Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cancer Sci ; 115(2): 600-610, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38037288

RÉSUMÉ

Oncolytic viruses have two anticancer functions: direct oncolysis and elicitation of antitumor immunity. We previously developed a novel fusogenic oncolytic vaccinia virus (FUVAC) from a non-fusogenic vaccinia virus (VV) and, by remodeling the tumor immune microenvironment, we demonstrated that FUVAC induced stronger oncolysis and antitumor immune responses compared with non-fusogenic VV. These functions depend strongly on cell-cell fusion induction. However, FUVAC tends to have decreased fusion activity in cells with low virus replication efficacy. Therefore, another combination strategy was required to increase cell-cell fusion in these cells. Histone deacetylase (HDAC) inhibitors suppress the host virus defense response and promote viral replication. Therefore, in this study, we selected an HDAC inhibitor, trichostatin A (TSA), as the combination agent for FUVAC to enhance its fusion-based antitumor potential. TSA was added prior to FUVAC treatment of murine tumor B16-F10 and CT26 cells. TSA increased the replication of both FUVAC and parental non-fusogenic VV. Moreover, TSA enhanced cell-cell fusion and FUVAC cytotoxicity in these tumor cells in a dose-dependent manner. Transcriptome analysis revealed that TSA-treated tumors showed altered expression of cellular component-related genes, which may affect fusion tolerance. In a bilateral tumor-bearing mouse model, combination treatment of TSA and FUVAC significantly prolonged mouse survival compared with either treatment alone or in combination with non-fusogenic VV. Our findings demonstrate that TSA is a potent enhancer of cell-cell fusion efficacy of FUVAC.


Sujet(s)
Tumeurs , Thérapie virale de cancers , Virus oncolytiques , Souris , Animaux , Inhibiteurs de désacétylase d'histone/pharmacologie , Virus de la vaccine/génétique , Virus de la vaccine/métabolisme , Fusion cellulaire , Tumeurs/génétique , Lignée cellulaire tumorale , Microenvironnement tumoral
2.
Cells ; 10(5)2021 04 23.
Article de Anglais | MEDLINE | ID: mdl-33922406

RÉSUMÉ

Engineered vaccinia virus serves as an oncolytic virus for cancer virotherapy. We evaluated the oncolytic characteristics of VGF- and O1-deleted recombinant mitogen-activated protein kinase (MAPK)-dependent vaccinia virus (MDRVV). We found that compared with viruses with the deletion of either gene alone, MDRVV is more attenuated in normal cells and can replicate in cancer cells that exhibit constitutive ERK1/2 activation in the MAPK pathway. We armed MDRVV with a bifunctional fusion gene encoding cytosine deaminase and uracil phosphoribosyltransferase (CD/UPRT), which converts 5-fluorocytosine (5-FC) into chemotherapeutic agents, and evaluated its oncolytic activity alone or in combination with 5-FC in human pancreatic cancer cell lines, tumor mouse models of peritoneal dissemination and liver metastasis, and ex vivo-infected live pancreatic cancer patient-derived tissues. CD/UPRT-armed MDRVV alone could efficiently eliminate pancreatic cancers, and its antitumor effects were partially enhanced in combination with 5-FC in vitro and in vivo. Moreover, the replication of MDRVV was detected in tumor cells of patient-derived, surgically resected tissues, which showed enlarged nuclei and high expression of pERK1/2 and Ki-67, and not in stromal cells. Our findings suggest that systemic injections of CD/UPRT-armed MDRVV alone or in combination with 5-FC are promising therapeutic strategies for pancreatic ductal adenocarcinoma.


