Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Med Virol ; 95(3): e28633, 2023 03.
Article in English | MEDLINE | ID: mdl-36866703

ABSTRACT

Burkitt lymphoma (BL) is a B cell malignancy associated with the Epstein-Barr virus (EBV). Most BL cases are characterized by a t(8;14) chromosomal translocation involving the MYC oncogene and the immunoglobulin heavy chain gene (IGH). The role of EBV in promoting this translocation remains largely unknown. Here we provide the experimental evidence that EBV reactivation from latency leads to an increase in the proximity between the MYC and IGH loci, otherwise located far away in the nuclear space both in B-lymphoblastoid cell lines and in patients' B-cells. Specific DNA damage within the MYC locus, followed by the MRE11-dependent DNA repair plays a role in this process. Using a CRISPR/Cas9-based B cell model to induce specific DNA double strand breaks in MYC and IGH loci, we have shown that the MYC-IGH proximity induced by EBV reactivation leads to an increased t(8;14) translocation frequency.


Subject(s)
Burkitt Lymphoma , Epstein-Barr Virus Infections , Humans , Herpesvirus 4, Human/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Genes, Immunoglobulin Heavy Chain
2.
Gene Ther ; 30(1-2): 167-171, 2023 02.
Article in English | MEDLINE | ID: mdl-32999452

ABSTRACT

B-cell lines and primary PBMCs are notoriously hard to transfect, thus making genome editing, ectopic gene expression, or gene silencing experiments particularly tedious. Here we propose a novel efficient and reproducible protocol for electrotransfection of lymphoblastoid, B-cell lymphoma, leukemia cell lines, and B cells from PBMCs. The proposed protocol requires neither costly equipment nor expensive reagents; it can be used with small or large plasmids. Transfection and viability rates of about 79% and 58%, respectively, have been routinely achieved by optimizing the salt concentration in the electrotransfection medium and the amount of plasmid used. A validation of the protocol was obtained via the generation of a TP53-/- RPMI8866 lymphoblastoid cell line which should prove useful in future hematological and blood cancer studies.


Subject(s)
Ectopic Gene Expression , Gene Editing , Humans , Gene Editing/methods , Transfection , Cell Line , Plasmids
3.
Trends Cancer ; 5(5): 268-271, 2019 05.
Article in English | MEDLINE | ID: mdl-31174839

ABSTRACT

DUX4, a double homeobox transcription factor, has been mostly studied in facioscapulohumeral dystrophy (FSHD), a pathology linked to a deletion of subtelomeric repeats on chromosome 4q. More recently, however, the gene has been associated with various sarcomas and haematological malignancies. Drugs developed for FSHD could be tested on cancer cells to develop efficient treatment strategies for both pathologies.


Subject(s)
Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Neoplasms/genetics , Disease Susceptibility , Epigenesis, Genetic , Gene Rearrangement , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Humans , Molecular Targeted Therapy , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Protein Binding , Protein Interaction Domains and Motifs
4.
J Cell Physiol ; 234(9): 15678-15685, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30701532

ABSTRACT

Individuals infected with human immunodeficiency virus (HIV) are at increased risk for Burkitt lymphoma, a B-cell malignancy which occurs after a chromosomal translocation rearranging the MYC oncogene with an immunoglobulin gene locus, usually the IGH heavy chain gene locus. We have previously reported that the HIV protein Tat which circulates in all HIV-positive individuals whatever their immune status caused an increased rate of colocalization between IGH and MYC in B-cells nuclei. We here present in vitro evidence that Tat activates the expression of the AICDA gene that encodes the activation-induced cytidine deaminase whose physiological function is to create double-strand breaks for immunoglobulin gene maturation. In the presence of Tat, DNA damage was observed concomitantly in both MYC and IGH, followed by DNA repair by nonhomologous end joining. AICDA was further found overexpressed in vivo in peripheral blood B-cells from HIV-infected individuals. Thus, the capacity of Tat to spontaneously penetrate B-cells could be sufficient to favor the occurrence of MYC-IGH oncogenic rearrangements during erroneous repair, a plausible cause for the increased incidence of Burkitt lymphoma in the HIV-infected population.

