Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Brain Dev ; 44(8): 562-566, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35469635

ABSTRACT

BACKGROUND: Chromosome 18q duplications are associated with a range of phenotypes often similar to complete trisomy 18, variably including poor growth, feeding difficulties, congenital malformations and dysmorphic facial features. Although 18q duplication patients may have seizures and developmental impairment, brain MRI typically shows only variable degrees of cerebral atrophy. PATIENT: We present a boy with a 52.2 Mb 18q duplication in whom brain MRI in the neonatal period showed striking white matter abnormalities, most notable in the frontal lobes. His clinical presentation was otherwise in keeping with trisomy 18, including characteristic facial features, hypotonia, cardiac malformation, rocker bottom feet, pectus excavatum, short and broad thumbs and halluces, and diabetes insipidus. CONCLUSION: Since not previously reported in association with 18q duplication, the observation of cerebral white matter anomalies is particularly interesting. This radiologic pattern is a well-recognized feature of 18q deletion syndrome, hypothesized by many to occur due to haploinsufficiency of MBP, the gene encoding myelin basic protein. The mechanisms leading to the white matter anomalies in this patient remain unexplained.


Subject(s)
White Matter , Chromosome Deletion , Chromosome Duplication/genetics , Chromosomes , Chromosomes, Human, Pair 18/genetics , Humans , Trisomy/genetics , Trisomy 18 Syndrome , White Matter/diagnostic imaging
2.
Oncogene ; 41(3): 309-320, 2022 01.
Article in English | MEDLINE | ID: mdl-34743206

ABSTRACT

While aneuploidy is a main enabling characteristic of cancers, it also creates specific vulnerabilities. Here we demonstrate that Ran inhibition targets epithelial ovarian cancer (EOC) survival through its characteristic aneuploidy. We show that induction of aneuploidy in rare diploid EOC cell lines or normal cells renders them highly dependent on Ran. We also establish an inverse correlation between Ran and the tumor suppressor NR1D1 and reveal the critical role of Ran/NR1D1 axis in aneuploidy-associated endogenous DNA damage repair. Mechanistically, we show that Ran, through the maturation of miR4472, destabilizes the mRNA of NR1D1 impacting several DNA repair pathways. We showed that NR1D1 interacts with both PARP1 and BRCA1 leading to the inhibition of DNA repair. Concordantly, loss of Ran was associated with NR1D1 induction, accumulation of DNA damages, and lethality of aneuploid EOC cells. Our findings suggest a synthetic lethal strategy targeting aneuploid cells based on their dependency to Ran.


Subject(s)
GTP Phosphohydrolases/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Ovarian Neoplasms/genetics , Aneuploidy , Animals , Female , Humans , Mice
3.
Cell Cycle ; 19(2): 163-178, 2020 01.
Article in English | MEDLINE | ID: mdl-31885322

ABSTRACT

The cell cycle is tightly regulated by cyclins and their catalytic moieties, the cyclin-dependent kinases (CDKs). Cyclin D1, in association with CDK4/6, acts as a mitogenic sensor and integrates extracellular mitogenic signals and cell cycle progression. When deregulated (overexpressed, accumulated, inappropriately located), cyclin D1 becomes an oncogene and is recognized as a driver of solid tumors and hemopathies. Recent studies on the oncogenic roles of cyclin D1 reported non-canonical functions dependent on the partners of cyclin D1 and its location within tumor cells or tissues. Support for these new functions was provided by various mouse models of oncogenesis. Finally, proteomic and transcriptomic data identified complex cyclin D1 networks. This review focuses on these aspects of cyclin D1 pathophysiology, which may be crucial for targeted therapy.Abbreviations: aa, amino acid; AR, androgen receptor; ATM, ataxia telangectasia mutant; ATR, ATM and Rad3-related; CDK, cyclin-dependent kinase; ChREBP, carbohydrate response element binding protein; CIP, CDK-interacting protein; CHK1/2, checkpoint kinase 1/2; CKI, CDK inhibitor; DDR, DNA damage response; DMP1, cyclin D-binding myb-like protein; DSB, double-strand DNA break; DNA-PK, DNA-dependent protein kinase; ER, estrogen receptor; FASN, fatty acid synthase; GSK3ß, glycogen synthase-3ß; HAT, histone acetyltransferase; HDAC, histone deacetylase; HK2, hexokinase 2; HNF4α, and hepatocyte nuclear factor 4α; HR, homologous recombination; IR, ionizing radiation; KIP, kinase inhibitory protein; MCL, mantle cell lymphoma; NHEJ, non-homologous end-joining; PCAF, p300/CREB binding-associated protein; PGC1α, PPARγ co-activator 1α; PEST, proline-glutamic acid-serine-threonine, PK, pyruvate kinase; PPAR, peroxisome proliferator-activated receptor; RB1, retinoblastoma protein; ROS, reactive oxygen species; SRC, steroid receptor coactivator; STAT, signal transducer and activator of transcription; TGFß, transforming growth factor ß; UPS, ubiquitin-proteasome system; USP22, ubiquitin-specific peptidase 22; XPO1 (or CRM1) exportin 1.


Subject(s)
Cyclin D1/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Nucleus/metabolism , Cyclin D1/chemistry , DNA Damage , Humans , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/pathology
4.
Sci Rep ; 7(1): 13946, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29066743

ABSTRACT

Mantle cell lymphoma (MCL) is a hematologic neoplasm characterised by the t(11;14)(q13;q32) translocation leading to aberrant cyclin D1 expression. The cell functions of cyclin D1 depend on its partners and/or subcellular distribution, resulting in different oncogenic properties. We observed the accumulation of cyclin D1 in the cytoplasm of a subset of MCL cell lines and primary cells. In primary cells, this cytoplasmic distribution was correlated with a more frequent blastoid phenotype. We performed immunoprecipitation assays and mass spectrometry on enriched cytosolic fractions from two cell lines. The cyclin D1 interactome was found to include several factors involved in adhesion, migration and invasion. We found that the accumulation of cyclin D1 in the cytoplasm was associated with higher levels of migration and invasiveness. We also showed that MCL cells with high cytoplasmic levels of cyclin D1 engrafted more rapidly into the bone marrow, spleen, and brain in immunodeficient mice. Both migration and invasion processes, both in vivo and in vitro, were counteracted by the exportin 1 inhibitor KPT-330, which retains cyclin D1 in the nucleus. Our data reveal a role of cytoplasmic cyclin D1 in the control of MCL cell migration and invasion, and as a true operator of MCL pathogenesis.


Subject(s)
Cell Movement , Cyclin D1/metabolism , Cytoplasm/metabolism , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Active Transport, Cell Nucleus , Adult , Aged , Aged, 80 and over , Animals , Cell Nucleus/metabolism , Cell Transformation, Neoplastic , Cytosol/metabolism , Female , Humans , Male , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Proteomics
5.
Oncotarget ; 7(29): 45214-45224, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27286258

ABSTRACT

The interactions of multiple myeloma (MM) cells with their microenvironment are crucial for pathogenesis. MM cells could interact differentially with their microenvironment depending on the type of cyclin D they express. We established several clones that constitutively express cyclin D1 from the parental RPMI8226 MM cell line and analyzed the impact of cyclin D1 expression on cell behavior. We performed a gene expression profiling study on cyclin D1-expressing vs. control cells and validated the results by semi-quantitative RT-PCR. The expression of cyclin D1 altered the transcription of genes that control adhesion and migration. We confirmed that cyclin D1 increases cell adhesion to stromal cells and fibronectin, stabilizes F-actin fibers, and enhances chemotaxis and inflammatory chemokine secretion. Both control and cyclin D1-expressing cells were more resistant to acute carfilzomib treatment when cultured on stromal cells than in suspension. However, this resistance was specifically reduced in cyclin D1-expressing cells after pomalidomide pre-treatment that modifies tumor cell/microenvironment interactions. Transcriptomic analysis revealed that cyclin D1 expression was also associated with changes in the expression of genes controlling metabolism. We also found that cyclin D1 expression disrupted the redox balance by producing reactive oxygen species. The resulting oxidative stress activated the p44/42 mitogen-activated protein kinase (or ERK1/2) signaling pathway, increased cell adhesion to fibronectin or stromal cells, and controlled drug sensitivity.Our results have uncovered a new function for cyclin D1 in the control of redox metabolism and interactions of cyclin D1-expressing MM cells with their bone marrow microenvironment.


