Volume 132, Issue 1, Pages (January 2007)

Slides:



Advertisements
Similar presentations
Figure S1 A MRC5 Control Chk1 TdR(h): γH2AX Cleaved casp3 RPA34
Advertisements

Volume 131, Issue 1, Pages (July 2006)
Volume 124, Issue 7, Pages (June 2003)
Volume 133, Issue 1, Pages (July 2007)
Volume 14, Issue 4, Pages (May 2004)
Volume 132, Issue 2, Pages (February 2007)
Volume 145, Issue 2, Pages (August 2013)
Volume 60, Issue 2, Pages (August 2001)
Volume 131, Issue 1, Pages (July 2006)
Volume 135, Issue 3, Pages (September 2008)
Membrane-Tethered Intracellular Domain of Amphiregulin Promotes Keratinocyte Proliferation  Stefan W. Stoll, Philip E. Stuart, Sylviane Lambert, Alberto.
by Anita Boyapati, Ming Yan, Luke F. Peterson, Joseph R
Tocotrienols Induce Apoptosis and Autophagy in Rat Pancreatic Stellate Cells Through the Mitochondrial Death Pathway  Mariana Rickmann, Eva C. Vaquero,
Volume 132, Issue 2, Pages (February 2007)
Volume 133, Issue 1, Pages (July 2007)
The Mitochondrial Protein hTID-1 Partners With the Caspase-Cleaved Adenomatous Polyposis Cell Tumor Suppressor to Facilitate Apoptosis  Jiang Qian, Erin.
Volume 133, Issue 1, Pages (July 2007)
Induction of DNA double‐strand breaks and activation of the ataxia telangiectasia mutated pathway in post‐mitotic cells in response to camptothecin. Induction.
MK-8776, a novel Chk1 inhibitor, exhibits an improved radiosensitizing effect compared to UCN-01 by exacerbating radiation-induced aberrant mitosis  Motofumi.
Volume 129, Issue 3, Pages (September 2005)
Volume 35, Issue 3, Pages (August 2009)
Volume 118, Issue 4, Pages (April 2000)
Volume 134, Issue 1, Pages (January 2008)
Volume 117, Issue 4, Pages (October 1999)
Volume 135, Issue 3, Pages (September 2008)
Volume 135, Issue 1, Pages (July 2008)
Volume 4, Issue 1, Pages (July 2001)
Volume 30, Issue 1, Pages (April 2008)
The DNA-Mismatch Repair Enzyme hMSH2 Modulates UV-B-Induced Cell Cycle Arrest and Apoptosis in Melanoma Cells  Markus Seifert, Stefan J. Scherer, Wilfried.
Volume 123, Issue 1, Pages (July 2002)
Volume 136, Issue 4, Pages e3 (April 2009)
A DNA Damage Response Pathway Controlled by Tel1 and the Mre11 Complex
Volume 115, Issue 1, Pages (July 1998)
Volume 138, Issue 5, Pages e2 (May 2010)
A p53-Dependent Checkpoint Pathway Prevents Rereplication
Volume 138, Issue 1, Pages (January 2010)
Volume 131, Issue 4, Pages (October 2006)
Decreased Growth Inhibitory Responses of Squamous Carcinoma Cells to Interferon-γ Involve Failure to Recruit cki Proteins into cdk2 Complexes  Beth L.
Akio Horiguchi, Mototsugu Oya, Ken Marumo, Masaru Murai 
Volume 145, Issue 2, Pages (August 2013)
Volume 133, Issue 4, Pages (October 2007)
The Mammalian UV Response
A Role for H2B Ubiquitylation in DNA Replication
Volume 132, Issue 5, Pages (May 2007)
Volume 8, Issue 1, Pages (January 1998)
Volume 35, Issue 3, Pages (August 2009)
Volume 17, Issue 1, Pages (January 2005)
Katja Wassmann, Théodora Niault, Bernard Maro  Current Biology 
Raymond L. Warters, Patrick J. Adamson, Christopher D. Pond, Sancy A
A Role for H2B Ubiquitylation in DNA Replication
Volume 56, Issue 5, Pages (December 2014)
José Antonio Tercero, Maria Pia Longhese, John F.X Diffley 
Volume 15, Issue 6, Pages (September 2004)
New Histone Incorporation Marks Sites of UV Repair in Human Cells
Volume 8, Issue 3, Pages (January 1998)
The Prolyl Isomerase Pin1 Functions in Mitotic Chromosome Condensation
Interferon-γ-Mediated Growth Regulation of Melanoma Cells: Involvement of STAT1- Dependent and STAT1-Independent Signals  Anja Bosserhoff  Journal of Investigative.
Volume 47, Issue 4, Pages (August 2012)
Cdc18 Enforces Long-Term Maintenance of the S Phase Checkpoint by Anchoring the Rad3-Rad26 Complex to Chromatin  Damien Hermand, Paul Nurse  Molecular.
Volume 67, Issue 6, Pages (June 2005)
Robin M. Ricke, Anja-Katrin Bielinsky  Molecular Cell 
Volume 3, Issue 5, Pages (May 2001)
Volume 132, Issue 2, Pages (February 2007)
Ultraviolet-B-Induced G1 Arrest is Mediated by Downregulation of Cyclin-Dependent Kinase 4 in Transformed Keratinocytes Lacking Functional p53  Arianna.
Sean P. Cullen, Conor J. Kearney, Danielle M. Clancy, Seamus J. Martin 
Volume 22, Issue 3, Pages (May 2006)
Volume 41, Issue 4, Pages (February 2011)
GADD45 Regulates G2/M Arrest, DNA Repair, and Cell Death in Keratinocytes Following Ultraviolet Exposure  Tomoko Maeda, Atef N. Hanna, Adrian B. Sim,
RA-9 induces G2–M cell-cycle arrest and caspase-mediated apoptosis in ovarian cancer cells. RA-9 induces G2–M cell-cycle arrest and caspase-mediated apoptosis.
Yun-Gui Yang, Tomas Lindahl, Deborah E. Barnes  Cell 
Presentation transcript:

Volume 132, Issue 1, Pages 221-235 (January 2007) 5-ASA Affects Cell Cycle Progression in Colorectal Cells by Reversibly Activating a Replication Checkpoint  M. Gloria Luciani, Christoph Campregher, John M. Fortune, Thomas A. Kunkel, Christoph Gasche  Gastroenterology  Volume 132, Issue 1, Pages 221-235 (January 2007) DOI: 10.1053/j.gastro.2006.10.016 Copyright © 2007 AGA Institute Terms and Conditions

Figure 1 Dose-response effect of 5-ASA on the growth of human colon cells. HCT116 (open bars), HT29 (shaded bars), and LoVo (solid bars) cells were plated at a density of 104 cells/well in 96-well microtiter plates. Twenty-four hours after seeding, the cells were treated with 5-ASA-containing culture medium (0 to 20 mmol/L) for 48 hours. The antiproliferative effects were measured by the MTT assay. The values are indicated as relative units (RU) to the control and represent the mean of 4 separate measurements. 5-ASA caused a dose-dependent growth reduction in all colon cancer cells. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions

Figure 2 Cell cycle distribution in 5-ASA-treated cells. (A) HT29, HCT116, and LoVo cell lines were treated with 5, 10, or 20 mmol/L 5-ASA for 48 hours. After staining with propidium iodide, the cellular DNA content was visualized by flow cytometry, and the cell cycle distribution was analyzed using CellQuest flow cytometry software (BD Technology). All cell lines showed changes in the cell cycle profile following 5-ASA exposure within 48 hours of treatment. In further experiments, HCT116 cells (B) and HT29 cells (C) were exposed to 5, 10, or 20 mmol/L 5-ASA for 48 hours. Following treatment, 5-ASA was removed, and the cells were pulsed with 10 μmol/L BrdU for 1 hour. After permeabilization and DNA-antigen unmasking, samples were stained with anti-BrdU antibody to visualize replicating cells and PI to visualize the total DNA content. Cells were analyzed by flow cytometry using CellQuest flow cytometry software (BD Technology). Both cell lines showed an increase in the percentage of cells in S phase following treatment with 5-ASA. (D) HCT116 cells were treated with low doses of 5-ASA (1.25–5 mmol/L) for 120 hours (upper panels) or 168 hours. After pulsing with 10 μmol/L BrdU and staining with anti-BrdU antibody and PI, cells were analyzed by flow cytometry. At later time points, an increase in the S-phase arrested population is visible at 1.25 mmol/L 5-ASA. In the flow cytometry dot blot, the upper region (R2) contains cells in S phase, the lower left region (R3) represents cells in G1, and the lower right region (R4) represents cells in G2/M. The flow cytometry profiles are representative of at least 3 independent experiments. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions

Figure 2 Cell cycle distribution in 5-ASA-treated cells. (A) HT29, HCT116, and LoVo cell lines were treated with 5, 10, or 20 mmol/L 5-ASA for 48 hours. After staining with propidium iodide, the cellular DNA content was visualized by flow cytometry, and the cell cycle distribution was analyzed using CellQuest flow cytometry software (BD Technology). All cell lines showed changes in the cell cycle profile following 5-ASA exposure within 48 hours of treatment. In further experiments, HCT116 cells (B) and HT29 cells (C) were exposed to 5, 10, or 20 mmol/L 5-ASA for 48 hours. Following treatment, 5-ASA was removed, and the cells were pulsed with 10 μmol/L BrdU for 1 hour. After permeabilization and DNA-antigen unmasking, samples were stained with anti-BrdU antibody to visualize replicating cells and PI to visualize the total DNA content. Cells were analyzed by flow cytometry using CellQuest flow cytometry software (BD Technology). Both cell lines showed an increase in the percentage of cells in S phase following treatment with 5-ASA. (D) HCT116 cells were treated with low doses of 5-ASA (1.25–5 mmol/L) for 120 hours (upper panels) or 168 hours. After pulsing with 10 μmol/L BrdU and staining with anti-BrdU antibody and PI, cells were analyzed by flow cytometry. At later time points, an increase in the S-phase arrested population is visible at 1.25 mmol/L 5-ASA. In the flow cytometry dot blot, the upper region (R2) contains cells in S phase, the lower left region (R3) represents cells in G1, and the lower right region (R4) represents cells in G2/M. The flow cytometry profiles are representative of at least 3 independent experiments. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions

Figure 3 5-ASA-induced S-phase accumulation is p53- and MMR independent. (A) HCT116, HCT116p53−/− (p53-null), and HT29 were blotted for the presence or not of the p53 protein. As shown by the Western blot, no expression of p53 was detected in HCT116p53−/− cells. (B) HCT116 and HCT116+chr3 were analyzed for the expression of the MMR proteins hMSH2 and hMLH1. While HCT116+chr3 expressed both MMR components, only hMSH2 was detectable in HCT116 cells. (C) HCT116, HCT116+chr3 (expressing hMHL1), and HCT116p53−/− cell lines were treated with 0 or 10 mmol/L 5-ASA for 48 hours. Following treatment, 5-ASA was removed, and the cells pulsed with 10 μmol/L BrdU for 1 hour. After permeabilization and DNA-antigen unmasking, samples were stained with anti-BrdU antibody and PI. Cells were analyzed by flow cytometry using CellQuest flow cytometry software (BD Technology). All cell lines showed a comparable increase in the percentage of cells in S phase following treatment with 10 mmol/L 5-ASA. In the flow cytometry dot blot, the upper region (R2) contains cells in S phase, the lower left region (R3) represents cells in G1, and the lower right region (R4) represents cells in G2/M. The flow cytometry profiles are representative of at least 3 independent experiments. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions

Figure 4 5-ASA induces a replication checkpoint by inducing phosphorylation of checkpoint proteins Chk1 and Rad17 and activates the p53-dependent pathway. (A) Phosphorylation status of Chk1 at Ser317 was analyzed in total lysates of HCT116 and HT29 cells treated with 0, 20, or 40 mmol/L 5-ASA for 8 hours. The checkpoint kinase resulted strongly phosphorylated at the ATR-targeted residue 317 in both cell lines following treatment, consistent with the activation of a replication checkpoint in these cells. An antibody anti-α-tubulin was used as loading control to show an equal amount of proteins in our Western blot. (B) HCT116 cells were treated with 0, 20, or 40 mmol/L 5-ASA for 2–8 hours One hundred micrograms of total lysates were analyzed by Western blot for the phosphorylation status of p53 Ser15, Chk1 Ser354, and Rad17 Ser645, as well as for the total levels of p21waf1/cip1 All checkpoint proteins tested were phosphorylated within 2 hours of 5-ASA treatment, and phosphorylation correlated with increased expression of the CDK-inhibitor p21. An antibody anti-α-tubulin was used as loading control to show an equal amount of proteins in our Western blot. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions

Figure 5 Checkpoint sensors ATR and claspin and replication proteins cdc45, MCMs, and RPA are recruited onto the DNA upon treatment with 5-ASA. (A) Chromatin-bound fractions of untreated HCT116 were analyzed at time points 0, 2, 4, 8, 16, and 24 hours for the presence of replication proteins recruited onto the DNA. No significant difference in the loading of ATR or claspin was detected in our samples. (B) The DNA-bound protein fraction of HCT116 cells treated with 20 or 40 mmol/L 5-ASA for 2–8 hours was analyzed by Western blot for the presence of checkpoint sensor proteins ATR and Claspin, as well as the replication proteins MCM2, cdc45, and RPA. Following exposure to 5-ASA, ATR, claspin, cdc45, and RPA accumulated onto origins of DNA within 2 hours, whereas levels of MCM2, a component of the helicase MCM2-7 complex, increased onto the chromatin after 8 hours, suggesting a reduction in the DNA replication rate in treated cells. Accumulation of checkpoint proteins onto origins of replication correlated with increased levels of phosphorylation of the γH2AX at Ser139 (bottom panel), consistent with the activation of an ATR-dependent replication checkpoint. Total levels of the histone H3 are also shown as loading control. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions

Figure 6 Colon cells present aberrant mitotic cells upon 5-ASA-treatment. HT29 (A and B) and HCT116 (C) cells were exposed to 20 mmol/L 5-ASA for 48 hours. Cells were fixed in 4% PFA, permeabilized, and incubated with mAb antitubulin followed by FITC-conjugated secondary anti-mouse IgG. Cells were visualized using a Carl Zeiss Axioplan 2 fluorescence microscope, and images were captured with a Photometrics CoolSNAP fx digital camera (Roper Scientific) and processed using MetaMorph (Molecular Devices) and Canvas X (ACD Systems) software (original magnification, ×300). Arrows show aberrant mitotic cells (HT29; A and B) or giant cells with multilobular nuclei (HCT116; C) that appeared upon 48-hour treatment with 5-ASA. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions

Figure 7 5-ASA-dependent S-phase arrest is reversible in colorectal cancer cells. HCT116 cells were treated with 0 or 10 mmol/L 5-ASA for 48 hours. Viable cells were either reverted to normal medium or kept in 10 mmol/L 5-ASA for an additional 48 hours. Cells were pulse labelled with 10 μmol/L BrdU, harvested, and the BrdU-positive cells, as well as the total DNA content, analyzed upon anti-BrdU and propidium iodide staining (PI) by flow cytometry. The S-phase arrest observed at 10 mmol/L 5-ASA was almost completely reversible upon drug removal, indicating that this concentration of 5-ASA does not cause permanent cell damage. In the flow cytometry dot blot, the upper region (R2) contains cells in S phase, the lower left region (R3) represents cells in G1, and the lower right region (R4) represents cells in G2/M. The flow cytometry profiles are representative of at least 3 independent experiments. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions

Figure 8 Model of activation of the 5-ASA-induced checkpoint responses in colon cells. Following environmental signals, 5-ASA may synergize with checkpoint sensor proteins upstream of an ATR-dependent replication checkpoint pathway. ATR, activated by association with ATRIP and recruited onto DNA replication origins, phosphorylates Chk1 on Ser345 and Ser317 and Rad17 on Ser645. The loading of claspin onto foci of replication also mediates stabilization and activation of the checkpoint kinase Chk1 and mediates the activation of the S-phase arrest. The ultimate target of Chk1 that mediates the replication stalling is yet unknown. In addition, ATR phosphorylates the tumor suppressor protein p53 on Ser15, which, although not involved in the replication checkpoint, ultimately results in p53 stabilization and transcriptional activation of downstream targets such as the CDK inhibitor p21waf1/cip1. Similarly, targets of the checkpoint kinase Chk1, not involved in the replication checkpoint, such as Cdc25A/C, are expected to be phosphorylated. Gastroenterology 2007 132, 221-235DOI: (10.1053/j.gastro.2006.10.016) Copyright © 2007 AGA Institute Terms and Conditions