5.
Nucleic Acids Res ; 46(22): e135, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30215776

ABSTRACT

Analysis of large-scale interphase genome positioning with reference to a nuclear landmark has recently been studied using sequencing-based single cell approaches. However, these approaches are dependent upon technically challenging, time consuming and costly high throughput sequencing technologies, requiring specialized bioinformatics tools and expertise. Here, we propose a novel, affordable and robust microscopy-based single cell approach, termed Topokaryotyping, to analyze and reconstruct the interphase positioning of genomic loci relative to a given nuclear landmark, detectable as banding pattern on mitotic chromosomes. This is accomplished by proximity-dependent histone labeling, where biotin ligase BirA fused to nuclear envelope marker Emerin was coexpressed together with Biotin Acceptor Peptide (BAP)-histone fusion followed by (i) biotin labeling, (ii) generation of mitotic spreads, (iii) detection of the biotin label on mitotic chromosomes and (iv) their identification by karyotyping. Using Topokaryotyping, we identified both cooperativity and stochasticity in the positioning of emerin-associated chromatin domains in individual cells. Furthermore, the chromosome-banding pattern showed dynamic changes in emerin-associated domains upon physical and radiological stress. In summary, Topokaryotyping is a sensitive and reliable technique to quantitatively analyze spatial positioning of genomic regions interacting with a given nuclear landmark at the single cell level in various experimental conditions.


Subject(s)
Karyotyping/methods , Mitosis , Nuclear Envelope/metabolism , Single-Cell Analysis/methods , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , In Situ Hybridization, Fluorescence , Interphase , Membrane Proteins/metabolism , Microscopy, Confocal , Nuclear Envelope/genetics , Nuclear Proteins/metabolism , Reproducibility of Results
6.
Oncogene ; 37(18): 2410-2421, 2018 05.
Article in English | MEDLINE | ID: mdl-29440708

ABSTRACT

The globotriaosylceramide Gb3 is a glycosphingolipid expressed on a subpopulation of germinal center B lymphocytes which has been recognized as the B cell differentiation antigen CD77. Among tumoral cell types, Gb3/CD77 is strongly expressed in Burkitt's lymphoma (BL) cells as well as other solid tumors including breast, testicular and ovarian carcinomas. One known ligand of Gb3/CD77 is Verotoxin-1 (VT-1), a Shiga toxin produced in specific E. coli strains. Previously, we have reported that in BL cells, VT-1 induces apoptosis via a caspase-dependent and mitochondria-dependent pathway. Yet, the respective roles of various apoptogenic factors remained to be deciphered. Here, this apoptotic pathway was found to require cleavage of the BID protein by caspase-8 as well as activation of two other apoptogenic proteins, BAK and BAX. Surprisingly however, t-BID, the truncated form of BID resulting from caspase-8 cleavage, played no role in the conformational changes of BAK and BAX. Rather, their activation occurred under the control of full length BID (FL-BID). Indeed, introducing a non-cleavable form of BID (BID-D59A) into BID-deficient BL cells restored BAK and BAX activation following VT-1 treatment. Still, t-BID was involved along with FL-BID in the BAK-dependent and BAX-dependent cytosolic release of CYT C and SMAC/DIABLO from the mitochondrial intermembrane space: FL-BID was found to control the homo-oligomerization of both BAK and BAX, likely contributing to the initial release of CYT C and SMAC/DIABLO, while t-BID was needed for their hetero-oligomerization and ensuing release amplification. Together, our results reveal a functional cooperation between BAK and BAX during VT-1-induced apoptosis and, unexpectedly, that activation of caspase-8 and production of t-BID were not mandatory for initiation of the cell death process.


