Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Oncogene ; 37(2): 241-254, 2018 01 11.
Article in English | MEDLINE | ID: mdl-28925399

ABSTRACT

Glioblastomas (GBM) are lethal primitive brain tumours characterized by a strong intra-tumour heterogeneity. We observed in GBM tissues the coexistence of functionally divergent micro-territories either enriched in more differentiated and non-mitotic cells or in mitotic undifferentiated OLIG2 positive cells while sharing similar genomic abnormalities. Understanding the formation of such functionally divergent micro-territories in glioblastomas (GBM) is essential to comprehend GBM biogenesis, plasticity and to develop therapies. Here we report an unexpected anti-proliferative role of beta-catenin in non-mitotic differentiated GBM cells. By cell type specific stimulation of miR-302, which directly represses cyclin D1 and stemness features, beta-catenin is capable to change its known proliferative function. Nuclear beta-catenin accumulation in non-mitotic cells is due to a feed forward mechanism between DOCK4 and beta-catenin, allowed by increased GSK3-beta activity. DOCK4 over expression suppresses selfrenewal and tumorigenicity of GBM stem-like cells. Accordingly in the frame of GBM median of survival, increased level of DOCK4 predicts improved patient survival.


Subject(s)
GTPase-Activating Proteins/metabolism , Glioblastoma/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , beta Catenin/metabolism , Adult , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain/pathology , Cell Nucleus/metabolism , Cell Proliferation , Feedback, Physiological , GTPase-Activating Proteins/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Male , Mice , Mice, Inbred NOD , MicroRNAs/genetics , Mitosis , Neoplastic Stem Cells/cytology , Oligodendrocyte Transcription Factor 2/metabolism , Primary Cell Culture , RNA, Small Interfering/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult , beta Catenin/genetics
2.
J Exp Bot ; 66(16): 5043-53, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25911742

ABSTRACT

The homeodomain-leucine zipper (HD-Zip) class of transcription factors is unique to plants. HD-Zip proteins bind to DNA exclusively as dimers recognizing dyad symmetric sequences and act as positive or negative regulators of gene expression. On the basis of sequence homology in the HD-Zip DNA-binding domain, HD-Zip proteins have been grouped into four families (HD-Zip I-IV). Each HD-Zip family can be further divided into subfamilies containing paralogous genes that have arisen through genome duplication. Remarkably, all the members of the HD-Zip IIγ and -δ clades are regulated by light quality changes that induce in the majority of the angiosperms the shade-avoidance response, a process regulated at multiple levels by auxin. Intriguingly, it has recently emerged that, apart from their function in shade avoidance, the HD-Zip IIγ and -δ transcription factors control several auxin-regulated developmental processes, including apical embryo patterning, lateral organ polarity, and gynoecium development, in a white-light environment. This review presents recent advances in our understanding of HD-Zip II protein function in plant development, with particular emphasis on the impact of loss-of-function HD-Zip II mutations on auxin distribution and response. The review also describes evidence demonstrating that HD-Zip IIγ and -δ genes are directly and positively regulated by HD-Zip III transcription factors, primary determinants of apical shoot development, known to control the expression of several auxin biosynthesis, transport, and response genes. Finally, the interplay between HD-Zip II and III transcription factors in embryo apical patterning and organ polarity is discussed.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/genetics , Gene Expression Regulation, Plant , Homeodomain Proteins/genetics , Indoleacetic Acids/metabolism , Leucine Zippers , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Oncogene ; 33(26): 3451-62, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-23912459

ABSTRACT

Glioblastoma multiforme (GBM) are highly invasive and angiogenic malignancies with a median survival time from diagnosis of <15 months. Previous work has revealed robust overexpression of fibronectin (FN) mRNA in GBM, although immunohistochemical staining of FN in these tumors is typically associated with the angiogenic vasculature. Here we sought to examine the expression of tumor cell FN and address its possible involvement in the invasive phenotype of GBM. We found that FN was expressed and assembled into fibrillar arrays in human tumors and in established GBM lines. Cultured cells spontaneously formed dense cellular networks and spheroid-like domes. Depletion of FN by targeted-short hairpin RNA expression disrupted matrix assembly and multicellular network organization by exerting profound effects on cell adhesion and motility. Although FN depletion enhanced persistent directional migration of single cells, it compromised collective invasion of spheroids through a laminin-rich matrix and sensitized cells to ionizing radiation. In orthotopic grafts, FN depletion significantly reduced tumor growth and angiogenesis. Together our results show that FN produced by the tumor cells has a role in GBM pathophysiology and they provide insights into the implications that targeting FN interactions may have for combating this dreaded disease.


