Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Crit Rev Immunol ; 40(2): 93-133, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32749091

RESUMEN

We have previously demonstrated that natural killer (NK) cells are the main immune effectors that can mediate selection and differentiation of different cancer stem cells and undifferentiated tumors via lysis and secreted or membrane-bound interferon-γ and tumor necrosis factor-α, respectively. This leads to growth inhibition and tumor metastasis curtailment. In this review, we present an overview of our findings on NK cell biology and its significance in selection and differentiation of stem-like tumors using in vitro and in vivo studies conducted in nonobese diabetic/severe combined immunodeficiency (scid)/interleukin-Rγ--, humanized-bone-marrow/liver/thymus (hu-BLT) mice, and those of human cancer patients. Moreover, we present recent advances in NK cell expansion and therapeutic delivery and discuss the superiority of allogeneic supercharged NK cells over their autologous counterparts for cancer treatment. We review potential loss of NK cell numbers and function at neoplastic and preneoplastic stages of tumorigenesis as a potential mechanism for pancreatic cancer induction and progression. We believe that NK cells should be placed highly in the armamentarium of tumor immunotherapy due to their indispensable role in targeting cancer stem-like/poorly differentiated tumors and a variety of other key NK cell functions that are discussed in this report, including their role in CD8+ T-cell expansion and targeting gene knockout or dedifferentiated tumors. The combination of allogeneic supercharged NK cells and other immunotherapeutic strategies such as oncolytic viruses, antibody-dependent cellular cytotoxicity-inducing antibodies, checkpoint inhibitors, chimeric antigen receptor (CAR)-T cells and CAR-NK cells, chemotherapeutics, and radiotherapeutic strategies can be used for optimal eradication of tumors.


Asunto(s)
Inmunidad , Huésped Inmunocomprometido , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Biomarcadores , Diferenciación Celular/inmunología , Terapia Combinada , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Humanos , Ratones Noqueados , Neoplasias/patología , Neoplasias/terapia
2.
Molecules ; 24(11)2019 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-31167364

RESUMEN

Currently available drugs for treatment of glioblastoma, the most aggressive brain tumor, remain inefficient, thus a plethora of natural compounds have already been shown to have antimalignant effects. However, these have not been tested for their impact on tumor cells in their microenvironment-simulated cell models, e.g., mesenchymal stem cells in coculture with glioblastoma cell U87 (GB). Mesenchymal stem cells (MSC) chemotactically infiltrate the glioblastoma microenvironment. Our previous studies have shown that bone-marrow derived MSCs impair U87 growth and invasion via paracrine and cell-cell contact-mediated cross-talk. Here, we report on a plant-derived protein, obtained from Crataeva tapia tree Bark Lectin (CrataBL), having protease inhibitory/lectin activities, and demonstrate its effects on glioblastoma cells U87 alone and their cocultures with MSCs. CrataBL inhibited U87 cell invasion and adhesion. Using a simplified model of the stromal microenvironment, i.e., GB/MSC direct cocultures, we demonstrated that CrataBL, when added in increased concentrations, caused cell cycle arrest and decreased cocultured cells' viability and proliferation, but not invasion. The cocultured cells' phenotypes were affected by CrataBL via a variety of secreted immunomodulatory cytokines, i.e., G-CSF, GM-CSF, IL-6, IL-8, and VEGF. We hypothesize that CrataBL plays a role by boosting the modulatory effects of MSCs on these glioblastoma cell lines and thus the effects of this and other natural lectins and/or inhibitors would certainly be different in the tumor microenvironment compared to tumor cells alone. We have provided clear evidence that it makes much more sense testing these potential therapeutic adjuvants in cocultures, mimicking heterogeneous tumor-stroma interactions with cancer cells in vivo. As such, CrataBL is suggested as a new candidate to approach adjuvant treatment of this deadly tumor.


Asunto(s)
Capparaceae/química , Células Madre Mesenquimatosas/efectos de los fármacos , Corteza de la Planta/química , Extractos Vegetales/farmacología , Lectinas de Plantas/farmacología , Inhibidores de Proteasas/farmacología , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Citocinas/biosíntesis , Glioblastoma/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Metaloproteasas/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Extractos Vegetales/química , Lectinas de Plantas/química , Inhibidores de Proteasas/química
3.
Arch Toxicol ; 92(5): 1893-1903, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29549414

