13 research outputs found

    Long term culture of mesenchymal stem cells in hypoxia promotes a genetic program maintaining their undifferentiated and multipotent status

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>In the bone marrow, hematopietic and mesenchymal stem cells form a unique niche in which the oxygen tension is low. Hypoxia may have a role in maintaining stem cell fate, self renewal and multipotency. However, whereas most studies addressed the effect of transient <it>in vitro </it>exposure of MSC to hypoxia, permanent culture under hypoxia should reflect the better physiological conditions.</p> <p>Results</p> <p>Morphologic studies, differentiation and transcriptional profiling experiments were performed on MSC cultured in normoxia (21% O<sub>2</sub>) versus hypoxia (5% O<sub>2</sub>) for up to passage 2. Cells at passage 0 and at passage 2 were compared, and those at passage 0 in hypoxia generated fewer and smaller colonies than in normoxia. In parallel, MSC displayed (>4 fold) inhibition of genes involved in DNA metabolism, cell cycle progression and chromosome cohesion whereas transcripts involved in adhesion and metabolism (CD93, ESAM, VWF, PLVAP, ANGPT2, LEP, TCF1) were stimulated. Compared to normoxic cells, hypoxic cells were morphologically undifferentiated and contained less mitochondrias. After this lag phase, cells at passage 2 in hypoxia outgrew the cells cultured in normoxia and displayed an enhanced expression of genes (4-60 fold) involved in extracellular matrix assembly (SMOC2), neural and muscle development (NOG, GPR56, SNTG2, LAMA) and epithelial development (DMKN). This group described herein for the first time was assigned by the Gene Ontology program to "plasticity".</p> <p>Conclusion</p> <p>The duration of hypoxemia is a critical parameter in the differentiation capacity of MSC. Even in growth promoting conditions, hypoxia enhanced a genetic program that maintained the cells undifferentiated and multipotent. This condition may better reflect the <it>in vivo </it>gene signature of MSC, with potential implications in regenerative medicine.</p

    The human RNA polymerase II-associated factor 1 (hPaf1): a new regulator of cell-cycle progression.

    Get PDF
    BACKGROUND: The human PAF (hPAF) complex is part of the RNA polymerase II transcription apparatus and regulates multiple steps in gene expression. Further, the yeast homolog of hPaf1 has a role in regulating the expression of a subset of genes involved in the cell-cycle. We therefore investigated the role of hPaf1 during progression of the cell-cycle. METHODOLOGY/FINDINGS: Herein, we report that the expression of hPaf1, a subunit of the hPAF complex, increases with cell-cycle progression and is regulated in a cell-cycle dependant manner. hPaf1 specifically regulates a subclass of genes directly implicated in cell-cycle progression during G1/S, S/G2, and G2/M. In prophase, hPaf1 aligns in filament-like structures, whereas in metaphase it is present within the pole forming a crown-like structure, surrounding the centrosomes. Moreover, hPaf1 is degraded during the metaphase to anaphase transition. In the nucleus, hPaf1 regulates the expression of cyclins A1, A2, D1, E1, B1, and Cdk1. In addition, expression of hPaf1 delays DNA replication but favors the G2/M transition, in part through microtubule assembly and mitotic spindle formation. CONCLUSION/SIGNIFICANCE: Our results identify hPaf1 and the hPAF complex as key regulators of cell-cycle progression. Mutation or loss of stoichiometry of at least one of the members may potentially lead to cancer development

    The Human RNA Polymerase II-Associated Factor 1 (hPaf1): A New Regulator of Cell-Cycle Progression

    Get PDF
    The human PAF (hPAF) complex is part of the RNA polymerase II transcription apparatus and regulates multiple steps in gene expression. Further, the yeast homolog of hPaf1 has a role in regulating the expression of a subset of genes involved in the cell-cycle. We therefore investigated the role of hPaf1 during progression of the cell-cycle.Herein, we report that the expression of hPaf1, a subunit of the hPAF complex, increases with cell-cycle progression and is regulated in a cell-cycle dependant manner. hPaf1 specifically regulates a subclass of genes directly implicated in cell-cycle progression during G1/S, S/G2, and G2/M. In prophase, hPaf1 aligns in filament-like structures, whereas in metaphase it is present within the pole forming a crown-like structure, surrounding the centrosomes. Moreover, hPaf1 is degraded during the metaphase to anaphase transition. In the nucleus, hPaf1 regulates the expression of cyclins A1, A2, D1, E1, B1, and Cdk1. In addition, expression of hPaf1 delays DNA replication but favors the G2/M transition, in part through microtubule assembly and mitotic spindle formation.Our results identify hPaf1 and the hPAF complex as key regulators of cell-cycle progression. Mutation or loss of stoichiometry of at least one of the members may potentially lead to cancer development

    hPaf1 is ubiquitinylated and degraded at the metaphase/anaphase transition.