Subject(s)
Cell Adhesion/genetics , Fibronectins/metabolism , Glioblastoma/pathology , Animals , Basement Membrane/cytology , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Extracellular Matrix , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/mortality , Humans , Integrin alpha5beta1/metabolism , Mice , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA Interference , RNA, Small Interfering , Spheroids, Cellular , Tumor Cells, Cultured
4.
Cell Death Differ ; 19(2): 232-44, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21720384

ABSTRACT

Glioblastoma multiforme (GBM) is the most common form of primary brain tumor in adults, often characterized by poor survival. Glioma-initiating cells (GiCs) are defined by their extensive self-renewal, differentiation, and tumor initiation properties. GiCs are known to be involved in tumor growth and recurrence, and in resistance to conventional treatments. One strategy to efficiently target GiCs in GBM consists in suppressing their stemness and consequently their tumorigenic properties. In this study, we show that the miR-302-367 cluster is strongly induced during serum-mediated stemness suppression. Stable miR-302-367 cluster expression is sufficient to suppress the stemness signature, self-renewal, and cell infiltration within a host brain tissue, through inhibition of the CXCR4 pathway. Furthermore, inhibition of CXCR4 leads to the disruption of the sonic hedgehog (SHH)-GLI-NANOG network, which is involved in self-renewal and expression of the embryonic stem cell-like signature. In conclusion, we demonstrated that the miR-302-367 cluster is able to efficiently trigger a cascade of inhibitory events leading to the disruption of GiCs stem-like and tumorigenic properties.


Subject(s)
Glioma/genetics , MicroRNAs/genetics , Multigene Family/genetics , Neoplastic Stem Cells/pathology , Receptors, CXCR4/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cell Lineage , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/pathology , Humans , Mice , Neoplastic Stem Cells/metabolism , Receptors, CXCR4/genetics , Serum
5.
Cell Death Differ ; 16(5): 728-37, 2009 May.
Article in English | MEDLINE | ID: mdl-19219066

ABSTRACT

After genotoxic stress, normal cells trigger DNA repair or, if unable to repair, undergo apoptosis to eradicate the cells that bear the risk of becoming tumorigenic. Here we show that repression of the transcription factor, activating transcription factor 3 (ATF3), after ultraviolet (UV)-mediated genotoxic stress impairs the DNA repair process. We provide evidence that ATF3 directly regulates the proliferating cell nuclear antigen (PCNA)-associated factor KIAA0101/p15(PAF). We further show that the expressions of ATF3 and p15(PAF) is sufficient to trigger the DNA repair machinery, and that attenuation of their expression alters DNA repair mechanisms. We show that the expression of p15(PAF) compensates for a lack of ATF3 expression, thereby constituting a major effector of ATF3 in the DNA repair process. In addition, we provide evidence that p15(PAF) expression is required for the correct function of PCNA during DNA repair, as prevention of their interaction significantly alters DNA repair mechanisms. Finally, defective DNA repair, because of the downregulation of p15(PAF) expression, rendered the cells more sensitive to UV-induced cell death. Therefore, our results suggest ATF3 and p15(PAF) as novel gatekeepers of genomic integrity after UV exposure.


Subject(s)
Activating Transcription Factor 3/metabolism , Carrier Proteins/metabolism , DNA Repair , Apoptosis , Cell Line , DNA Damage , DNA-Binding Proteins , Down-Regulation , Genome , HeLa Cells , Humans , Proliferating Cell Nuclear Antigen/metabolism , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Ultraviolet Rays
6.
Cell Death Differ ; 15(9): 1472-80, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18511933