RESUMEN

The problem of the currently used routine genotoxicity tests is relatively low predictivity of in vitro tests for in vivo genotoxicity and carcinogenicity. An important reason is considered to be inadequate expression of xenobiotic-metabolizing enzymes in indicator cell lines. The aim of our study was to generate metabolically active differentiated hepatic progenies (hDHP) from human adipose tissue-derived mesenchymal stem cells (hASC) for genotoxicity testing. hDHP, generated using a three-step hepatic differentiation procedure, expressed hepatic properties such as glycogen storage and albumin secretion. The results of the comet assay demonstrated comparable sensitivity of hASC and hDHP to detect DNA damage induced by a direct acting genotoxic agent tert-butylhydroperoxide. Exposure to model indirect acting genotoxins benzo(a)pyrene, aflatoxin B1, and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine did not induce DNA damage in hASC, while hDHP cells detected DNA damage induced by benzo(a)pyrene and aflatoxin B1, indicating their metabolic activity. The gene and protein expression analysis confirmed the presence of key enzymes involved in metabolism of the three genotoxins in hDHP cells. Moreover, the exposure of hDHP to the model pro-carcinogens altered the expression of selected metabolic genes. hDHP were further immortalized with hTERT transfection, resulting in a stable cell line that can be matured to metabolically active hDHP ready for genotoxicity testing in only 2 weeks. The advantage of these immortalized cells is their prolonged replicative life span and consequently limitless supply of hDHP cells. We conclude that hDHP cells have a great potential for the application in the routine genotoxicity testing and are therefore worth further investigations.


Asunto(s)
Tejido Adiposo/citología , Hígado/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Pruebas de Mutagenicidad/métodos , Aflatoxina B1/toxicidad , Benzo(a)pireno/toxicidad , Diferenciación Celular , Línea Celular , Ensayo Cometa/métodos , Enzimas/genética , Enzimas/metabolismo , Femenino , Células Hep G2 , Humanos , Imidazoles/toxicidad , Células Madre Mesenquimatosas/fisiología
4.
Radiol Oncol ; 48(3): 257-66, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25177240

RESUMEN

BACKGROUND: Glioblastoma multiforme (GBM) is a brain tumour with a very high patient mortality rate, with a median survival of 47 weeks. This might be improved by the identification of novel diagnostic, prognostic and predictive therapy-response biomarkers, preferentially through the monitoring of the patient blood. The aim of this study was to define the impact of GBM in terms of alterations of the plasma protein levels in these patients. MATERIALS AND METHODS: We used a commercially available antibody array that includes 656 antibodies to analyse blood plasma samples from 17 healthy volunteers in comparison with 17 blood plasma samples from patients with GBM. RESULTS: We identified 11 plasma proteins that are statistically most strongly associated with the presence of GBM. These proteins belong to three functional signalling pathways: T-cell signalling and immune responses; cell adhesion and migration; and cell-cycle control and apoptosis. Thus, we can consider this identified set of proteins as potential diagnostic biomarker candidates for GBM. In addition, a set of 16 plasma proteins were significantly associated with the overall survival of these patients with GBM. Guanine nucleotide binding protein alpha (GNAO1) was associated with both GBM presence and survival of patients with GBM. CONCLUSIONS: Antibody array analysis represents a useful tool for the screening of plasma samples for potential cancer biomarker candidates in small-scale exploratory experiments; however, clinical validation of these candidates requires their further evaluation in a larger study on an independent cohort of patients.

5.
iScience ; 27(9): 110604, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39252971

RESUMEN

Glioblastoma (GB) is the most common primary malignant brain tumor, characterized by resistance to therapy. Despite aggressive treatment options, GB remains an incurable disease. Invasiveness and heterogeneity are key GB features that cannot be studied in preclinical in vitro models. In this study, we investigated the effects of standard therapy using patient-derived GB organoids (GBOs). GBOs reflect the complexity and heterogeneity of the original tumor tissue. No significant effect on GBO viability or invasion was observed after irradiation and temozolomide treatment. E3 ubiquitin-protein ligase (MDM2), cyclin-dependent kinase inhibitor 1A (CDKN1A), and the serine/threonine kinases ATM and ATR were upregulated at the gene and protein levels after treatment. Our results show that the p53 pathway and DNA-damage response mechanisms were triggered, suggesting that GBOs recapitulate GB therapy resistance. GBOs thus provide a highly efficient platform to assess the specific responses of GB patients to therapy and to further explore therapy resistance.

