Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 31(4): 898-907, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21233447

ABSTRACT

OBJECTIVE: Smad-interacting protein-1 (Sip1/ZEB2) is a transcriptional repressor of the telomerase reverse transcriptase catalytic subunit (Tert) and has recently been identified as a key regulator of embryonic cell fate with a phenotypic effect similar, in our opinion, to that reported for nitric oxide (NO). Remarkably, SIP1/ZEB2 is a known target of the microRNA 200 (miR-200) family. In this light, we postulated that Sip1/ZEB2 and the miR-200 family could play a role during the NO-dependent differentiation of mES. METHODS AND RESULTS: The results of the present study show that Sip1/ZEB2 expression is downregulated during the NO-dependent expression of mesendoderm and early cardiovascular precursor markers, including Flk1 and CXCR4 in mES. Coincidently, members of the miR-200 family, namely miR-429, -200a, -200b, and -200c, were transcriptionally induced in parallel to mouse Tert. This regulation occurred at the level of chromatin. Remarkably, miR-429/miR-200a overexpression or Sip1/ZEB2 knockdown by short hairpin RNA interference elicited a gene expression pattern similar to that of NO regardless of the presence of leukemia inhibitory factor. CONCLUSIONS: These results are the first demonstrating that the miR-200 family and Sip1/ZEB2 transcription factor are regulated by NO, indicating an unprecedented molecular circuitry important for telomerase regulation and early differentiation of mES.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Nitric Oxide/metabolism , Repressor Proteins/metabolism , Signal Transduction , Animals , Cell Differentiation/drug effects , Cells, Cultured , Chromatin Assembly and Disassembly , Embryonic Stem Cells/drug effects , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Leukemia Inhibitory Factor/metabolism , Mice , Nitric Oxide Donors/pharmacology , RNA Interference , RNA, Messenger/metabolism , Repressor Proteins/genetics , Signal Transduction/drug effects , Telomerase/metabolism , Time Factors , Transcription, Genetic , Transfection , Zinc Finger E-box Binding Homeobox 2
2.
J Cell Physiol ; 222(2): 401-10, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19890834

ABSTRACT

The capability of PARP activity inhibitors to prevent DNA damage recovery suggested the use of these drugs as chemo- and radio-sensitisers for cancer therapy. Our research, carried out on cultured human M14 melanoma cells, was aimed to examine if PJ-34, a potent PARP activity inhibitor of second generation, was per se able to affect the viability of these cancer cells without any DNA damaging agents. Using time-lapse videomicroscopy, we evidenced that 10 microM PJ-34 treatment induced severe mitotic defects leading to dramatic reduction of cell proliferation and to cell death. PJ-34 cytotoxic effect was further confirmed by analysis of cell viability and clonogenic assay. Absence of canonic apoptosis markers allowed us to exclude this kind of cell death. No single and/or double stranded DNA damage was evidenced. Immunofluorescence analysis showed an aberrant mitotic scenario in several cells and subsequent multinucleation suggesting an atypical way for cells to die: the mitotic catastrophe. The detection of aberrant accumulation of polymerised actin inside the nucleolus was noteworthy. Taken together, our results demonstrate that, targeting PARP activity by PJ-34, cancer cell survival is affected independently of DNA damage repair. Two findings are remarkable: (a) cisplatin concentration can be reduced by three quarters if it is followed by treatment with 10 microM PJ-34 for 24 h to obtain the same cytotoxic effect; (b) effects dependent on PJ-34 treatment are reversible. Our data suggest that, to reduce the harm done to non-tumour cells during chemotherapy with cisplatin, the latter could be coupled with PJ-34 treatment.


Subject(s)
Actins/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Nucleolus/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Melanoma/enzymology , Mitosis/drug effects , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Cell Death , Cell Line, Tumor , Cell Nucleolus/metabolism , Cell Survival/drug effects , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Humans , Melanoma/pathology , Microscopy, Video , Poly(ADP-ribose) Polymerases/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...