ABSTRACT

In this study, we describe a novel activating transcription factor 3 (ATF3)-dependent death pathway triggered by ultraviolet (UV) irradiation. We demonstrate that ATF3 contributes to UV-induced apoptosis through the regulation of hypoxia inducible factor (Hif)-2alpha expression, which in turn induces the expression of proapoptotic genes, such as Caspase7 or TRAIL (tumor necrosis factor (ligand) superfamily, member 10). Gain of function of Hif-2alpha as well as ATF3 is sufficient to trigger cell death, whereas loss of function of both proteins drastically inhibits UV-induced apoptosis. Repression of Hif-2alpha strongly impairs ATF3-mediated death, providing evidences that Hif-2alpha is the major death effector of ATF3. In addition, Hif-1alpha, already known as a proapoptotic gene, upon UV irradiation, is not able to compensate for the lack of Hif-2alpha expression, thereby confirming the major contribution of Hif-2alpha in UV-mediated cell death. We further demonstrate that this cascade of gene activation depends on p38 and c-Jun N-terminal kinase (JNK) activity. Impairment of such a pathway is likely to contribute to oncogenesis by promoting survival of cells that could accumulate severe chromosomal alterations.


Subject(s)
Activating Transcription Factor 3/physiology , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Ultraviolet Rays , Activating Transcription Factor 3/biosynthesis , Activating Transcription Factor 3/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , RNA, Messenger/metabolism , Signal Transduction
7.
Oncogene ; 26(39): 5772-83, 2007 Aug 23.
Article in English | MEDLINE | ID: mdl-17404577

ABSTRACT

P27kip is a key inhibitory protein of the cell-cycle progression, which is rapidly downregulated in early G1 phase by a post-translational mechanism involving the proteosomal degradation. In this study, using a wounding model that induces cell-cycle entry of human dermal fibroblasts, we demonstrate that p27mRNA is downregulated when cells progress into the G1 phase, and then it returns to its basal level when cells approach the S phase. By using a quantitative polymerase chain reaction screening we identified inhibitors of differentiation (Id3), a bHLH transcriptional repressor, as a candidate mediator accounting for p27 mRNA decrease. Id3 silencing, using an small interfering RNA approach, reversed the injury mediated p27 downregulation demonstrating that Id3 is involved in the transcriptional repression of p27. Reporter gene experiments and a chromatin immunoprecipitation assay showed that Id3 likely exerts its repressive action through ELK1 inhibition. By inhibiting early p27 downregulation, Id3 depletion blocked (i) the G1-phase progression as assessed by the inhibition of pRb phosphorylation and p130 degradation and (ii) the G1/S transition as observed by the inhibition of cyclin A induction, demonstrating that p27 mRNA decrease is required for cell proliferation. Apart from its effect on the early p27 diminution, Id3 appears also involved in the control of the steady-state level of p27 at the G1/S boundary. In conclusion, this study identifies a novel mechanism of p27 regulation which besides p27 protein degradation also implicates a transcriptional mechanism mediated by Id3.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , G1 Phase/physiology , Gene Expression Regulation , Inhibitor of Differentiation Proteins/physiology , Neoplasm Proteins/physiology , RNA, Messenger/metabolism , S Phase/physiology , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Dermis/injuries , Dermis/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunoblotting , Luciferases , Mice , NIH 3T3 Cells , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection , Wound Healing/physiology
8.
Biochem Biophys Res Commun ; 293(1): 112-9, 2002 Apr 26.
Article in English | MEDLINE | ID: mdl-12054571

ABSTRACT

Regeneration of skeletal muscle upon injury is a complex process, involving activation of satellite cells, followed by migration, fusion, and regeneration of damaged myofibers. Previous work concerning the role of the mitogen activated protein (MAP) kinase signaling pathways in muscle injury comes primarily from studies using chemically induced wounding. The purpose of this study was to test the hypothesis that physical injury to skeletal muscle cells in vitro activates the MAP kinase signaling pathways. We demonstrate that extracellular signal regulated kinases (ERKs) 1, 2, and p38 are rapidly and transiently activated in response to injury in C2C12 cells, and are primarily localized to cells adjacent to the wound bed. Culture medium from wounded cells is able to stimulate activation of p38 but not ERK in unwounded cells. These results suggest that both ERK and p38 are involved in the response of muscle cells to physical injury in culture, and reflect what is seen in whole tissues in vivo.


Subject(s)
MAP Kinase Signaling System/physiology , Muscle, Skeletal/injuries , Wounds and Injuries/physiopathology , Animals , Cells, Cultured , Culture Media , Enzyme Activation , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Time Factors , Wounds and Injuries/pathology , p38 Mitogen-Activated Protein Kinases
9.
Exp Cell Res ; 261(1): 220-8, 2000 Nov 25.
Article in English | MEDLINE | ID: mdl-11082292

ABSTRACT

In this study we show that the addition of fresh culture medium to high-density growth-arrested 7TD1 cells induces a strong and transient stimulation of the c-Jun NH2 terminal kinase activity (Jun kinase/JNK), a marked increase in cyclin D2 expression, the phosphorylation of pRb, and the transition from G(1) to S phase. The stimulation of cyclin D2 expression and the induction of JNK activity appear to be the consequences of the alkalinization of the extracellular medium. Indeed both parameters (i) can be induced, regardless of cell dilution, by the addition of a weak base such as triethylamine, and (ii) are together inhibited by (N-ethyl-N-isopropyl)amiloride, a specific inhibitor of the Na(+)/H(+) exchanger. We provide a strong argument indicating the existence of a direct correlation between JNK1 activation and cyclin D2 stimulation. Indeed, we demonstrate that cyclin D2 expression is blocked by SB 202190, an agent known to inhibit both JNK and p38(MAPK), but not by SB 203580, a specific inhibitor of p38(MAPK). Furthermore, we also observed that DMSO and forskolin, two agents that inhibit the proliferation of 7TD1 cells, inhibit in parallel cyclin D2 and JNK1. Altogether our results suggest that (i) JNK1 participates in the signaling pathway which controls the expression of cyclin D2 and (ii) that the inhibition of JNK1 by DMSO and forskolin could explain, at least in part, the antiproliferative action of these drugs in 7TD1 cells.


Subject(s)
Cell Cycle/physiology , Cyclins/metabolism , Hybridomas/cytology , Hybridomas/physiology , Mitogen-Activated Protein Kinases/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Cell Cycle/drug effects , Cyclin D2 , Enzyme Activation , Enzyme Inhibitors/pharmacology , Ethylamines/pharmacology , G1 Phase , Humans , Imidazoles/pharmacology , Interleukin-6/pharmacology , JNK Mitogen-Activated Protein Kinases , Mice , Pyridines/pharmacology , Recombinant Proteins/pharmacology , S Phase , p38 Mitogen-Activated Protein Kinases
10.
Oncogene ; 18(22): 3324-33, 1999 Jun 03.
Article in English | MEDLINE | ID: mdl-10362353

ABSTRACT

p27[KIP1] (p27) is a cyclin dependent kinase inhibitor, involved in the negative regulation of G1 progression in response to a number of anti-proliferative signals. In this study we show, in growing mouse hybridoma (7TD1) and human myeloma (U266) cell lines, that p27 is highly expressed but slightly upregulated when cells are arrested, regardless to the phases of the cell cycle. In contrast, the specific blockade of these cells in early G1 phase reveals the induction of a protein of 23 kDa (p23) specifically recognized by polyclonal anti-p27 antibodies raised against the NH2 terminal part of p27 but not by anti-p21[CIP1] antibodies. Experiments using caspase inhibitors strongly suggest that p23 results from the proteolysis of p27 by a 'caspase-3-like' protease. This cleavage leads to the cytosolic sequestration of p23 but does not alter its binding properties to CDK2 and CDK4 kinases. Indeed, p23 associated in vivo with high molecular weight complexes and coprecipitated with CDK2 and CDK4. We demonstrate by transfection experiments in SaOS-2 cells that p23 induces a G1 phase growth arrest by inhibition of cyclin/CDK2 activity. In summary we describe here a caspase-cleaved form of p27, induced in absence of detectable apoptosis and likely involved in cell cycle regulation.