Subject(s)
Cyclin D1/physiology , Multiple Myeloma/pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Chemokines/metabolism , Cyclin D1/genetics , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Tumor Microenvironment
6.
Apoptosis ; 18(9): 1048-59, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23673989

ABSTRACT

Programmed cell death is an important factor in tissue homeostasis. Lot of work has been performed to characterize the caspase-dependent cell death. Caspase-independent cell death, although important in many physiological situations, is less investigated. In this work we show that two caspase-independent effectors of cell death, namely apoptosis-inducing factor and leukocyte elastase inhibitor derived DNase II interact and can cooperate to induce cell death. These results contribute to the knowledge of molecular pathways of cell death, an important issue in the development of new therapeutic strategies for the treatment of cancer or neurodegenerative diseases.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis , Endodeoxyribonucleases/metabolism , Neurodegenerative Diseases/metabolism , Serpins/metabolism , Animals , Apoptosis Inducing Factor/genetics , Caspases/metabolism , Cell Line , Endodeoxyribonucleases/genetics , Humans , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/genetics , Protein Binding , Serpins/genetics
7.
BMC Cancer ; 12: 234, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22691188

ABSTRACT

BACKGROUND: The finite replicative lifespan of cells, termed cellular senescence, has been proposed as a protective mechanism against the proliferation of oncogenically damaged cells, that fuel cancer. This concept is further supported by the induction of premature senescence, a process which is activated when an oncogene is expressed in normal primary cells as well as following intense genotoxic stresses. Thus, deregulation of genes that control this process, like the tumor suppressor p53, may contribute to promoting cancer by allowing cells to bypass senescence. A better understanding of the genes that contribute to the establishment of senescence is therefore warranted. Necdin interacts with p53 and is also a p53 target gene, although the importance of Necdin in the p53 response is not clearly understood. METHODS: In this study, we first investigated Necdin protein expression during replicative senescence and premature senescence induced by gamma irradiation and by the overexpression of oncogenic RasV12. Gain and loss of function experiments were used to evaluate the contribution of Necdin during the senescence process. RESULTS: Necdin expression declined during replicative aging of IMR90 primary human fibroblasts or following induction of premature senescence. Decrease in Necdin expression seemed to be a consequence of the establishment of senescence since the depletion of Necdin in human cells did not induce a senescence-like growth arrest nor a flat morphology or SA-ß-galactosidase activity normally associated with senescence. Similarly, overexpression of Necdin did not affect the life span of IMR90 cells. However, we demonstrate that in normal human cells, Necdin expression mimicked the effect of p53 inactivation by increasing radioresistance. CONCLUSION: This result suggests that Necdin potentially attenuate p53 signaling in response to genotoxic stress in human cells and supports similar results describing an inhibitory function of Necdin over p53-dependent growth arrest in mice.


Subject(s)
DNA Damage/radiation effects , Fibroblasts/radiation effects , Nerve Tissue Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Radiation Tolerance/physiology , Cell Line , Cell Proliferation/radiation effects , Cellular Senescence/physiology , Cellular Senescence/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Gamma Rays , Humans , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/metabolism
8.
Cancer Res ; 71(5): 1690-9, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21343394

ABSTRACT

Cyclin D1 is a cell cycle regulatory protein that acts at the G1-S transition, following its binding to and activation by the cyclin-dependent kinases 4 or 6. Cyclin D1 is absent from the entire B-cell lineage but is present in a large percentage of 2 types of malignant B-cell hemopathy--mantle cell lymphoma and multiple myeloma--suggesting a major role of this protein in the malignancy process. We show here, in an experimental model of cyclin D1 fusion protein transduction in mature B cells, that, cyclin D1 inhibits total mitochondrial activity. Cyclin D1 is localized at the outer mitochondrial membrane, bound to a voltage-dependent anion channel through its central domain, and it competes with hexokinase 2 for binding to this channel. The bound cyclin D1 decreases the supply of ADP, ATP, and metabolites, thereby reducing energy production. This function of cyclin D1 was also reported by others in normal and transformed mammary epithelial cells, suggesting that it may be ubiquitous.


Subject(s)
B-Lymphocytes/metabolism , Cyclin D1/metabolism , Mitochondria/metabolism , Blotting, Western , Cell Line , Cell Proliferation , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Membrane Potential, Mitochondrial/physiology , Microscopy, Confocal , Recombinant Fusion Proteins/metabolism
9.
Cell Signal ; 23(1): 171-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20837141

ABSTRACT

p27(Kip1) is a cyclin-dependent kinase inhibitor that plays a critical role in regulating G(1)/S transition, and whose activity is, in part, regulated through interactions with D-type cyclins. We have generated the BD1-9 cell line, a BaF3 pro-B cells derivative in which cyclin D1 can be induced rapidly and reversibly by ponasterone A. The induction of cyclin D1 expression leads to a targeted p27(Kip1) accumulation in both cytoplasmic and nuclear compartments. But, only the p27(Kip1) form phosphorylated on serine 10 (pSer10-p27(Kip1)) accumulates in BD1-9 cells. We found that the binding of cyclin D1 and pSer10-p27(Kip1) prevents p27(Kip1) degradation by the cytoplasmic Kip1 ubiquitylation-promoting complex (KPC) proteosomic pathway. Importantly, the nuclear CDK2 activity which is crucial for G(1)/S transition is not altered by p27(Kip1) increase. Using siRNA techniques, we revealed that p27(Kip1) inhibition does not affect the distribution of BD1-9 cells in the different phases of the cell cycle. Our study demonstrates that aberrant cyclin D1 expression acts as a p27(Kip1) trap in B lymphocytes but does not induce p27(Kip1) relocation from the nucleus to the cytoplasm and does not modulate the G(1)/S transition. Since our cellular model mimics what observed in aggressive lymphomas, our data bring new insights into the understanding of their physiopathology.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Precursor Cells, B-Lymphoid/metabolism , Animals , Cell Line , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Ecdysterone/analogs & derivatives , Ecdysterone/pharmacology , G1 Phase , Mice , Phosphorylation , Precursor Cells, B-Lymphoid/immunology , Protein Stability , RNA Interference , RNA, Small Interfering/metabolism , S Phase
10.
Mol Cancer ; 9: 103, 2010 May 10.
Article in English | MEDLINE | ID: mdl-20459741

ABSTRACT

BACKGROUND: Aberrant expression of cyclin D1 is a common feature in multiple myeloma (MM) and always associated with mantle cell lymphoma (MCL). CCND1 gene is alternatively spliced to produce two cyclin D1 mRNA isoforms which are translated in two proteins: cyclin D1a and cyclin D1b. Both isoforms are present in MM cell lines and primary cells but their relative role in the tumorigenic process is still elusive. RESULTS: To test the tumorigenic potential of cyclin D1b in vivo, we generated cell clones derived from the non-CCND1 expressing MM LP-1 cell line, synthesizing either cyclin D1b or cyclin K, a structural homolog and viral oncogenic form of cyclin D1a. Immunocompromised mice injected s.c. with LP-1K or LP-1D1b cells develop tumors at the site of injection. Genome-wide analysis of LP-1-derived cells indicated that several cellular processes were altered by cyclin D1b and/or cyclin K expression such as cell metabolism, signal transduction, regulation of transcription and translation. Importantly, cyclin K and cyclin D1b have no major action on cell cycle or apoptosis regulatory genes. Moreover, they impact differently cell functions. Cyclin K-expressing cells have lost their migration properties and display enhanced clonogenic capacities. Cyclin D1b promotes tumorigenesis through the stimulation of angiogenesis. CONCLUSIONS: Our study indicates that cyclin D1b participates into MM pathogenesis via previously unrevealed actions.


Subject(s)
Cyclin D1/metabolism , Cyclins/metabolism , Multiple Myeloma/metabolism , Animals , Cell Cycle/physiology , Cell Line , Cell Movement/physiology , Cell Separation , Chick Embryo , Cyclin D1/genetics , Cyclins/genetics , Female , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Nude , Multiple Myeloma/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...