    No full text
    <p>Panc1 cells were grown at low density on cover slips for 24 h, and arrested at G1/S (T0) and G2/M (N0) boundaries by double thymidine and thymidine/nocodazole blocks, respectively. Acetone/methanol (1∶1)-fixed Panc1 cells were labeled using a FITC-conjugated anti-hPaf1 monoclonal antibody and counterstained by propidium iodide. Up to 10 fields were observed by confocal microscopy for cells after 12 h of double thymidine treatment release (T+12) and compared with 10 fields of thymidine/nocodazole treated cells just before release (N0). Representative pictures are presented in A). The black arrows indicate the inter-chromatine accumulation of hPaf1 in the prophase for the cells in N0; accumulation was not detected at T+12. B) Proteins from nuclear extracts performed at T0, T+8, N0, and S0 (shake off cells in N0) were immunoprecipitated using the rabbit polyclonal antibody anti-ubiquitin (Calbiochem), and immunoblotted with anti-hPaf1 antibody. An aliquot of the nuclear protein extracts was loaded as a control for hPaf1 expression (input), and immunoprecipitation was carried out using an isotype IgG control antibody as a negative control. C) hPaf1 degradation by the proteasome was confirmed using the proteasome inhibitor MG132. After double thymidine block, the cells were released either in ethanol control or MG132 (20 ”M) for 12 h. Protein lysates were imunoprecipitated using the rabbit polyclonal antibody anti-ubiquitin (Calbiochem) and immunoblotted with anti-hPaf1 antibody. D) Panc1, CD18/HPAF, and human fibroblast cells were arrested at the G1/S boundary and released for 12 h to reach a maximum of mitosis. The cells were analyzed for hPaf1 expression and sub cellular localization by confocal. In the prophase, hPaf1 was found aligned on filament-like structures (D3) to be concentrated at the pole in the metaphase (D1, D2: black arrow). The Z-section (D4, D5, and D6) revealed that in the metaphase, hPaf1 formed a crown at both poles of the cells, potentially surrounding the centrosomes (white arrow). E) CD18/HPAF cells were arrested at the G1/S boundary and released for 12 h to reach a maximum of mitosis. The cells were analyzed for hPaf1 and tubulin expression and subcellular localization by confocal. In metaphase, hPaf1 co-localized with tubulin, confirming the centrosomal localization of hPaf1.</p

    hPaf1 is a key regulator for temporal transcription of cyclins.

    No full text
    <p>A) Chromatine immunoprecipitation using hPaf1 anitbody for cyclins A1, A2, E1, D1, B1, and the Cdk1. An aliquot of the DNA extract before immunoprecipitation was used as a positive control for the PCR amplification. B) Analysis of cyclins E1, B1, D1, and A2 expression was done by relative competitive dropping-PCR on T0 and T+8 synchronized Panc1 cells. To measure the effect of hPaf1 expression, hPaf1 was down-regulated using the siRNA technology. C) Values of the dropping-PCR were adjusted for assay variation by dividing the integrated optical density of cyclins A2, B1, D1, and E1 mRNA by the integrated optical density of GAPDH mRNA. D) Suppression of hPaf1 expression by the Panc1 cells after transfection with siRNA oligonucleotide was measured on the same RNAs used for the dropping-PCR by one-step real time RT-PCR using a TaqMan probe following the absolute standard curve method. Standard curves were established using hPaf1 and GAPDH mimics. Sequences and conditions used in this experiment are available upon request.</p

    Localization of hPaf1 during the cell cycle in Panc1 cells.

    No full text
    <p>hPaf1 expression was monitored in a synchronously growing population of Panc1 cells. Panc1 cells were arrested at the G1/S boundary by double thymidine block, then released in normal medium containing serum (time 0 = T0) and analyzed every 2 h for 24 h by flow cytometry (after propidium iodide staining), confocal microscopy, and Western blotting of cytoplasmic and nuclear cell fractions. A) Eighty percent of the cells were blocked at the G1/S boundary. After release, the cells progressed into the cell cycle synchronously to reach a peak of mitosis after 10–12 h. Representative fields from the confocal analysis are presented in B). The cells were grown at low density on cover slips for 24 h, and arrested at the G1/S boundary by double thymidine incorporation. The acetone/methanol (1∶1)-fixed Panc1 cells were labeled using a FITC-conjugated anti-hPaf1 monoclonal antibody and counterstained by propidium iodide. The amount of fluorescence detected for the cells in T+12 (G2/M) was twice the level detected in T0 (G1/S). Up to 10 fields were monitored for each phase of the cell cycle. Cells in the prophase, metaphase, anaphase, and telophase showed very low to no expression of hPaf1. C) Nuclear and cytoplasmic protein extracts were prepared every 2 h and analyzed by immunoblotting using anti-hPaf1 and anti-hLeo1 antibodies. The JLA-20 antibody recognizing ÎČ actin was used as a loading control.</p

    hPaf1 during cell cycle progression.

    No full text
    <p>hPaf1 expression initiates in early G1, increases during the S-phase and reaches a maximum level in early G2. Synthesized as a 60 kDa protein, hPaf1 translocates as an 80 kDa protein into the nucleus. In the nucleus, hPaf1 directly regulates the expression of the cyclins A1, A2, D1, E1, B1. In S phase, hPaf1 interacts with the DNA polymerase α-primase complex and delays the onset of DNA replication. At mitosis, hPaf1 migrates to the cellular pole, forming a crown surrounding the centrosomes. It is then possibly shuttled via tubulin filaments to be degraded by the proteasome.</p
    corecore