Subject(s)
Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/physiology , Burkitt Lymphoma/pathology , Shiga Toxins/pharmacology , Apoptosis/genetics , BH3 Interacting Domain Death Agonist Protein/chemistry , BH3 Interacting Domain Death Agonist Protein/genetics , Burkitt Lymphoma/genetics , Caspase 8/metabolism , HEK293 Cells , Humans , Protein Domains/genetics , Protein Domains/physiology , Protein Isoforms/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology
7.
Oncotarget ; 8(25): 40079-40089, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28445143

ABSTRACT

The immunoglobulin heavy chain (IGH) gene loci are subject to specific recombination events during B-cell differentiation including somatic hypermutation and class switch recombination which mark the end of immunoglobulin gene maturation in germinal centers of secondary lymph nodes. These two events rely on the activity of activation-induced cytidine deaminase (AID) which requires DNA double strand breaks be created, a potential danger to the cell. Applying 3D-fluorescence in situ hybridization coupled with immunofluorescence staining to a previously described experimental system recapitulating normal B-cell differentiation ex vivo, we have kinetically analyzed the radial positioning of the two IGH gene loci as well as their proximity with the nucleolus, heterochromatin and γH2AX foci. Our observations are consistent with the proposal that these IGH gene rearrangements take place in a specific perinucleolar "recombination compartment" where AID could be sequestered thus limiting the extent of its potentially deleterious off-target effects.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Nucleolus/immunology , Immunoglobulin Heavy Chains/immunology , B-Lymphocytes/metabolism , Cell Line, Tumor , Cell Nucleolus/metabolism , Cells, Cultured , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Humans , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , In Situ Hybridization, Fluorescence/methods , Lymphocyte Activation/immunology , Microscopy, Confocal , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology
8.
Oncotarget ; 7(40): 65090-65108, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27556182

ABSTRACT

We performed transcriptome profiling of human immortalized myoblasts (MB) transiently expressing double homeobox transcription factor 4 (DUX4) and double homeobox transcription factor 4 centromeric (DUX4c) and identified 114 and 70 genes differentially expressed in DUX4- and DUX4c-transfected myoblasts, respectively. A significant number of differentially expressed genes were involved in inflammation, cellular migration and chemotaxis suggesting a role for DUX4 and DUX4c in these processes. DUX4 but not DUX4c overexpression resulted in upregulation of the CXCR4 (C-X-C motif Receptor 4) and CXCL12 (C-X-C motif ligand 12 also known as SDF1) expression in human immortalized myoblasts. In a Transwell cell migration assay, human bone marrow-derived mesenchymal stem cells (BMSCs) were migrating more efficiently towards human immortalized myoblasts overexpressing DUX4 as compared to controls; the migration efficiency of DUX4-transfected BMSCs was also increased. DUX4c overexpression in myoblasts or in BMSCs had no impact on the rate of BMSC migration. Antibodies against SDF1 and CXCR4 blocked the positive effect of DUX4 overexpression on BMSC migration. We propose that DUX4 controls the cellular migration of mesenchymal stem cells through the CXCR4 receptor.


Subject(s)
Cell Movement/physiology , Chemokine CXCL12/metabolism , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Receptors, CXCR4/metabolism , Cells, Cultured , Humans , Myoblasts/metabolism , Transcriptome
9.
Free Radic Biol Med ; 99: 244-258, 2016 10.
Article in English | MEDLINE | ID: mdl-27519269

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is one of the three most common muscular dystrophies in the Western world, however, its etiology remains only partially understood. Here, we provide evidence of constitutive DNA damage in in vitro cultured myoblasts isolated from FSHD patients and demonstrate oxidative DNA damage implication in the differentiation of these cells into phenotypically-aberrant myotubes. Double homeobox 4 (DUX4), the major actor in FSHD pathology induced DNA damage accumulation when overexpressed in normal human myoblasts, and RNAi-mediated DUX4 inhibition reduced the level of DNA damage in FSHD myoblasts. Addition of tempol, a powerful antioxidant, to the culture medium of proliferating DUX4-transfected myoblasts and FSHD myoblasts reduced the level of DNA damage, suggesting that DNA alterations are mainly due to oxidative stress. Antioxidant treatment during the myogenic differentiation of FSHD myoblasts significantly reduced morphological defects in myotube formation. We propose that the induction of DNA damage is a novel function of the DUX4 protein affecting myogenic differentiation of FSHD myoblasts.


Subject(s)
Homeodomain Proteins/genetics , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Myoblasts/metabolism , Oxidative Stress , Antioxidants/pharmacology , Case-Control Studies , Cell Differentiation , Cyclic N-Oxides/pharmacology , DNA Damage , Gene Expression Profiling , Gene Expression Regulation , Gene Ontology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/metabolism , Humans , Molecular Sequence Annotation , Multigene Family , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts/pathology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spin Labels , Transfection
10.
Histochem Cell Biol ; 145(4): 475-83, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26860865

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is a progressive muscular dystrophy linked to a deletion of a subset of D4Z4 macrosatellite repeats accompanied by a chromatin relaxation of the D4Z4 array on chromosome 4q. In vitro, FSHD primary myoblasts show altered expression of oxidative-related genes and are more susceptible to oxidative stress. Double homeobox 4 (DUX4) gene, encoded within each D4Z4 unit, is normally transcriptionally silenced but is found aberrantly expressed in skeletal muscles of FSHD patients. Its expression leads to a deregulation of DUX4 target genes including those implicated in redox balance. Here, we assessed DNA repair efficiency of oxidative DNA damage in FSHD myoblasts and DUX4-transfected myoblasts. We have shown that the DNA repair activity is altered neither in FSHD myoblasts nor in immortalized human myoblasts transiently expressing DUX4. DNA damage caused by moderate doses of an oxidant is efficiently repaired while FSHD myoblasts exposed for 24 h to high levels of oxidative stress accumulated more DNA damage than normal myoblasts, suggesting that FSHD myoblasts remain more vulnerable to oxidative stress at high doses of oxidants.


Subject(s)
DNA Damage , DNA Repair , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts, Skeletal/metabolism , Oxidative Stress , Cells, Cultured , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hydrogen Peroxide/pharmacology , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/pathology , Oxidative Stress/drug effects
11.
J Cell Biochem ; 117(7): 1506-10, 2016 07.
Article in English | MEDLINE | ID: mdl-26873538

ABSTRACT

The immunoglobulin heavy chain (IGH) locus is submitted to intra-chromosomal DNA breakages and rearrangements during normal B cell differentiation that create a risk for illegitimate inter-chromosomal translocations leading to a variety of B-cell malignancies. In most Burkitt's and Mantle Cell lymphomas, specific chromosomal translocations juxtapose the IGH locus with a CMYC or Cyclin D1 (CCND1) gene, respectively. 3D-fluorescence in situ hybridization was performed on normal peripheral B lymphocytes induced to mature in vitro from a naive state to the stage where they undergo somatic hypermutation (SHM) and class switch recombination (CSR). The CCND1 genes were found very close to the IGH locus in naive B cells and further away after maturation. In contrast, the CMYC alleles became localized closer to an IGH locus at the stage of SHM/CSR. The colocalization observed between the two oncogenes and the IGH locus at successive stages of B-cell differentiation occurred in the immediate vicinity of the nucleolus, consistent with the known localization of the RAGs and AID enzymes whose function has been demonstrated in IGH physiological rearrangements. We propose that the chromosomal events leading to Mantle Cell lymphoma and Burkitt's lymphoma are favored by the colocalization of CCND1 and CMYC with IGH at the time the concerned B cells undergo VDJ recombination or SHM/CSR, respectively. J. Cell. Biochem. 117: 1506-1510, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
B-Lymphocytes/metabolism , Cell Differentiation/physiology , Cyclin D1/metabolism , Gene Rearrangement, B-Lymphocyte, Heavy Chain/physiology , Immunoglobulin Heavy Chains/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Alleles , B-Lymphocytes/cytology , Cyclin D1/genetics , Genetic Loci/physiology , Humans , Immunoglobulin Heavy Chains/genetics , In Situ Hybridization, Fluorescence , Proto-Oncogene Proteins c-myc/genetics
12.
J Cell Physiol ; 231(1): 62-71, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26218298

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is a neuromuscular disease with a prevalence that could reach 1 in 8,000 characterized by progressive asymmetric muscle weakness. Myoblasts isolated from FSHD muscles exhibit morphological differentiation defects and show a distinct transcription profile. These abnormalities may be linked to the muscle weakness in FSHD patients. We have tested whether fusion of FSHD myoblasts with primary myoblasts isolated from healthy individuals could correct the differentiation defects. Our results show that the number of hybrid myotubes with normal phenotype increased with the percentage of normal myoblasts initially cultured. We demonstrated that a minimum of 50% of normal nuclei is required for a phenotypic correction of the FSHD phenotype. Moreover, transcriptomic profiles of phenotypically corrected hybrid myotubes showed that the expression of deregulated genes in FSHD myotubes became almost normal. The number of deregulated pathways also decreased from 39 in FSHD myotubes to one in hybrid myotubes formed with 40% FSHD and 60% normal myoblasts. We thus propose that while phenotypical and functional correction of FSHD is feasible, it requires more than 50% of normal myoblasts, it creates limitations for cell therapy in the FSHD context.


Subject(s)
Cell Differentiation/physiology , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts/cytology , Adult , Cell Differentiation/genetics , Cells, Cultured , Female , Humans , Male , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Phenotype , Young Adult
13.
Autophagy ; 11(12): 2275-87, 2015.
Article in English | MEDLINE | ID: mdl-26565591

ABSTRACT

The Epstein-Barr virus (EBV) is associated with various lymphoproliferative disorders and lymphomas. We have previously demonstrated that treating wild-type TP53-expressing B cell lines with the TP53 pathway activator nutlin-3 induced apoptosis in EBV-negative and EBV-positive latency I cells whereas EBV-positive latency III cells remained much more apoptosis-resistant. Here, we report a constitutively high level of autophagy in these resistant cells which express high levels of the proautophagic protein BECN1/Beclin 1 based, at least in part, on the activation of the NFKB signaling pathway by the viral protein LMP1. Following treatment with nutlin-3, several autophagy-stimulating genes were upregulated both in EBV-negative and EBV-positive latency III cells. However the process of autophagy was only triggered in the latter and was associated with an upregulation of SESN1/sestrin 1 and inhibition of MTOR more rapid than in EBV-negative cells. A treatment with chloroquine, an inhibitor of autophagy, potentiated the apoptotic effect of nutlin-3, particularly in those EBV-positive cells which were resistant to apoptosis induced by nutlin-3 alone, thereby showing that autophagy participates in this resistant phenotype. Finally, using immunohistochemical staining, clinical samples from various B cell lymphoproliferations with the EBV-positive latency II or III phenotype were found to harbor a constitutively active autophagy.


Subject(s)
Apoptosis/drug effects , Autophagy , B-Lymphocytes/cytology , B-Lymphocytes/virology , Herpesvirus 4, Human , Lymphoma/pathology , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression/drug effects , Humans , Imidazoles/pharmacology , Lymphoma/virology , Piperazines/pharmacology , Tumor Suppressor Protein p53/metabolism
14.
Aging Cell ; 14(3): 322-33, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25720734

ABSTRACT

The H3.3 histone variant has been a subject of increasing interest in the field of chromatin studies due to its two distinguishing features. First, its incorporation into chromatin is replication independent unlike the replication-coupled deposition of its canonical counterparts H3.1/2. Second, H3.3 has been consistently associated with an active state of chromatin. In accordance, this histone variant should be expected to be causally involved in the regulation of gene expression, or more generally, its incorporation should have downstream consequences for the structure and function of chromatin. This, however, leads to an apparent paradox: In cells that slowly replicate in the organism, H3.3 will accumulate with time, opening the way to aberrant effects on heterochromatin. Here, we review the indications that H3.3 is expected both to be incorporated in the heterochromatin of slowly replicating cells and to retain its functional downstream effects. Implications for organismal aging are discussed.


Subject(s)
Aging/genetics , Chromatin/genetics , DNA Replication/genetics , Histones/metabolism , Transcriptional Activation/genetics , Animals , Histones/genetics , Humans , Sex Factors
15.
Eur J Hum Genet ; 22(9): 1117-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24448543

ABSTRACT

Mechanisms that regulate attachment of the scaffold/matrix attachment regions (S/MARs) to the nuclear matrix remain largely unknown. We have studied the effect of simple sequence length polymorphism (SSLP), DNA methylation and chromatin organization in an S/MAR implicated in facioscapulohumeral dystrophy (FSHD), a hereditary disease linked to a partial deletion of the D4Z4 repeat array on chromosome 4q. This FSHD-related nuclear matrix attachment region (FR-MAR) loses its efficiency in myoblasts from FSHD patients. Three criteria were found to be important for high-affinity interaction between the FR-MAR and the nuclear matrix: the presence of a specific SSLP haplotype in chromosomal DNA, the methylation of one specific CpG within the FR-MAR and the absence of histone H3 acetylated on lysine 9 in the relevant chromatin fragment.


Subject(s)
Epigenesis, Genetic , Matrix Attachment Regions/genetics , Microsatellite Repeats/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Nuclear Matrix/metabolism , Polymorphism, Genetic , Acetylation , Adult , Base Sequence , Cell Line, Tumor , Cells, Cultured , Chromatin/metabolism , CpG Islands , DNA Methylation , Female , Histones/metabolism , Humans , Male , Middle Aged , Molecular Sequence Data , Myoblasts/metabolism , Protein Binding
16.
Blood ; 123(13): 2044-53, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24452204

ABSTRACT

In mantle cell lymphoma (MCL), one allele of the cyclin D1 (Ccnd1) gene is translocated from its normal localization on chromosome 11 to chromosome 14. This is considered as the crucial event in the transformation process of a normal naive B-cell; however, the actual molecular mechanism leading to Ccnd1 activation remains to be deciphered. Using a combination of three-dimensional and immuno-fluorescence in situ hybridization experiments, the radial position of the 2 Ccnd1 alleles was investigated in MCL-derived cell lines and malignant cells from affected patients. The translocated Ccnd1 allele was observed significantly more distant from the nuclear membrane than its nontranslocated counterpart, with a very high proportion of IgH-Ccnd1 chromosomal segments localized next to a nucleolus. These perinucleolar areas were found to contain active RNA polymerase II (PolII) clusters. Nucleoli are rich in nucleolin, a potent transcription factor that we found to bind sites within the Ccnd1 gene specifically in MCL cells and to activate Ccnd1 transcription. We propose that the Ccnd1 transcriptional activation in MCL cells relates to the repositioning of the rearranged IgH-Ccnd1-carrying chromosomal segment in a nuclear territory with abundant nucleolin and active PolII molecules. Similar transforming events could occur in Burkitt and other B-cell lymphomas.


Subject(s)
Cell Nucleolus/metabolism , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, Mantle-Cell/genetics , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Transcriptional Activation , Active Transport, Cell Nucleus/physiology , CCCTC-Binding Factor , Cell Line, Tumor , Cyclin D1/genetics , Genes, Neoplasm , HeLa Cells , Humans , Protein Transport , Repressor Proteins/metabolism , Nucleolin
17.
J Biol Chem ; 288(49): 34989-5002, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24145033

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant hereditary neuromuscular disorder linked to the deletion of an integral number of 3.3-kb-long macrosatellite repeats (D4Z4) within the subtelomeric region of chromosome 4q. Most genes identified in this region are overexpressed in FSHD myoblasts, including the double homeobox genes DUX4 and DUX4c. We have carried out a simultaneous miRNome/transcriptome analysis of FSHD and control primary myoblasts. Of 365 microRNAs (miRNAs) analyzed in this study, 29 were found to be differentially expressed between FSHD and normal myoblasts. Twenty-one microRNAs (miR-1, miR-7, miR-15a, miR-22, miR-30e, miR-32, miR-107, miR-133a, miR-133b, miR-139, miR-152, miR-206, miR-223, miR-302b, miR-331, miR-362, miR-365, miR-382, miR-496, miR-532, miR-654, and miR-660) were up-regulated, and eight were down-regulated (miR-15b, miR-20b, miR-21, miR-25, miR-100, miR-155, miR-345, and miR-594). Twelve of the miRNAs up-regulated in FHSD were also up-regulated in the cells ectopically expressing DUX4c, suggesting that this gene could regulate miRNA gene transcription. The myogenic miRNAs miR-1, miR-133a, miR-133b, and miR-206 were highly expressed in FSHD myoblasts, which nonetheless did not prematurely enter myogenic differentiation. This could be accounted for by the fact that in FSHD myoblasts, functionally important target genes, including cell cycle, DNA damage, and ubiquitination-related genes, escape myogenic microRNA-induced repression.


Subject(s)
MicroRNAs/genetics , MicroRNAs/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts, Skeletal/metabolism , Adult , Cell Differentiation/genetics , Cells, Cultured , Down-Regulation , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Humans , Male , Middle Aged , Muscle Development/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts, Skeletal/pathology , Up-Regulation , Young Adult
18.
BMC Genomics ; 14: 265, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23597168

ABSTRACT

BACKGROUND: miRNA profiling performed in myogenic cells and biopsies from skeletal muscles has previously identified miRNAs involved in myogenesis. RESULTS: Here, we have performed miRNA transcriptome profiling in human affinity-purified CD56+ myoblasts induced to differentiate in vitro. In total, we have identified 60 miRNAs differentially expressed during myogenic differentiation. Many were not known for being differentially expressed during myogenic differentiation. Of these, 14 (miR-23b, miR-28, miR-98, miR-103, miR-107, miR-193a, miR-210, miR-324-5p, miR-324-3p, miR-331, miR-374, miR-432, miR-502, and miR-660) were upregulated and 6 (miR-31, miR-451, miR-452, miR-565, miR-594 and miR-659) were downregulated. mRNA transcriptome profiling performed in parallel resulted in identification of 6,616 genes differentially expressed during myogenic differentiation. CONCLUSIONS: This simultaneous miRNA/mRNA transcriptome profiling allowed us to predict with high accuracy target genes of myogenesis-related microRNAs and to deduce their functions.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/genetics , Muscle Development/genetics , Muscle, Skeletal/metabolism , Myoblasts/metabolism , RNA, Messenger/metabolism , CD56 Antigen/genetics , Down-Regulation , Gene Expression Profiling , Humans , MicroRNAs/physiology , RNA, Messenger/genetics , Up-Regulation
19.
PLoS One ; 8(1): e53033, 2013.
Article in English | MEDLINE | ID: mdl-23326377

ABSTRACT

Little is known about differences between induced pluripotent stem cells produced from tissues originating from the same germ layer. We have generated human myoblast-derived iPS cells by retroviral transduction of human primary myoblasts with the OCT3/4, SOX2, KLF4 and MYC coding sequences and compared them to iPS produced from human primary fibroblasts. When cultivated in vitro, these iPS cells proved similar to human embryonic stem cells in terms of morphology, expression of embryonic stemness markers and gene promoter methylation patterns. Embryonic bodies were derived that expressed endodermal, mesodermal as well as ectodermal markers. A comparative analysis of transcription patterns revealed significant differences in the gene expression pattern between myoblast- and fibroblast-derived iPS cells. However, these differences were reduced in the mesenchymal stem cells derived from the two iPS cell types were compared.


Subject(s)
Cell Differentiation/genetics , Fibroblasts/metabolism , Gene Expression Profiling , Induced Pluripotent Stem Cells/metabolism , Myoblasts/metabolism , Animals , Cells, Cultured , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Germ Layers/cytology , Germ Layers/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Muscle Development/genetics , Myoblasts/cytology , Octamer Transcription Factor-3/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-myc/genetics , Retroviridae/genetics , SOXB1 Transcription Factors/genetics , Transduction, Genetic
20.
Genome Res ; 23(2): 331-40, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23038767

ABSTRACT

We have developed an approach termed PUB-NChIP (proximity utilizing biotinylation with native ChIP) to purify and study the protein composition of chromatin in proximity to a nuclear protein of interest. It is based on coexpression of (1) a protein of interest, fused with the bacterial biotin ligase BirA, together with (2) a histone fused to a biotin acceptor peptide (BAP), which is specifically biotinylated by BirA-fusion in the proximity of the protein of interest. Using the RAD18 protein as a model, we demonstrate that the RAD18-proximal chromatin is enriched in some H4 acetylated species. Moreover, the RAD18-proximal chromatin containing a replacement histone H2AZ has a different pattern of H4 acetylation. Finally, biotin pulse-chase experiments show that the H4 acetylation pattern starts to resemble the acetylation pattern of total H4 after the proximity of chromatin to RAD18 has been lost.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/metabolism , Nuclear Proteins/metabolism , Acetylation , Biotinylation/methods , Cell Line , Histones , Humans , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL
...