Subject(s)
CDC2-CDC28 Kinases , Caspases/metabolism , Cell Cycle Proteins , G1 Phase/physiology , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins , Tumor Suppressor Proteins , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Caspase 3 , Caspase Inhibitors , Caspases/genetics , Caspases/immunology , Cell Division/drug effects , Cell Division/physiology , Cell Line , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Cyclins/immunology , Cyclins/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytosol/metabolism , Dimethyl Sulfoxide/pharmacology , G1 Phase/drug effects , Humans , Hybridomas/drug effects , Hybridomas/metabolism , Hybridomas/pathology , Interleukin-6/metabolism , Mice , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/immunology , Molecular Weight , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Oligopeptides/pharmacology , Peptide Fragments/immunology , Protein Serine-Threonine Kinases/metabolism , Time Factors , Tumor Cells, Cultured
11.
Oncogene ; 17(9): 1159-66, 1998 Sep 03.
Article in English | MEDLINE | ID: mdl-9764826

ABSTRACT

Dimethylsulfoxide (DMSO) was shown to inhibit the proliferation of several B cell lines including Raji, Daudi, and SKW6-CL4 but the mechanisms involved in this growth arrest are still unclear. We show that in 7TD1 mouse hybridoma cells a DMSO-induced reversible G1 arrest involves inactivation of Rb kinases, cyclin D2/CDK4 and cyclin E/CDK2. This occurs by at least three distinct mechanisms. Inhibition of cyclin D2 neosynthesis leads to a dramatic decrease of cyclinD2/CDK4 complexes. This in turn enables the redistribution of p27[KIP1] from cyclin D2/CDK4 to cyclin E/CDK2 complexes. In addition, the simultaneous accumulation of p21[CIP1] entails increasing association with cyclin D3/CDK4 and cyclin E/CDK2. Thus, p21[CIP1] and p27[KIP1], act in concert to inhibit cyclin E/CDK2 activity which, together with CDK4 inactivation, confers a G1-phase arrest.


Subject(s)
Anti-Inflammatory Agents/pharmacology , B-Lymphocytes/drug effects , CDC2-CDC28 Kinases , Cell Cycle Proteins , Cyclins/antagonists & inhibitors , Cyclins/drug effects , Dimethyl Sulfoxide/pharmacology , G1 Phase/physiology , Proto-Oncogene Proteins , Tumor Suppressor Proteins , Administration, Topical , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Division/physiology , Cyclin A/antagonists & inhibitors , Cyclin A/metabolism , Cyclin D2 , Cyclin E/antagonists & inhibitors , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Cyclins/metabolism , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Hybridomas/cytology , Hybridomas/drug effects , Hybridomas/physiology , Mice , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/genetics , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/metabolism
13.
Minerva Anestesiol ; 56(12): 1433-7, 1990 Dec.
Article in Italian | MEDLINE | ID: mdl-2100321

ABSTRACT

Eighteen patients aged between 55 and 65 and affected by respiratory insufficient were included in the study. Serum levels of PTH, total calcium, Ca++ and phosphorus were measured. Special attention was focused on PTH concentrations with in some case provedo to be above normal. Two hypotheses of pathogenesis are put forward. The first suggests that reduced Ca++ levels are responsible for anomalous PTH serum concentrations, whereas the second suggests that this role may be played by hypoxia. The latter would indirectly or directly affect parathyroids, either increasing PTH hyperincretion or slowing down the metabolism of the hormone by kidneys and liver which are in turn negatively affected by low blood oxygen tension.


Subject(s)
Hyperparathyroidism/etiology , Respiratory Insufficiency/complications , Aged , Female , Humans , Hyperparathyroidism/blood , Male , Middle Aged , Parathyroid Hormone/blood , Respiratory Insufficiency/blood
14.
Leuk Res ; 10(10): 1249-54, 1986.
Article in English | MEDLINE | ID: mdl-3464813

ABSTRACT

The expression of two G1-specific clones, p2A9 and p4F1, of two cell cycle-related oncogenes, c-myc and c-myb and of one S phase-specific gene, the H3 histone gene, was explored in 11 cases of acute myeloid leukemia. Both Northern blot analysis and in-situ hybridization were employed. Differential patterns of gene expression were observed. In 6 out of 11 cases a considerable or high expression of the p2A9 and p4F1 clones and of c-myc and c-myb oncogenes was observed. In 2 cases a high expression of c-myc was matched by low or absent expression of the other genes examined. In 3 cases the expression of 2A9, 4F1, c-myc and c-myb was very low or undetectable. In two of these cases a considerable expression of the H3 histone gene was observed.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Oncogenes , Histones/genetics , Humans , Interphase , Nucleic Acid Hybridization
SELECTION OF CITATIONS
SEARCH DETAIL