Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 10345, 2024 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710795

RESUMEN

Skeletal bone function relies on both cells and cellular niches, which, when combined, provide guiding cues for the control of differentiation and remodeling processes. Here, we propose an in vitro 3D model based on human fetal osteoblasts, which eases the study of osteocyte commitment in vitro and thus provides a means to examine the influences of biomaterials, substances or cells on the regulation of these processes. Aggregates were formed from human fetal osteoblasts (hFOB1.19) and cultivated under proliferative, adipo- and osteoinductive conditions. When cultivated under osteoinductive conditions, the vitality of the aggregates was compromised, the expression levels of the mineralization-related gene DMP1 and the amount of calcification and matrix deposition were lower, and the growth of the spheroids stalled. However, within spheres under growth conditions without specific supplements, self-organization processes occur, which promote extracellular calcium deposition, and osteocyte-like cells develop. Long-term cultivated hFOB aggregates were free of necrotic areas. Moreover, hFOB aggregates cultivated under standard proliferative conditions supported the co-cultivation of human monocytes, microvascular endothelial cells and stromal cells. Overall, the model presented here comprises a self-organizing and easily accessible 3D osteoblast model for studying bone marrow formation and in vitro remodeling and thus provides a means to test druggable molecular pathways with the potential to promote life-long bone formation and remodeling.


Asunto(s)
Diferenciación Celular , Técnicas de Cocultivo , Osteoblastos , Humanos , Osteoblastos/metabolismo , Osteoblastos/citología , Microambiente Celular , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Osteogénesis , Agregación Celular , Células Cultivadas
2.
Bioengineering (Basel) ; 9(2)2022 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-35200413

RESUMEN

The wide use of 3D-organotypic cell models is imperative for advancing our understanding of basic cell biological mechanisms. For this purpose, easy-to-use enabling technology is required, which should optimally link standardized assessment methods to those used for the formation, cultivation, and evaluation of cell aggregates or primordial tissue. We thus conceived, manufactured, and tested devices which provide the means for cell aggregation and online monitoring within a hanging drop. We then established a workflow for spheroid manipulation and immune phenotyping. This described workflow conserves media and reagent, facilitates the uninterrupted tracking of spheroid formation under various conditions, and enables 3D-marker analysis by means of 3D epifluorescence deconvolution microscopy. We provide a full description of the low-cost manufacturing process for the fluidic devices and microscopic assessment tools, and the detailed blueprints and building instructions are disclosed. Conclusively, the presented compilation of methods and techniques promotes a quick and barrier-free entry into 3D cell biology.

3.
Front Physiol ; 6: 362, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26696897

RESUMEN

Multipotential mesenchymal stromal cells (MSC) are present as a rare subpopulation within any type of stroma in the body of higher animals. Prominently, MSC have been recognized to reside in perivascular locations, supposedly maintaining blood vessel integrity. During tissue damage and injury, MSC/pericytes become activated, evade from their perivascular niche and are thus assumed to support wound healing and tissue regeneration. In vitro MSC exhibit demonstrated capabilities to differentiate into a wide variety of tissue cell types. Hence, many MSC-based therapeutic approaches have been performed to address bone, cartilage, or heart regeneration. Furthermore, prominent studies showed efficacy of ex vivo expanded MSC to countervail graft-vs.-host-disease. Therefore, additional fields of application are presently conceived, in which MSC-based therapies potentially unfold beneficial effects, such as amelioration of non-healing conditions after tendon or spinal cord injury, as well as neuropathies. Working along these lines, MSC-based scientific research has been forged ahead to prominently occupy the clinical stage. Aging is to a great deal stochastic by nature bringing forth changes in an individual fashion. Yet, is aging of stem cells or/and their corresponding niche considered a determining factor for outcome and success of clinical therapies?

4.
Mol Ther ; 21(11): 2032-42, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23939023

RESUMEN

Recombinant vesicular stomatitis virus (VSV) shows promise for the treatment of hepatocellular carcinoma (HCC), but its safety and efficacy when administered in a setting of hepatic fibrosis, which occurs in the majority of clinical cases, is unknown. We hypothesized that VSV could provide a novel benefit to the underlying fibrosis, due to its ability to replicate and cause cell death specifically in activated hepatic stellate cells. In addition to the ability of VSV to produce a significant oncolytic response in HCC-bearing rats in the background of thioacetamide-induced hepatic fibrosis without signs of hepatotoxicity, we observed a significant downgrading of fibrosis stage, a decrease in collagen content in the liver, and modulation of gene expression in favor of fibrotic regression. Together, this work suggests that VSV is not only safe and effective for the treatment of HCC with underlying fibrosis, but it could potentially be developed for clinical application as a novel antifibrotic agent.


Asunto(s)
Carcinoma Hepatocelular/terapia , Cirrosis Hepática/complicaciones , Cirrosis Hepática/terapia , Neoplasias Hepáticas/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus de la Estomatitis Vesicular Indiana/genética , Animales , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Células Hep G2 , Humanos , Infusiones Intraarteriales , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática Experimental/inducido químicamente , Neoplasias Hepáticas/complicaciones , Masculino , Ratas , Ratas Endogámicas BUF , Tioacetamida
5.
J Virol ; 86(9): 4844-55, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22345438

RESUMEN

Vesicular stomatitis virus (VSV), a negative-sense single-stranded-RNA rhabdovirus, is an extremely promising oncolytic agent for cancer treatment. Since oncolytic virotherapy is moving closer to clinical application, potentially synergistic combinations of oncolytic viruses and molecularly targeted antitumor agents are becoming a meaningful strategy for cancer treatment. Mitogen-activated protein kinase (MAPK) inhibitors have been shown to impair liver cell proliferation and tumor development, suggesting their potential use as therapeutic agents for hepatocellular carcinoma (HCC). In this work, we show that the impairment of MAPK in vitro did not interfere with the oncolytic properties of VSV in HCC cell lines. Moreover, the administration of MAPK inhibitors did not restore the responsiveness of HCC cells to alpha/beta interferon (IFN-α/ß). In contrast to previous reports, we show that JNK inhibition by the inhibitor SP600125 is not responsible for VSV attenuation in HCC cells and that this compound acts by causing a posttranslational modification of the viral glycoprotein.


Asunto(s)
Antracenos/farmacología , Antivirales/farmacología , Glicoproteínas de Membrana/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Vesiculovirus/efectos de los fármacos , Vesiculovirus/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Cricetinae , Activación Enzimática/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Interferón Tipo I/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Vesiculovirus/genética , Virión/efectos de los fármacos , Liberación del Virus/efectos de los fármacos
6.
PLoS One ; 5(6): e10988, 2010 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-20539760

RESUMEN

The intrinsic oncolytic specificity of vesicular stomatitis virus (VSV) is currently being exploited to develop alternative therapeutic strategies for hepatocellular carcinoma (HCC). Identifying key regulators in diverse transduction pathways that define VSV oncolysis in cancer cells represents a fundamental prerequisite to engineering more effective oncolytic viral vectors and adjusting combination therapies. After having identified defects in the signalling cascade of type I interferon induction, responsible for attenuated antiviral responses in human HCC cell lines, we have now investigated the role of cell proliferation and translation initiation. Cell cycle progression and translation initiation factors eIF4E and eIF2Bepsilon have been recently identified as key regulators of VSV permissiveness in T-lymphocytes and immortalized mouse embryonic fibroblasts, respectively. Here, we show that in HCC, decrease of cell proliferation by cell cycle inhibitors or siRNA-mediated reduction of G(1) cyclin-dependent kinase activities (CDK4) or cyclin D1 protein expression, do not significantly alter viral growth. Additionally, we demonstrate that translation initiation factors eIF4E and eIF2Bepsilon are negligible in sustaining VSV replication in HCC. Taken together, these results indicate that cellular proliferation and the initiation phase of cellular protein synthesis are not essential for successful VSV oncolysis of HCC. Moreover, our observations indicate the importance of cell-type specificity for VSV oncolysis, an important aspect to be considered in virotherapy applications in the future.