6.
Artículo en Inglés | MEDLINE | ID: mdl-36016679

RESUMEN

Tartary buckwheat flavonoids (TBFs) exhibit diverse biological activities, with antioxidant, antidiabetes, anti-inflammatory, and cholesterol-lowering properties. In this study, we investigated the role of TBFs in attenuating glucose and lipid disturbances in diabetic mice and hence preventing the occurrence of diabetes-related colon lesions in mice by regulating the gut microbiota. The results showed that TBFs (1) reversed blood glucose levels and body weight changes; (2) improved levels of serum total cholesterol (TC), triglycerides (TGs), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and fasting insulin; and (3) significantly reduced diabetes-related colon lesions in diabetic mice. In addition, TBFs also affected the diabetes-related imbalance of the gut microbiota and enriched beneficial microbiota, including Akkermansia and Prevotella. The TBF also selectively increased short-chain fatty acid-producing bacteria, including Roseburia and Odoribacter, and decreased the abundance of the diabetes-related gut microbiota, including Escherichia, Mucispirillum, and Bilophila. The correlation analysis indicated that TBFs improved metabolic parameters related to key communities of the gut microbiota. Our data suggested that TBFs alleviated glucose and lipid disturbances and improved colon lesions in diabetic mice, possibly by regulating the community composition of the gut microbiota. This regulation of the gut microbiota composition may explain the observed effects of TBFs to alleviate diabetes-related symptoms.

7.
Oncotarget ; 9(30): 21296-21312, 2018 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-29765540

RESUMEN

Glioblastoma is the most aggressive brain tumor with poor overall survival bellow 2 years. The natural compounds with anti-cancer properties, are thus gaining attention for possible adjuvant GBM treatment. In various cancer models Enterolobium contortisiliquum Trypsin Inhibitor (EcTI) proved to have anti-cancer effects. Here, we investigated the EcTI effects on GBM U87 cells and on mesenchymal stem cells (MSC) compared to their direct coculture (MSC/U87). MSC are present in tumor stroma, modulating GBM cells phenotype, and also represent potential drug delivery vehicle due to their tumor tropism. We showed that in p53-wild type U87 cells, metabolic activity was less affected by EcTI as in MSC monocuture, but the metabolic rate of mixed coculture was significantly reduced at lower EcTI concentration. Under coculture condition, EcTI potentiated MSC induced cell cycle arrest, possible due to highly increased p53, p21 and lower D1 expression, but there was no effect on apoptosis. Accordingly, in the coculture EcTI also enhanced Ca2+ signalling mediated via bradykinin receptor 2, being associated with nitric oxide release that highly impaired proliferation and invasion. The mechanism did not seem to involve changes in cell adhesion but rather it down-regulated the ß1 integrin signaling with associated p-FAK in U87 cells, both supporting inhibition of invasion. Finally, some cytokines were down-regulated, indicating that EcTI inhibition of signalling might be mediated by cytokines. In conclusion, these results indicate that in cocultured MSC/U87 cells EcTI impairs the metabolic activity, proliferation, and reduced invasion, possibly associated with observed cytokines secretion. In this context, we confirmed that the plant derived protein potentiated the anticancer effects, induced by MSC, as represented by GBM U87 cell line.

8.
Oncotarget ; 7(1): 593-609, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26573230

RESUMEN

The cancer stem cell model suggests that glioblastomas contain a subpopulation of stem-like tumor cells that reproduce themselves to sustain tumor growth. Targeting these cells thus represents a novel treatment strategy and therefore more specific markers that characterize glioblastoma stem cells need to be identified. In the present study, we performed transcriptomic analysis of glioblastoma tissues compared to normal brain tissues revealing sensible up-regulation of CD9 gene. CD9 encodes the transmembrane protein tetraspanin which is involved in tumor cell invasion, apoptosis and resistance to chemotherapy. Using the public REMBRANDT database for brain tumors, we confirmed the prognostic value of CD9, whereby a more than two fold up-regulation correlates with shorter patient survival. We validated CD9 gene and protein expression showing selective up-regulation in glioblastoma stem cells isolated from primary biopsies and in primary organotypic glioblastoma spheroids as well as in U87-MG and U373 glioblastoma cell lines. In contrast, no or low CD9 gene expression was observed in normal human astrocytes, normal brain tissue and neural stem cells. CD9 silencing in three CD133+ glioblastoma cell lines (NCH644, NCH421k and NCH660h) led to decreased cell proliferation, survival, invasion, and self-renewal ability, and altered expression of the stem-cell markers CD133, nestin and SOX2. Moreover, CD9-silenced glioblastoma stem cells showed altered activation patterns of the Akt, MapK and Stat3 signaling transducers. Orthotopic xenotransplantation of CD9-silenced glioblastoma stem cells into nude rats promoted prolonged survival. Therefore, CD9 should be further evaluated as a target for glioblastoma treatment.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Células Madre Neoplásicas/metabolismo , Tetraspanina 29/genética , Animales , Biomarcadores de Tumor/metabolismo , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Técnicas de Cultivo de Órganos , Interferencia de ARN , Ratas Desnudas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia , Tetraspanina 29/metabolismo , Trasplante Heterólogo , Regulación hacia Arriba
9.
Protein Pept Lett ; 22(4): 322-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25642990

RESUMEN

Numerous small molecules including cytokines primarily associated with immune response have been shown to play a role in normal mesenchymal stem cells (MSC) and tumour cells' communication. One characteristic that distinguishes MSC from fibroblast and other cells of mesenchymal origin is their pro-tumour migratory behaviour. Recognizing the cytokines as key players of the MSC/tumour cell cross-talk and understanding their intracellular signalling, should lead to a development of more efficient anti-tumour therapies. Those are urgently needed for improving the treatment of patients with glioblastoma multiformae (GBM) that are suffering from most aggressive and incurable type of brain tumours. So far, the "cytokine signalling interference" approach, employing genetically modified MSCs and GBM cells in animal xenograft models pointed to the mechanisms underlying tumour - directed migration and immunomodulatory role of MSCs. There, MSC's effects on tumour growth were shown to vary substantially, and to depend on the type of the cells and the animal model used. This review is focusing on the cytokines produced by MSCs and their involvement in proliferation, migration, angiogenesis, apoptosis and immune cell infiltration. Recently, targeted therapies have emerged as a promising modality for GBM treatment. New approaches, combining these with MSCs as cellular vectors for modulating cytokines and cytokine receptors' signalling in GBM may thus prove more efficient at inhibiting glioma progression.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Citocinas/metabolismo , Glioblastoma/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/patología , Sistemas de Liberación de Medicamentos , Glioblastoma/patología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología
10.
J Histochem Cytochem ; 63(10): 749-61, 2015 10.
Artículo en Inglés | MEDLINE | ID: mdl-26109632

RESUMEN

Zebrafish (Danio rerio) and their transparent embryos are becoming an increasingly popular tool for studying processes involved in tumor progression and in the search for novel tumor treatment approaches. The xenotransplantation of fluorescently labeled mammalian cancer cells into zebrafish embryos is an approach enabling relatively high-throughput in vivo analyses. The small size of the embryos as well as the relative simplicity of their manipulation and maintenance allow for large numbers of embryos to be processed efficiently in a short time and at low cost. Furthermore, the possibility of fluorescence microscopic imaging of tumor progression within zebrafish embryos and larvae holds unprecedented potential for the real-time visualization of these processes in vivo. This review presents the methodologies of xenotransplantation studies on zebrafish involving research on tumor invasion, proliferation, tumor-induced angiogenesis and screening for antitumor therapeutics. We further focus on the application of these zebrafish to the study of glioma; in particular, its most common and malignant form, glioblastoma.


Asunto(s)
Modelos Animales de Enfermedad , Glioma/patología , Pez Cebra , Animales , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales/métodos , Glioma/tratamiento farmacológico , Humanos , Microscopía/métodos , Trasplante de Neoplasias/patología , Trasplante Heterólogo/métodos , Pez Cebra/embriología , Pez Cebra/crecimiento & desarrollo
11.
J Histochem Cytochem ; 63(7): 481-93, 2015 07.
Artículo en Inglés | MEDLINE | ID: mdl-25809793

RESUMEN

Poor survival of high-grade glioma is at least partly caused by glioma stem-like cells (GSLCs) that are resistant to therapy. GSLCs reside in niches in close vicinity of endothelium. The aim of the present study was to characterize proteins that may be functional in the GSLC niche by performing immunohistochemistry on serial cryostat sections of human high-grade glioma samples. We have found nine niches in five out of five high-grade glioma samples that were all surrounding arterioles with CD31+ endothelial cells and containing cellular structures that were CD133+ and nestin+. All nine niches expressed stromal-derived factor-1α (SDF-1α), its receptor C-X-C chemokine receptor type 4 (CXCR4), osteopontin and cathepsin K. SDF-1α plays a role in homing of CXCR4+ stem cells and leukocytes, whereas osteopontin and cathepsin K promote migration of cancer cells and leukocytes. Leukocyte-related markers, such as CD68, macrophage matrix metalloprotease-9, CD177 and neutrophil elastase were often but not always detected in the niches. We suggest that SDF-1α is involved in homing of CXCR4+ GSLCs and leukocytes and that cathepsin K and osteopontin are involved in the migration of GSLCs out of the niches.


Asunto(s)
Catepsina K/metabolismo , Quimiocina CXCL12/metabolismo , Glioma/patología , Células Madre Neoplásicas , Osteopontina/metabolismo , Receptores CXCR4/metabolismo , Nicho de Células Madre/fisiología , Antígeno AC133 , Adulto , Anciano , Antígenos CD/metabolismo , Arteriolas/metabolismo , Arteriolas/patología , Biomarcadores de Tumor/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/irrigación sanguínea , Glioma/inmunología , Glioma/metabolismo , Glicoproteínas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Nestina/metabolismo , Activación Neutrófila , Neutrófilos/inmunología , Neutrófilos/metabolismo , Péptidos/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
PLoS One ; 9(11): e113688, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25419715

RESUMEN

Malignant gliomas are among the rarest brain tumours, and they have the worst prognosis. Grade IV astrocytoma, known as glioblastoma multiforme (GBM), is a highly lethal disease where the standard therapies of surgery, followed by radiation and chemotherapy, cannot significantly prolong the life expectancy of the patients. Tumour recurrence shows more aggressive form compared to the primary tumour, and results in patient survival from 12 to 15 months only. Although still controversial, the cancer stem cell hypothesis postulates that cancer stem cells are responsible for early relapse of the disease after surgical intervention due to their high resistance to therapy. Alternative strategies for GBM therapy are thus urgently needed. Nanobodies are single-domain antigen-binding fragments of heavy-chain antibodies, and together with classical antibodies, they are part of the camelid immune system. Nanobodies are small and stable, and they share a high degree of sequence identity to the human heavy chain variable domain, and these characteristics offer them advantages over classical antibodies or antibody fragments. We first immunised an alpaca with a human GBM stem-like cell line prepared from primary GBM cultures. Next, a nanobody library was constructed in a phage-display vector. Using nanobody phage-display technology, we selected specific GBM stem-like cell binders through a number of affinity selections, using whole cell protein extracts and membrane protein-enriched extracts from eight different GBM patients, and membrane protein-enriched extracts from two established GBM stem-like cell lines (NCH644 and NCH421K cells). After the enrichment, periplasmic extract ELISA was used to screen for specific clones. These nanobody clones were recloned into the pHEN6 vector, expressed in Escherichia coli WK6, and purified using immobilised metal affinity chromatography and size-exclusion chromatography. Specific nanobody:antigen pairs were obtained and mass spectrometry analysis revealed two proteins, TRIM28 and ß-actin, that were up-regulated in the GBM stem-like cells compared to the controls.


Asunto(s)
Actinas/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Animales , Afinidad de Anticuerpos/inmunología , Western Blotting , Neoplasias Encefálicas/diagnóstico , Camélidos del Nuevo Mundo , Línea Celular Tumoral , Glioblastoma/diagnóstico , Humanos , Masculino , Espectrometría de Masas , Datos de Secuencia Molecular , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Biblioteca de Péptidos , Proteómica/métodos , Homología de Secuencia de Aminoácido , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/metabolismo , Proteína 28 que Contiene Motivos Tripartito
13.
Exp Neurol ; 234(1): 208-19, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22230665

RESUMEN

The interaction of human mesenchymal stem cells (hMSCs) and tumor cells has been investigated in various contexts. HMSCs are considered as cellular treatment vectors based on their capacity to migrate towards a malignant lesion. However, concerns about unpredictable behavior of transplanted hMSCs are accumulating. In malignant gliomas, the recruitment mechanism is driven by glioma-secreted factors which lead to accumulation of both, tissue specific stem cells as well as bone marrow derived hMSCs within the tumor. The aim of the present work was to study specific cellular interactions between hMSCs and glioma cells in vitro. We show, that glioma cells as well as hMSCs differentially express connexins, and that they interact via gap-junctional coupling. Besides this so-called functional syncytium formation, we also provide evidence of cell fusion events (structural syncytium). These complex cellular interactions led to an enhanced migration and altered proliferation of both, tumor and mesenchymal stem cell types in vitro. The presented work shows that glioma cells display signs of functional as well as structural syncytium formation with hMSCs in vitro. The described cellular phenomena provide new insight into the complexity of interaction patterns between tumor cells and host cells. Based on these findings, further studies are warranted to define the impact of a functional or structural syncytium formation on malignant tumors and cell based therapies in vivo.


Asunto(s)
Conexina 43/metabolismo , Expresión Génica/fisiología , Células Gigantes/fisiología , Glioma/fisiopatología , Células Madre Mesenquimatosas/fisiología , Aminoácidos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Conexina 43/genética , Medios de Cultivo Condicionados/farmacología , Dextranos , Expresión Génica/efectos de los fármacos , Células Gigantes/efectos de los fármacos , Células Gigantes/patología , Glioma/patología , Humanos , ARN Mensajero/metabolismo , Rodaminas , Fracciones Subcelulares
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA