BL/6 mice. It is documented that apolipoprotein E (APOE), a lipoprotein know for its atheroprotective functions and de novo biogenesis of HDL-C, is associated with the accumulation of fat in the liver and other organs and regulates bone mass in mice. We further studied the mechanism of APOE in bone metabolism using well-characterized APOE knockout mice. We found that bone mass was remarkably reduced in APOE deficient mice fed Western-type diet (WTD) compared to wild type counterparts. Static (microCT-based) and dynamic histomorphometry showed that the reduced bone mass in APOΕ-/- mice is attributed to both decreased osteoblastic bone synthesis and elevated osteoclastic bone resorption. Interestingly, histologic analysis of femoral sections revealed a significant reduction in the number of bone marrow lipoblasts in APOΕ-/- compared to wild type mice under WTD. Analyses of whole bone marrow cells obtained from femora of both animal groups showed that APOE null mice had significantly reduced levels of the osteoblastic (RUNX2 and Osterix) and lipoblastic (PPARγ and CEBPα) cardinal regulators. Additionally, the modulators of bone remodeling RANK, RANKL, and cathepsin K were greatly increased, while OPG and the OPG/RANKL ratio were remarkably decreased in APOΕ-/- mice fed WTD, compared to their wild-type counterparts. These findings suggest that APOE deficiency challenged with WTD reduces osteoblastic and lipoblastic differentiation and activity, whereas it enhances osteoclastic function, ultimately resulting in reduced bone mass, in mice.


Asunto(s)
Apolipoproteínas E/deficiencia , Huesos/fisiología , Diferenciación Celular , Dieta Occidental/efectos adversos , Adiposidad , Animales , Peso Corporal , Médula Ósea/fisiología , Lipogénesis , Ratones Endogámicos C57BL , Osteoblastos/fisiología , Osteoclastos/fisiología
11.
Artículo en Inglés | MEDLINE | ID: mdl-29559954

RESUMEN

Proteoglycans (PGs), important constituents of the extracellular matrix, have been associated with cancer pathogenesis. Their unique structure consisting of a protein core and glycosaminoglycan chains endowed with fine modifications constitutes these molecules as capable cellular effectors important for homeostasis and contributing to disease progression. Indeed, differential expression of PGs and their interacting proteins has been characterized as specific for disease evolvement in various cancer types. Importantly, PGs to a large extent regulate the bioavailability of hormones, growth factors, and cytokines as well as the activation of their respective receptors which regulate phenotypic diversibility, gene expression and rates of recurrence in specific tumor types. Defining and targeting these effectors on an individual patient basis offers ground for the development of newer therapeutic approaches which may act as either supportive or a substitute treatment to the standard therapy protocols. This review discusses the roles of PGs in cancer progression, developing technologies utilized for the defining of the PG "signature" in disease, and how this may facilitate the generation of tailor-made cancer strategies.

12.
Exp Cell Res ; 361(1): 9-18, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28962916

RESUMEN

Fibrosarcoma is a tumor of mesenchymal origin, originating from fibroblasts. IGF-I is an anabolic growth factor which exhibits significant involvement in cancer progression. In this study, we investigated the possible participation of syndecan-2 (SDC-2), a cell membrane heparan sulfate (HS) proteoglycan on IGF-I dependent fibrosarcoma cell motility. Our results demonstrate that SDC-2-deficient HT1080 cells exhibit attenuated IGF-I-dependent chemotactic migration (p < 0.001). SDC-2 was found to co-localize to IGF-I receptor (IGF-IR) in a manner dependent on IGF-I activity (P ≤ 0.01). In parallel, the downregulation of SDC-2 significantly inhibited both basal and due to IGF-I action ERK1/2 activation, (p < 0.001). The phosphorylation levels of ezrin (Thr567), which is suggested to act as a signaling bridge between the cellular membrane receptors and actin cytoskeleton, were strongly enhanced by IGF-I at both 1h and 24h (p < 0.05; p < 0.01). The formation of an immunoprecipitative complex revealed an association between SDC2 and ezrin which was enhanced through IGF-I action (p < 0.05). Immunoflourescence demonstrated a co-localization of IGF-IR, SDC2 and ezrin upregulated by IGF-I action. IGF-I enhanced actin polymerization and ezrin/actin specific localization to cell membranes. Finally, treatment with IGF-I strongly increased SDC2 expression at both the mRNA and protein level (p < 0.001). Therefore, we propose a novel SDC2-dependent mechanism, where SDC2 is co-localized with IGF-IR and enhances its' IGFI-dependent downstream signaling. SDC2 mediates directly IGFI-induced ERK1/2 activation, it recruits ezrin, contributes to actin polymerization and ezrin/actin specific localization to cell membranes, ultimately facilitating the progression of IGFI-dependent fibrosarcoma cell migration.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteínas del Citoesqueleto/metabolismo , Fibrosarcoma/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Sindecano-2/metabolismo , Proteínas del Citoesqueleto/genética , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Transducción de Señal/efectos de los fármacos , Sindecano-2/genética , Células Tumorales Cultivadas
13.
IUBMB Life ; 69(11): 824-833, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28940845

RESUMEN

Syndecan 2 (SDC2) belongs to a four-member family of evolutionary conserved small type I transmembrane proteoglycans consisting of a protein core to which glycosaminoglycan chains are covalently attached. SDC2 is a cell surface heparan sulfate proteoglycan, which is increasingly drawing attention for its distinct characteristics and its participation in numerous cell functions, including those related to carcinogenesis. Increasing evidence suggests that the role of SDC2 in cancer pathogenesis is dependent on cancer tissue origin rendering its use as a biomarker/therapeutic target feasible. This mini review discusses the mechanisms, through which SDC2, in a distinct manner, modulates complex signalling networks to affect cancer progression. © 2017 IUBMB Life, 69(11):824-833, 2017.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias del Colon/genética , Fibrosarcoma/genética , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/genética , Neoplasias Pancreáticas/genética , Sindecano-2/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Membrana Celular/metabolismo , Membrana Celular/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Progresión de la Enfermedad , Femenino , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Especificidad de Órganos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Dominios Proteicos , Transducción de Señal , Sindecano-2/metabolismo
14.
Data Brief ; 9: 568-572, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27752527

RESUMEN

In this data article, the potential role of p53 tumor suppressor gene (p53) on the attachment ability of MCF-7 breast cancer cells was investigated. In our main article, "IGF-I/ EGF and E2 signaling crosstalk through IGF-IR conduit point affect breast cancer cell adhesion" (K. Voudouri, D. Nikitovic, A. Berdiaki, D. Kletsas, N.K. Karamanos, G.N. Tzanakakis, 2016) [1], we describe the key role of IGF-IR in breast cancer cell adhesion onto fibronectin (FN). p53 tumor suppressor gene is a principal regulator of cancer cell proliferation. Various data have demonstrated an association between p53 and IGF-IR actions on cell growth through its' putative regulation of IGF-IR expression. According to our performed experiments, p53 does not modify IGF-IR expression and does not affect basal MCF-7 cells adhesion onto FN. Moreover, technical details about the performance of adhesion assay onto the FN substrate were provided.

15.
Oncol Rep ; 36(5): 2471-2480, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27572115

RESUMEN

Soft tissue sarcomas are rare, heterogeneous tumors of mesenchymal origin with an aggressive behavior. Heparin is a mixture of heavily sulfated, linear glycosaminoglycan (GAG) chains, which participate in the regulation of various cell biological functions. Heparin is considered to have significant anticancer capabilities, although the mechanisms involved have not been fully defined. In the present study, the effects of unfractionated heparin (UFH) and low­molecular­weight heparin (LMWH) on B6FS fibrosarcoma cell motility were examined. Both preparations of heparin were shown to both enhance B6FS cell adhesion (p<0.01 and p<0.05), and migration (p<0.05), the maximal effect being evident at the concentration of 10 µg/ml. The utilization of FAK­deficient cells demonstrated that the participation of FAK was obligatory for heparin­dependent fibrosarcoma cell adhesion (p<0.05). The results of confocal microscopy indicated that heparin was taken up by the B6FS cells, and that UFH and LMWH induced F­actin polymerization. Heparitinase digestion demonstrated that the endogenous heparan sulfate (HS) chains did not affect the motility of the B6FS cells (p>0.05, not significant). In conclusion, both UFH and LMWH, through a FAK/actin cytoskeleton axis, promoted the adhesion and migration of B6FS fibrosarcoma cells. Thus, our findings indicate that the responsiveness of fibrosarcoma cells to the exogenous heparin/HS content of the cancer microenvironment may play a role in their ability to become mobile and metastasize.


Asunto(s)
Anticoagulantes/administración & dosificación , Fibrosarcoma/tratamiento farmacológico , Quinasa 1 de Adhesión Focal/genética , Sarcoma/tratamiento farmacológico , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Fibrosarcoma/genética , Fibrosarcoma/patología , Quinasa 1 de Adhesión Focal/metabolismo , Heparina/administración & dosificación , Heparina de Bajo-Peso-Molecular/administración & dosificación , Humanos , Sarcoma/genética , Sarcoma/patología
16.
Oncol Rep ; 36(4): 1787-92, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27499459

RESUMEN

Osteosarcoma (OS) is a primary bone tumor of mesenchymal origin mostly affecting children and adolescents. The OS extracellular matrix (ECM) is extensively altered as compared to physiological bone tissue. Indeed, the main characteristic of the most common osteoblastic subtype of OS is non­mineralized osteoid production. Parathyroid hormone (PTH) is a polypeptide hormone secreted by the chief cells of the parathyroid glands. The PTH-related peptide (PTHrP) may be comprised of 139, 141 or 173 amino acids and exhibits considerate N­terminal amino acid sequence homology with PTH. The function of PTH/PTHrP is executed through the activation of the PTH receptor 1 (PTHR1) and respective downstream intracellular pathways which regulate skeletal development, bone turnover and mineral ion homeostasis. Both PTHR1 and its PTH/PTHrP ligands have been shown to be expressed in OS and to affect the functions of these tumor cells. This review aims to highlight the less well known aspects of PTH/PTHrP functions in the progression of OS by focusing on ECM-dependent signaling.


Asunto(s)
Neoplasias Óseas/patología , Matriz Extracelular/metabolismo , Osteosarcoma/patología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Hormona Paratiroidea/metabolismo , Transducción de Señal/fisiología , Neoplasias Óseas/metabolismo , Humanos , Osteosarcoma/metabolismo
17.
Food Chem Toxicol ; 96: 302-8, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27554598

RESUMEN

We studied the feasibility of using a short-term culture of monocytes, isolated from peripheral donor blood, to assess the biological activity of different types of bionanomaterials (BNM): biodegradable polimeric particles, fiber and film substrates of micro- and nano-dimensions, fullerenes (F) and nanodiamonds (ND), which are either currently in use and/or potentially applicable in medicine. Additionally, the effect of creating a protein corona on ND and F particles was investigated. The cellular reduction of (4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) is a well-established tool for assessing the viability/metabolic activity of cells. The scanning electron microscopy assay can detect fine changes in cell morphology. In the present study BNM have been shown to affect; in a size, chemical composition and morphological characteristics-dependent manner, the ability of monocytes to reduce MTT as well as their morphology. Moreover, the specific effects of ND and F on MTT reduction and cell morphology were exhibited in a dose-dependent manner and sensitive to the formation of surface protein corona. Our results suggest that short-term culture of monocytes is a sensitive model system for assessing the biological effects of BMPs in vitro.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Diamante/química , Fulerenos/química , Nanopartículas del Metal/uso terapéutico , Microscopía Electrónica de Rastreo/métodos , Monocitos/ultraestructura , Células Cultivadas , Humanos , Técnicas In Vitro , Nanopartículas del Metal/química , Nanopartículas del Metal/ultraestructura
18.
Matrix Biol ; 56: 95-113, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27353258

RESUMEN

Epidermal growth factor (EGF)/insulin like growth factor-I (IGF-I) and Estradiol (E2) can regulate biological functions of hormone-dependent tumor cells. Fibronectin (FN) is a large glycoprotein abundantly expressed in breast cancer extracellular matrices (ECMs) postulated to be a marker of aggressiveness during cancer pathogenesis. In this study we demonstrate that IGF-I/EGF as well E2 strongly increase the adhesion of the MCF-7 breast cancer cells onto FN. Moreover, IGF-IR is necessary for the IGF-I-/EGF- and E2-induced cell adhesion. Erk1/2 inhibition abolished the IGF-I-/EGF-/E2-induced MCF-7 cell adhesion, suggesting that this regulation of cell adhesion is perpetrated through Erk1/2 downstream signaling. Erk1/2 signaling was shown to modulate IGF-IR status as its' inhibition attenuates both IGF-IR expression and activation. Notably, EGF and E2 enhanced the mRNA as well as protein expression of IGF-IR in MCF-7 cells. Confocal microscopy demonstrated that treatment of MCF-7 cells with IGF-I or EGF induced actin reorganization, which was attenuated with Erk1/2 inhibition. Interestingly, IGF-I treatment induced a co-localization of IGF-IR and FAK, which was evident mostly at the cell membranes of MCF-7 cells. In summary, IGF-IR was shown to be a convergence point for the IGF-/EGF- and E2-dependent MCF-7 cell adhesion onto FN.


Asunto(s)
Factor de Crecimiento Epidérmico/fisiología , Estradiol/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Receptor IGF Tipo 1/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Activación Enzimática , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Células MCF-7 , Transporte de Proteínas , Receptor Cross-Talk , Receptor IGF Tipo 1/genética
19.
Oncol Lett ; 11(5): 3354-3360, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27123116

RESUMEN

Regression in melanoma is a frequent biological event of uncertain prognostic value as the lesion exhibits heterogeneous phenotypical features, both at the morphological and immunohistochemical level. In the present study, we examined the expression of tissue inhibitors of metalloproteinases (TIMP1, TIMP2 and TIMP3) in melanoma with regression. We specifically examined the expression levels of these TIMPs in regressed components (RC) and non-regressed components (NRC) of the tumor and compared their expression levels with those in non-regressed melanomas. We found that TIMP1 was overexpressed in the NRC of melanomas with partial regression (PR) compared with the NRC in melanomas with segmental regression (SR) (P=0.011). TIMP2 was overexpressed in the NRC of melanomas with PR compared with the NRC in melanomas with SR (PR/SR, P=0.009); or compared with the NRC in melanomas with simultaneous SR-PR (P=0.002); or compared with melanomas without regression (absence of regression) (P=0.037). Moreover, TIMP3 was overexpressed in the NRC of all melanomas with SR as compared to the RC component (P=0.007). Our findings on the differential expression of TIMP1, TIMP2 and TIMP3 in melanomas with regression support the hypothesis that the morphological differences identified in the melanoma regression spectrum may have a correlation with prognosis. This may explain the controversial findings within the literature concerning the biological and prognostic role of regression in melanoma.

20.
Biochim Biophys Acta ; 1860(4): 814-24, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26825774

RESUMEN

BACKGROUND: High levels of hyaluronan (HA) synthesis in various cancer tissues, including sarcomas, are correlated with tumorigenesis and malignant transformation. RHAMM (receptor for hyaluronic acid-mediated motility) is overexpressed during tumor development in different malignancies. ß-Catenin is a crucial downstream mediator of the Wnt signaling cascade which facilitates carcinogenic events characterized by deregulated cell proliferation. METHODS: Real-time PCR, in vitro cell proliferation assay, siRNA transfection, flow cytometry, immunoprecipitation, western blotting and immunofluorescence were utilized. RESULTS: The reduction of RHAMM expression was strongly correlated with an inhibition of HT1080 fibrosarcoma cell growth (p≤0.01). LMWHA, in a RHAMM-dependent manner increases cell growth of HT1080 cells (p≤0.01). Both basal and LMWHA dependent growth of HT1080 cells was attenuated by ß-catenin deficiency (p≤0.01). ß-Catenin cytoplasmatic deposition is positively regulated by RHAMM (p≤0.01). Immunoflourescence and immunoprecipitation suggest that RHAMM/ß-catenin form an intracellular complex. Transfection experiments identified c-myc as candidate downstream mediator of RHAMM/ß-catenin effects on HT1080 fibrosarcoma cell proliferation. CONCLUSIONS: LMWHA/RHAMM downstream signaling regulates fibrosarcoma cell growth in a ß-catenin/c-myc dependent manner. GENERAL SIGNIFICANCE: The present study suggests that RHAMM is a novel ß-catenin intracellular binding partner, protecting ß-catenin from degradation and supporting the nuclear translocation of this key cellular mediator, which results in c-myc activation and enhanced fibrosarcoma cell growth.


Asunto(s)
Núcleo Celular/metabolismo , Proliferación Celular , Proteínas de la Matriz Extracelular/biosíntesis , Fibrosarcoma/metabolismo , Receptores de Hialuranos/biosíntesis , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular Tumoral , Núcleo Celular/genética , Proteínas de la Matriz Extracelular/genética , Fibrosarcoma/genética , Humanos , Receptores de Hialuranos/genética , Proteínas Proto-Oncogénicas c-myc/genética , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...