Sujet(s)
Cytosine deaminase/métabolisme , Flucytosine/pharmacologie , Mitogen-Activated Protein Kinases/métabolisme , Thérapie virale de cancers/méthodes , Tumeurs du pancréas/thérapie , Pentosyltransferases/métabolisme , Virus de la vaccine/génétique , Animaux , Apoptose , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/thérapie , Prolifération cellulaire , Association thérapeutique , Cytosine deaminase/génétique , Femelle , Humains , Tumeurs du foie/génétique , Tumeurs du foie/secondaire , Tumeurs du foie/thérapie , Souris , Souris SCID , Mitogen-Activated Protein Kinases/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Pentosyltransferases/génétique , Tumeurs du péritoine/génétique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/thérapie , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
3.
Mol Ther ; 29(5): 1782-1793, 2021 05 05.
Article de Anglais | MEDLINE | ID: mdl-33348052

RÉSUMÉ

Oncolytic viruses induce antitumor immunity following direct viral oncolysis. However, their therapeutic effects are limited in distant untreated tumors because their antitumor function depends on indirect antitumor immunity. Here, we generated a novel fusogenic oncolytic vaccinia virus (FUVAC) and compared its antitumor activity with that of its parental non-fusogenic virus. Compared with the parent, FUVAC exerted the cytopathic effect and induced immunogenic cell death in human and murine cancer cells more efficiently. In a bilateral tumor-bearing syngeneic mouse model, FUVAC administration significantly inhibited tumor growth in both treated and untreated tumors. However, its antitumor effects were completely suppressed by CD8+ T cell depletion. Notably, FUVAC reduced the number of tumor-associated immune-suppressive cells in treated tumors, but not in untreated tumors. Mice treated with FUVAC before an immune checkpoint inhibitor (ICI) treatment achieved complete response (CR) in both treated and untreated tumors, whereas ICI alone did not show antitumor activity. Mice achieving CR rejected rechallenge with the same tumor cells, suggesting establishment of a long-term tumor-specific immune memory. Thus, FUVAC improves the tumor immune microenvironment and enhances systemic antitumor immunity, suggesting that, alone and in combination with ICI, it is a novel immune modulator for overcoming oncolytic virus-resistant tumors.


Sujet(s)
Tumeurs du côlon/thérapie , Inhibiteurs de points de contrôle immunitaires/administration et posologie , Virus de la vaccine/physiologie , Cellules A549 , Animaux , Lignée cellulaire tumorale , Tumeurs du côlon/immunologie , Association thérapeutique , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Souris , Mutation , Thérapie virale de cancers , Virus oncolytiques/physiologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Virus de la vaccine/génétique , Pénétration virale , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Sci Transl Med ; 12(526)2020 01 15.
Article de Anglais | MEDLINE | ID: mdl-31941828

RÉSUMÉ

The immune status of the tumor microenvironment is a key indicator in determining the antitumor effectiveness of immunotherapies. Data support the role of activation and expansion of tumor-infiltrating lymphocytes (TILs) in increasing the benefit of immunotherapies in patients with solid tumors. We found that intratumoral injection of a tumor-selective oncolytic vaccinia virus encoding interleukin-7 (IL-7) and IL-12 into tumor-bearing immunocompetent mice activated the inflammatory immune status of previously poorly immunogenic tumors and resulted in complete tumor regression, even in distant tumor deposits. Mice achieving complete tumor regression resisted rechallenge with the same tumor cells, suggesting establishment of long-term tumor-specific immune memory. Combining this virotherapy with anti-programmed cell death-1 (PD-1) or anti-cytotoxic T lymphocyte antigen 4 (CTLA4) antibody further increased the antitumor activity as compared to virotherapy alone, in tumor models unresponsive to either of the checkpoint inhibitor monotherapies. These findings suggest that administration of an oncolytic vaccinia virus carrying genes encoding for IL-7 and IL-12 has antitumor activity in both directly injected and distant noninjected tumors through immune status changes rendering tumors sensitive to immune checkpoint blockade. The benefit of intratumoral IL-7 and IL-12 expression was also observed in humanized mice bearing human cancer cells. These data support further investigation in patients with non-inflamed solid tumors.


Sujet(s)
Interleukine-12/métabolisme , Interleukine-7/métabolisme , Virus oncolytiques/génétique , Animaux , Antigène CTLA-4/immunologie , Femelle , Inhibiteurs de points de contrôle immunitaires , Souris , Virus de la vaccine/génétique
5.
Biochem Biophys Res Commun ; 516(3): 831-838, 2019 08 27.
Article de Anglais | MEDLINE | ID: mdl-31262449

RÉSUMÉ

The promising anti-tumor effects of oncolytic vaccinia virus (OVV) have been demonstrated. Further, we previously showed that long non-coding RNA (lncRNA) urothelial carcinoma-associated 1 (UCA1) enhances OVV cell-to-cell spread via the activation of Cdc42 in ovarian cancer. However, its role in other cancer types and the molecular mechanism underlying its effects remain to be explored. In this study, we first demonstrated that UCA1 upregulates OVV cell-to-cell spread but not its binding, entry, and replication in colorectal cancer cells. Functional analysis indicated that Cdc42 activation and filopodia formation play an important role in this process. Moreover, expression analysis of various miRNAs suggested that UCA1 inhibits both miR-18a and miR-182, thereby promoting Cdc42 activation, which in turn, regulates OVV cell-to-cell spread. Furthermore, UCA1 was found to modulate tumor malignancy, drug resistance, and sensitivity to OVV via different miRNAs in colorectal cancer. These findings indicate that a three-marker panel, which includes UCA1 expression, Cdc42 activation, and filopodia formation, could potentially be used to predict the therapeutic effect of OVV in colorectal cancer.


Sujet(s)
Régulation de l'expression des gènes tumoraux , microARN/génétique , ARN long non codant/génétique , Virus de la vaccine/génétique , Protéine G cdc42/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Cellules Caco-2 , Prolifération cellulaire , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/thérapie , Cellules HCT116 , Cellules HT29 , Humains , microARN/métabolisme , Thérapie virale de cancers/méthodes , Virus oncolytiques/génétique , Virus oncolytiques/métabolisme , Pseudopodes/métabolisme , Pseudopodes/anatomopathologie , ARN long non codant/métabolisme , Transduction du signal , Virus de la vaccine/métabolisme , Réplication virale , Protéine G cdc42/métabolisme
6.
Mol Ther Oncolytics ; 14: 159-171, 2019 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-31236440

RÉSUMÉ

Vaccinia virus (VV) has been utilized in oncolytic virotherapy, but it risks a host antiviral immune response. VV has an extracellular enveloped virus (EEV) form consisting of a normal virion covered with a host-derived outer membrane that enables its spread via circulation while evading host immune mechanisms. However, the immune resistance of EEV is only partial, owing to expression of the surface protein B5R, which has four short consensus repeat (SCR) domains that are targeted by host immune factors. To engineer a more effective virus for oncolytic virotherapy, we developed an enhanced immune-evading oncolytic VV by removing the SCRs from the attenuated strain LC16mO. Although deletion of only the SCRs preserved viral replication, progeny production, and oncolytic activity, deletion of whole B5R led to attenuation of the virus. Importantly, SCR-deleted EEV had higher neutralization resistance than did B5R-wild-type EEV against VV-immunized animal serum; moreover, it retained oncolytic function, thereby prolonging the survival of tumor-bearing mice treated with anti-VV antibody. These results demonstrate that partial SCR deletion increases neutralization escape without affecting the oncolytic potency of VV, making it useful for the treatment of tumors under the anti-virus antibody existence.

7.
Mol Ther Oncolytics ; 13: 35-48, 2019 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-31011626

RÉSUMÉ

Oncolytic vaccinia virus (OVV) has demonstrated appropriate safety profiles for clinical development. Although designed to kill cancer cells efficiently, OVV sensitivity varies in individual cancers, and predictive biomarkers of therapeutic responses have not been identified. Here we found that OVV was much more efficient in KFTX paclitaxel-resistant ovarian cancer cells compared to that in KFlow paclitaxel-sensitive cells. Microarray analysis identified long non-coding RNA urothelial carcinoma-associated 1 (UCA1) upregulation, which contributed to both enhanced paclitaxel resistance and OVV spread. In addition, UCA1 expression correlated with efficient OVV spread in other ovarian cell lines and primary cancer cell cultures. When host pathways underlying OVV spread were analyzed, differences were detected in the activation of the Rho GTPase Cdc42, suggesting that filopodia formation enhances OVV cell-to-cell spread and tumor migration. Moreover, we established a clinically relevant mouse model of peritoneal metastasis using KFTX or KFlow cells. Paclitaxel exerted anti-tumor effects on KFlow, but not KFTX, tumors. In mice bearing KFTX cells after paclitaxel failure, OVV treatment induced the regression of residual tumors and improved survival. Our findings demonstrated that UCA1 promotes OVV cell-to-cell spread in ovarian cancer, resulting in enhanced therapeutic outcome.

8.
BMC Biotechnol ; 15: 58, 2015 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-26088202

RÉSUMÉ

BACKGROUND: Human artificial chromosome (HAC) vectors have some unique characteristics as compared with conventional vectors, carrying large transgenes without size limitation, showing persistent expression of transgenes, and existing independently from host genome in cells. With these features, HACs are expected to be promising vectors for modifications of a variety of cell types. However, the method of introduction of HACs into target cells is confined to microcell-mediated chromosome transfer (MMCT), which is less efficient than other methods of vector introduction. Application of Measles Virus (MV) fusogenic proteins to MMCT instead of polyethylene glycol (PEG) has partly solved this drawback, whereas the tropism of MV fusogenic proteins is restricted to human CD46- or SLAM-positive cells. RESULTS: Here, we show that retargeting of microcell fusion by adding anti-Transferrin receptor (TfR) single chain antibodies (scFvs) to the extracellular C-terminus of the MV-H protein improves the efficiency of MV-MMCT to human fibroblasts which originally barely express both native MV receptors, and are therefore resistant to MV-MMCT. Efficacy of chimeric fusogenic proteins was evaluated by the evidence that the HAC, tagged with a drug-resistant gene and an EGFP gene, was transferred from CHO donor cells into human fibroblasts. Furthermore, it was demonstrated that no perturbation of either the HAC status or the functions of transgenes was observed on account of retargeted MV-MMCT when another HAC carrying four reprogramming factors (iHAC) was transferred into human fibroblasts. CONCLUSIONS: Retargeted MV-MMCT using chimeric H protein with scFvs succeeded in extending the cell spectrum for gene transfer via HAC vectors. Therefore, this technology could facilitate the systematic cell engineering by HACs.


Sujet(s)
Chromosomes artificiels humains/génétique , Virus de la rougeole/génétique , Protéines de fusion virale/génétique , Animaux , Cellules CHO , Lignée cellulaire , Cricetinae , Cricetulus , Fibroblastes , Techniques de transfert de gènes , Humains , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme
9.
Biochem Biophys Res Commun ; 431(2): 336-41, 2013 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-23291180

RÉSUMÉ

Human artificial chromosome (HAC) has several advantages as a gene therapy vector, including stable episomal maintenance and the ability to carry large gene inserts. Induced pluripotent stem (iPS) cells also have a great potential for gene therapy, which can be generated from an individual's own tissues and contribute to any tissues when reintroduced. A Sendai virus (SeV) vector with reprogramming factors is a powerful tool for generating iPS cells because of the high infection efficiency without the risk of integration into host chromosomes. In this study, we developed an iPS cell-mediated and integration-free coagulation factor VIII (FVIII) expression system using non-integrating SeV- and HAC-vectors. Multiple human FVIII genes, which were under the control of the megakaryocyte-specific platelet factor-4 (PF4) promoter for development of a treatment for hemophilia A, were inserted into a HAC vector (PF4-FVIII-HAC). The PF4-FVIII-HAC was introduced into SeV vector-mediated iPS cells derived from a mouse model of hemophilia A. After in vitro differentiation of iPS cells with the PF4-FVIII-HAC into megakaryocytes/platelets, the PF4-FVIII-HAC resulted in expression of FVIII. This study has developed the iPS cell-mediated PF4-driven FVIII expression system using two non-integrating vectors; therefore, this system may be a promising tool for safer gene- and cell-therapy of hemophilia A.


Sujet(s)
Chromosomes artificiels humains/génétique , Facteur VIII/génétique , Thérapie génétique/méthodes , Hémophilie A/thérapie , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/transplantation , Animaux , Différenciation cellulaire , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Vecteurs génétiques/génétique , Humains , Cellules souches pluripotentes induites/cytologie , Souris , Facteur-4 plaquettaire/génétique , Facteur-4 plaquettaire/métabolisme , Régions promotrices (génétique) , Virus Sendai
10.
PLoS One ; 6(10): e25961, 2011.
Article de Anglais | MEDLINE | ID: mdl-21998730

RÉSUMÉ

Human artificial chromosomes (HACs) have unique characteristics as gene-delivery vectors, including episomal transmission and transfer of multiple, large transgenes. Here, we demonstrate the advantages of HAC vectors for reprogramming mouse embryonic fibroblasts (MEFs) into induced pluripotent stem (iPS) cells. Two HAC vectors (iHAC1 and iHAC2) were constructed. Both carried four reprogramming factors, and iHAC2 also encoded a p53-knockdown cassette. iHAC1 partially reprogrammed MEFs, and iHAC2 efficiently reprogrammed MEFs. Global gene expression patterns showed that the iHACs, unlike other vectors, generated relatively uniform iPS cells. Under non-selecting conditions, we established iHAC-free iPS cells by isolating cells that spontaneously lost iHAC2. Analyses of pluripotent markers, teratomas and chimeras confirmed that these iHAC-free iPS cells were pluripotent. Moreover, iHAC-free iPS cells with a re-introduced HAC encoding Herpes Simplex virus thymidine kinase were eliminated by ganciclovir treatment, indicating that the HAC safeguard system functioned in iPS cells. Thus, the HAC vector could generate uniform, integration-free iPS cells with a built-in safeguard system.


Sujet(s)
Ingénierie cellulaire/méthodes , Chromosomes artificiels humains/génétique , Vecteurs génétiques/génétique , Cellules souches pluripotentes induites/cytologie , Animaux , Cellules CHO , Cricetinae , Cricetulus , Fibroblastes/cytologie , Gènes-suicide transgéniques/génétique , Humains , Cellules souches pluripotentes induites/métabolisme , Souris , Simplexvirus/génétique , Thymidine kinase/génétique
11.
J Hum Genet ; 56(10): 727-33, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21833006

RÉSUMÉ

Human artificial chromosome (HAC) has several advantages as a gene therapy vector, including stable episomal maintenance that avoids insertional mutations and the ability to carry large gene inserts. To examine the copy number effect on the gene expression levels and its stability for a long-term culture for a future application in gene therapy, we constructed a HAC vector carrying the human factor VIII (FVIII) complementary DNA, FVIII-HAC in Chinese hamster ovary (CHO) cells. One and more copies of FVIII gene on the HAC were expressed in the copy-number-dependent manner in the CHO cells. The HAC with 16 copies of FVIII, FVIII (16)-HAC, was transferred from CHO hybrids into a human immortalized mesenchymal stem cell using microcell-mediated chromosome transfer. The expression levels of HAC-derived FVIII transgene products were compared with transfected FVIII plasmids. The former showed expression levels consistent with those of the original clones, even after 50 population doublings, whereas the latter showed a remarkable decrease in expression despite unvarying DNA content, indicating that the gene on the HAC is resistant to gene silencing. These results suggest that the HAC-mediated therapeutic gene-expression system may be a powerful tool for stable expression of transgenes, and possibly for industrial production of gene products.


Sujet(s)
Chromosomes artificiels humains/génétique , Facteur VIII/génétique , Facteur VIII/métabolisme , Techniques de transfert de gènes , Vecteurs génétiques , Cellules souches mésenchymateuses/métabolisme , Animaux , Cellules CHO , Lignée cellulaire , Cricetinae , Cricetulus , Dosage génique , Thérapie génétique/méthodes , Humains , Transgènes/génétique
12.
Exp Anim ; 58(5): 537-42, 2009 Oct.
Article de Anglais | MEDLINE | ID: mdl-19897938

RÉSUMÉ

Although Tritrichomonas muris is a common parasite often detected in experimental animals including mice, its pathogenesis in host animals remains unclear. Proteomics can be used to specifically analyze biochemical host-parasite interaction and immune responses of the host to parasites. However, proteomics have not yet been applied to T. muris studies. Here, the effects of T. muris on the host were analyzed by proteomics. We found that 10 different proteins were expressed in T. muris-infected mice intestines compared with non-infected intestines. The identified proteins represented several functions mainly related to stress, immune response, metabolism and signal transduction. The results suggest that T. muris infection may affect processes that are acclimatizing to the environmental changes caused by the infection in the mouse intestine.


Sujet(s)
Interactions hôte-parasite , Intestins/parasitologie , Parasitoses animales/métabolisme , Protéomique , Trichomonase/métabolisme , Trichomonas/physiologie , Animaux , Muqueuse intestinale/métabolisme , Mâle , Souris , Souris de lignée C57BL , Parasitoses animales/immunologie , Organismes exempts d'organismes pathogènes spécifiques , Trichomonas/pathogénicité , Trichomonase/immunologie
13.
Biochem Biophys Res Commun ; 353(2): 259-67, 2007 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-17188235

RÉSUMÉ

Germline stem (GS) cells can only differentiate into germline cells, while multipotent germ stem (mGS) cells, like embryonic stem (ES) cells, can differentiate into various somatic cells and tissues. The proteomic profiles in GS and mGS cells were compared by two-dimensional gel electrophoresis. Ten down-regulated and 16 up-regulated proteins were differentially expressed in mGS cells in comparison to GS cells, and these proteomic characteristics were very much similar to those in ES cells indicating that multipotency of mGS and ES cells is based on a common molecular event(s). Protein identification by mass spectrometry revealed that these proteins were functionally involved in cell signaling, transcription factors, metabolism, and protein folding. The identified proteins in the present study may thus reveal its biological characteristics and functional property in self-renewal and multipotency.


Sujet(s)
Cellules souches embryonnaires/métabolisme , Régulation de l'expression des gènes au cours du développement/physiologie , Cellules germinales/métabolisme , Cellules souches multipotentes/métabolisme , Protéome/métabolisme , Animaux , Lignée cellulaire , Cellules souches embryonnaires/cytologie , Cellules germinales/cytologie , Souris , Souris de lignée C57BL , Cellules souches multipotentes/cytologie
14.
J Med Virol ; 78(11): 1456-64, 2006 Nov.
Article de Anglais | MEDLINE | ID: mdl-16998876

RÉSUMÉ

Transforming growth factor (TGF)-beta1 induces not only cell growth inhibition or apoptosis but also Epstein-Barr virus (EBV) reactivation in some Burkitt's lymphoma (BL) cell lines. The purpose of this study was to define the role of TGF-beta signaling molecules in response to TGF-beta1-mediated cell growth inhibition, apoptosis, and EBV reactivation in BL cell lines. First, we confirmed the effect of TGF-beta1 on the cell growth and EBV reactivation in six BL cell lines. TGF-beta1 induced cell growth inhibition and EBV reactivation in these cell lines but did not in Akata cells. To elucidate the mechanism of TGF-beta1 unresponsiveness in Akata cells, we studied the expression of TGF-beta receptors and the intracellular signaling molecules Smads. All cell lines expressed TGF-beta type I receptor, Smad2, Smad3, and Smad4. TGF-beta type II receptor (R-II) was expressed in all cell lines except Akata cells. Introduction of the TGF-beta R-II into Akata cells results in sensitivity to TGF-beta1-mediated growth inhibition, apoptosis, and EBV reactivation. In addition, to test a possibility to the transcriptional repression of the TGF-beta R-II gene in Akata cells, the effect of histone deacetylation (HDAC) inhibitor, trichostatin A (TSA) was examined. The expression of TGF-beta R-II in Akata cells was induced by TSA treatment. These results suggest that the lack of functional TGF-beta R-II impedes the progression of signals through TGF-beta1 and becomes a determinant of unresponsiveness to TGF-beta1-mediated growth inhibition and EBV reactivation.


Sujet(s)
Apoptose/physiologie , Herpèsvirus humain de type 4/physiologie , Récepteurs TGF-bêta/génétique , Facteur de croissance transformant bêta-1/métabolisme , Activation virale/physiologie , Lignée cellulaire , Régulation de l'expression des gènes , Humains , Protein-Serine-Threonine Kinases , Récepteur de type II du facteur de croissance transformant bêta , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE