Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 789, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964993

ABSTRACT

Several studies have linked the E3 ubiquitin ligase TRIP12 (Thyroid hormone Receptor Interacting Protein 12) to the cell cycle. However, the regulation and the implication of this protein during the cell cycle are largely unknown. In this study, we show that TRIP12 expression is regulated during the cell cycle, which correlates with its nuclear localization. We identify an euchromatin-binding function of TRIP12 mediated by a N-terminal intrinsically disordered region. We demonstrate the functional implication of TRIP12 in the mitotic entry by controlling the duration of DNA replication that is independent from its catalytic activity. We also show the requirement of TRIP12 in the mitotic progression and chromosome stability. Altogether, our findings show that TRIP12 is as a new chromatin-associated protein with several implications in the cell cycle progression and in the maintenance of genome integrity.


Subject(s)
Carrier Proteins/metabolism , Chromosomal Instability , Ubiquitin-Protein Ligases/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle/physiology , Chromosome Segregation , DNA Replication , Euchromatin/genetics , Euchromatin/metabolism , Gene Expression Regulation , Humans , Mice, SCID , Mitosis , Protein Domains , Ubiquitin-Protein Ligases/genetics
2.
Cancer Gene Ther ; 17(5): 315-24, 2010 May.
Article in English | MEDLINE | ID: mdl-19911032

ABSTRACT

Pancreatic cancer (PC) remains a life-threatening disease. Efficient therapeutic gene delivery to PC-derived cells continues to present challenges. We used self-inactivated lentiviral vectors to transduce PC-derived cells in vitro and in vivo. We showed that lentiviral vectors transduce PC-derived cell lines with high efficiency (>90%), regardless of the differentiation state of the cell. Next, we transferred human interferon beta (hIFN-beta) gene. Expression of hIFN-beta in PC cells using lentiviral vectors resulted in the inhibition of cell proliferation and the induction of cell death by apoptosis. In vivo, lentiviral administration of hIFN-beta prevented PC tumor progression for up to 15 days following gene therapy, and induced tumor regression/stabilization in 50% of the mice treated. Again, hIFN-beta expression resulted in cancer cell proliferation inhibition and apoptosis induction. We provide evidence that human immunodeficiency virus (HIV)-1-based lentiviral vectors are very efficient for gene transfer in PC-derived cells in vitro and in vivo. As a consequence, delivery of hIFN-beta stopped PC tumor progression. Thus, our approach could be applied to the 85% of PC patients with a locally advanced disease.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Lentivirus/genetics , Pancreatic Neoplasms/therapy , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Humans , Interferon-beta/genetics , Interferon-beta/physiology , Mice , Mice, SCID , Transduction, Genetic , Xenograft Model Antitumor Assays
3.
Cancer Gene Ther ; 14(1): 19-29, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16990845

ABSTRACT

Pancreatic cancer is one of the most aggressive and devastating human malignancies. There is an urgent need for more effective therapy for patients with advanced disease. In this context, genetic therapy potentially represents a rational new approach to treating pancreatic cancer, which could provide an adjunct to conventional options. Because of the promise of recombinant SV40 vectors, we tested their ability to deliver a transgene, and to target a transcript, so as to inhibit pancreatic tumors growth in vivo. BxPC3 and Capan-1 cells were efficiently transduced using SV40 vectors without selection, as compared to synthetic vectors PEI. SV40 vectors were as efficient as adenoviral vectors, and provided long-term transgene expression. Next, we devised a SV40-derived, targeted gene therapy approach of pancreatic cancer, by combining hTR tumor-specific promoter with sst2 somatostatin receptor tumor-suppressor gene. In vitro cell proliferation was strongly impaired following administration of SV(hTR-sst2). SV40-derived sst2-mediated antiproliferative effect was dependent on the local production of somatostatin. In vivo, intratumoral gene transfer of sst2 using rSV40 vectors resulted in a marked inhibition of Capan-1 tumor progression, and proliferation. These results represent the initial steps toward a novel approach to the gene therapy of pancreatic cancer using SV40 as a vector.


Subject(s)
Defective Viruses/physiology , Gene Transfer Techniques , Pancreatic Neoplasms/pathology , Simian virus 40/physiology , Virus Replication , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Polymerase Chain Reaction , Transduction, Genetic
4.
Gut ; 56(1): 107-14, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16891358

ABSTRACT

BACKGROUND: Peritoneal carcinomatosis from pancreatic cancer has a poor prognosis with a median survival of 3.1 months. This is mainly due to lack of effective treatment. Interleukin 12 (IL12) is a proinflammatory cytokine that has a potent antitumoral effect by stimulating innate and adoptive immunity. AIM: To examine the antitumoral effect and toxicity of intraperitoneal delivery of IL12 using an ex vivo gene therapy approach in a murine model of pancreatic peritoneal carcinomatosis. METHODS: Peritoneal carcinomatosis was generated by direct intraperitoneal inoculation of the pancreatic cancer cell line Capan-1 in athymic mice. Syngenic fibroblasts were genetically modified in vitro to secrete IL12 using a polycistronic TFG murine IL12 retroviral vector coding for both p35 and p40 murine IL12 subunits. Ex vivo gene therapy involved injection of the genetically modified fibroblasts intraperitoneally twice a week for 4 weeks. RESULTS: Treatment of pre-established peritoneal carcinomatosis with fibroblasts genetically modified to express IL12 induced a marked inhibition of tumour growth as measured by comparison of the weights of the intraperitoneal tumour nodules in the treated and control animals (3.52 (SD 0.47) v 0.93 (SD 0.21) g, p<0.05) and improved survival. This effect was associated with infiltration of the peritoneal tumour nodules with macrophages. Peritoneal lavage confirmed enhancement of the innate peritoneal inflammatory activity, with an increased number of activated macrophages and natural killer cells. Moreover, macrophages harvested from animals with peritoneal carcinomatosis and treated with IL12-expressing fibroblasts expressed an activated proinflammatory antitumoral M1 phenotype that included strongly enhanced reactive oxygen species and nitric oxide production. There was no treatment-related toxicity. CONCLUSION: Multiple injections of genetically modified fibroblasts to express IL12 is an effective and well-tolerated treatment for experimental murine pancreatic peritoneal carcinomatosis via activated innate immunity and in particular activated M1 macrophages.


Subject(s)
Antineoplastic Agents/immunology , Fibroblasts/immunology , Genetic Therapy/methods , Interleukin-12/immunology , Peritoneal Neoplasms/therapy , Animals , Cell Division/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Flow Cytometry/methods , Immunity, Innate/immunology , Immunohistochemistry/methods , Injections, Intraperitoneal , Interleukin-12/administration & dosage , Interleukin-12/genetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Nitric Oxide/biosynthesis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/immunology , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...