Asunto(s)
Carcinoma Hepatocelular/terapia , Ciclo Celular , Neoplasias Hepáticas/terapia , Viroterapia Oncolítica , Vesiculovirus/fisiología , Animales , Western Blotting , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Citometría de Flujo , Humanos , Neoplasias Hepáticas/patología , Ratones , ARN Interferente Pequeño , Replicación Viral
7.
Mol Ther ; 18(2): 275-84, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19809404

RESUMEN

Newcastle disease virus (NDV) is an intrinsically tumor-specific virus, which is currently under investigation as a clinical oncolytic agent. Several clinical trials have reported NDV to be a safe and effective agent for cancer therapy; however, there remains a clear need for improvement in therapeutic outcome. The endogenous NDV fusion (F) protein directs membrane fusion, which is required for virus entry and cell-cell fusion. Here, we report a novel NDV vector harboring an L289A mutation within the F gene, which resulted in enhanced fusion and cytotoxicity of hepatocellular carcinoma (HCC) cells in vitro, as compared with the rNDV/F3aa control virus. In vivo administration of the recombinant vector, termed rNDV/F3aa(L289A), via hepatic arterial infusion in immune-competent Buffalo rats bearing multifocal, orthotopic liver tumors resulted in tumor-specific syncytia formation and necrosis, with no evidence of toxicity to the neighboring hepatic parenchyma. Furthermore, the improved oncolysis conferred by the L289A mutation translated to significantly prolonged survival compared with control NDV. Taken together, rNDV/F(L289A) represents a safe, yet more effective vector than wild-type NDV for the treatment of HCC, making it an ideal candidate for clinical application in HCC patients.


Asunto(s)
Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Virus de la Enfermedad de Newcastle/fisiología , Viroterapia Oncolítica/métodos , Animales , Línea Celular , Línea Celular Tumoral , Pollos , Vectores Genéticos/efectos de los fármacos , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Interferón Tipo I/farmacología , Masculino , Virus de la Enfermedad de Newcastle/efectos de los fármacos , Virus de la Enfermedad de Newcastle/genética , Ratas
8.
Hepatology ; 48(6): 1864-73, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19003878

RESUMEN

UNLABELLED: Oncolytic virotherapy is a promising strategy for safe and effective treatment of malignancy. We have reported previously that recombinant vesicular stomatitis virus (VSV) vectors are effective oncolytic agents that can be safely administered via the hepatic artery in immunocompetent rats to treat multifocal hepatocellular carcinoma (HCC), resulting in tumor necrosis and prolonged survival. Though the results were encouraging, complete tumor regression was not observed, which led us to explore alternative approaches to further enhance the efficacy of VSV treatment. Transarterial embolization techniques have been shown to improve the efficiency and tumor selectivity of anticancer treatments. Degradable starch microspheres (DSM) are one such embolic agent that provides transient embolization of the therapeautic agent before being degraded by serum amylases. Here we demonstrate via dynamic contrast-enhanced magnetic resonance imaging that in our rat model of multifocal HCC, DSM injection into the hepatic artery results in a substantial reduction in tumor perfusion of systemically applied contrast agent. VSV, when administered in combination with DSM, results in enhanced tumor necrosis and synergistically prolongs survival when compared with VSV or DSM monotherapy. CONCLUSION: This regimen of viroembolization represents an innovative therapeutic modality that can augment the future development of transarterial oncolytic virus therapy for patients with advanced HCC.


Asunto(s)
Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/virología , Embolización Terapéutica/métodos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/virología , Viroterapia Oncolítica/métodos , Animales , Apoptosis , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Arteria Hepática , Neoplasias Hepáticas/patología , Masculino , Microesferas , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Ratas , Ratas Endogámicas BUF , Almidón , Vesiculovirus/genética
9.
Mol Ther ; 16(11): 1789-97, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18781139

RESUMEN

The intrinsic oncolytic specificity of vesicular stomatitis virus (VSV) is currently being exploited to develop alternative therapeutic strategies for hepatocellular carcinoma (HCC). We have observed earlier that, in contrast to cultured human HCC cells, primary human hepatocytes (PHHs) are refractory to VSV infection. Impairment of the type I interferon (IFN) pathway in HCC cells has been suggested to be the mechanism by which these cells become susceptible to VSV infection. The goal of this study was to elucidate the nature of the IFN defect in human HCC. We demonstrate here that the defect in IFN-beta signaling in HCC cells results from a deregulated IFN regulatory factor-3 (IRF3) pathway. Expression of IRF3-spliced variant (IRF3-nirs3) was constitutively observed in HCC cells and, importantly, also in primary HCC samples. In contrast, IRF3 was readily activated in PHHs after stimulation with dsRNA or infection with VSV. In addition, overexpression of IRF3-nirs3 significantly abrogated the IFN-beta response to VSV infection and improved viral growth. Our data provide evidence that aberrant splicing of IRF3 in HCC contributes to the defect in IFN-mediated antiviral defenses. This work may provide a potential molecular basis for selecting HCC patients for oncolytic VSV therapy in future clinical trials.


Asunto(s)
Empalme Alternativo , Factor 3 Regulador del Interferón/biosíntesis , Interferón beta/biosíntesis , Virus Oncolíticos/genética , Vesiculovirus/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Factor 3 Regulador del Interferón/genética , Interferón beta/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , Virus Oncolíticos/fisiología , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , ARN Bicatenario/fisiología , Vesiculovirus/fisiología , Replicación Viral
10.
Mol Ther ; 16(11): 1789-1797, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28189006

RESUMEN

The intrinsic oncolytic specificity of vesicular stomatitis virus (VSV) is currently being exploited to develop alternative therapeutic strategies for hepatocellular carcinoma (HCC). We have observed earlier that, in contrast to cultured human HCC cells, primary human hepatocytes (PHHs) are refractory to VSV infection. Impairment of the type I interferon (IFN) pathway in HCC cells has been suggested to be the mechanism by which these cells become susceptible to VSV infection. The goal of this study was to elucidate the nature of the IFN defect in human HCC. We demonstrate here that the defect in IFN-ß signaling in HCC cells results from a deregulated IFN regulatory factor-3 (IRF3) pathway. Expression of IRF3-spliced variant (IRF3-nirs3) was constitutively observed in HCC cells and, importantly, also in primary HCC samples. In contrast, IRF3 was readily activated in PHHs after stimulation with dsRNA or infection with VSV. In addition, overexpression of IRF3-nirs3 significantly abrogated the IFN-ß response to VSV infection and improved viral growth. Our data provide evidence that aberrant splicing of IRF3 in HCC contributes to the defect in IFN-mediated antiviral defenses. This work may provide a potential molecular basis for selecting HCC patients for oncolytic VSV therapy in future clinical trials.

11.
J Virol ; 79(9): 5507-15, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15827165

RESUMEN

Human plasmacytoid dendritic cells (PDC) are key sentinels alerting both innate and adaptive immune responses through production of huge amounts of alpha/beta interferon (IFN). IFN induction in PDC is triggered by outside-in signal transduction pathways through Toll-like receptor 7 (TLR7) and TLR9 as well as by recognition of cytosolic virus-specific patterns. TLR7 and TLR9 ligands include single-stranded RNA and CpG-rich DNA, respectively, as well as synthetic derivatives thereof which are being evaluated as therapeutic immune modulators promoting Th1 immune responses. Here, we identify the first viruses able to block IFN production by PDC. Both TLR-dependent and -independent IFN responses are abolished in human PDC infected with clinical isolates of respiratory syncytial virus (RSV), RSV strain A2, and measles virus Schwarz, in contrast to RSV strain Long, which we previously identified as a potent IFN inducer in human PDC (Hornung et al., J. Immunol. 173:5935-5943, 2004). Notably, IFN synthesis of PDC activated by the TLR7 and TLR9 agonists resiquimod (R848) and CpG oligodeoxynucleotide 2216 is switched off by subsequent infection by RSV A2 and measles virus. The capacity of RSV and measles virus of human PDC to shut down IFN production should contribute to the characteristic features of these viruses, such as Th2-biased immune pathology, immune suppression, and superinfection.


Asunto(s)
Células Dendríticas/virología , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Virus del Sarampión/patogenicidad , Glicoproteínas de Membrana/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Virus Sincitiales Respiratorios/patogenicidad , Células Dendríticas/inmunología , Humanos , Imidazoles/farmacología , Interferón-alfa/antagonistas & inhibidores , Interferón beta/antagonistas & inhibidores , Virus del Sarampión/inmunología , Glicoproteínas de Membrana/agonistas , Oligodesoxirribonucleótidos/farmacología , Receptores de Superficie Celular/agonistas , Virus Sincitiales Respiratorios/inmunología , Factores de Tiempo , Receptor Toll-Like 7 , Receptor Toll-Like 9 , Receptores Toll-Like
12.
J Gen Virol ; 85(Pt 4): 775-786, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15039520

RESUMEN

Mucosal epithelia are invaded from the apical surface during a primary infection by herpes simplex virus type 1 (HSV-1). HSV-1 progeny virus, synthesized from latently infected peripheral neurons that innervate such epithelia, reinfects the epithelia most likely from the basolateral surface. The epithelial cell lines MDCK and Caco-2 can be induced in vitro to differentiate into polarized cells with distinct apical and plasma membrane domains separated by tight junctions if they are cultured on porous membrane filters. Our data using these culture systems showed that highly polarized epithelial cells were not susceptible to apical HSV-1 infection. However, HSV-1 infected these cells if added from the basolateral surface or if a depletion of extracellular Ca(2+) had weakened the strength of the cell-cell contacts. Basolateral infection and apical infection after the Ca(2+) switch required an intact microtubule network for genome targeting to the nucleus. This system can be used to identify the microtubule motors that HSV-1 uses during virus entry in polarized epithelial cells.


Asunto(s)
Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/patogenicidad , Animales , Células CACO-2 , Señalización del Calcio , Adhesión Celular , Moléculas de Adhesión Celular/fisiología , Diferenciación Celular , Línea Celular , Polaridad Celular , Chlorocebus aethiops , Cricetinae , Perros , Células Epiteliales/citología , Células Epiteliales/fisiología , Células Epiteliales/virología , Humanos , Microtúbulos/fisiología , Nectinas , Receptores de Superficie Celular/fisiología , Receptores Virales/fisiología , Células Vero
13.
J Virol ; 77(16): 8661-8, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12885884

RESUMEN

We have previously shown that the nonstructural (NS) proteins NS1 and NS2 of bovine respiratory syncytial virus (BRSV) mediate resistance to the alpha/beta interferon (IFN)-mediated antiviral response. Here, we show that they, in addition, are able to prevent the induction of beta IFN (IFN-beta) after virus infection or double-stranded RNA stimulation. In BRSV-infected MDBK cells upregulation of IFN-stimulated genes (ISGs) such as MxA did not occur, although IFN signaling via JAK/STAT was found intact. In contrast, infection with recombinant BRSVs lacking either or both NS genes resulted in efficient upregulation of ISGs. Biological IFN activity and IFN-beta were detected only in supernatants of cells infected with the NS deletion mutants but not with wild-type (wt) BRSV. Subsequent analyses of IFN-beta promoter activity showed that infection of cells with the double deletion mutant BRSV DeltaNS1/2, but not with BRSV wt, resulted in a significant increase in IFN-beta gene promoter activity. Induction of the IFN-beta promoter depends on the activation of three distinct transcription factors, NF-kappaB, ATF-2/c-Jun, and IFN regulatory factor 3 (IRF-3). Whereas NF-kappaB and ATF-2/c-Jun activities were readily detectable and comparable in both wt BRSV- and BRSV DeltaNS1/2-infected cells, phosphorylation and transcriptional activity of IRF-3, however, were observed only after BRSV DeltaNS1/2 infection. NS protein-mediated inhibition of IRF-3 activation and IFN induction should have considerable impact on the pathogenesis and immunogenicity of BRSV.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Virus Sincitial Respiratorio Bovino/fisiología , Factores de Transcripción/antagonistas & inhibidores , Proteínas no Estructurales Virales/fisiología , Activación Viral/fisiología , Animales , Línea Celular , Proteínas de Unión al ADN/fisiología , Eliminación de Gen , Factores Reguladores del Interferón , Interferón beta/genética , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Transcripción/fisiología , Proteínas no Estructurales Virales/genética , Proteínas Virales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA