1
|
Oropeza E, Seker S, Carrel S, Mazumder A, Lozano D, Jimenez A, VandenHeuvel SN, Noltensmeyer DA, Punturi NB, Lei JT, Lim B, Waltz SE, Raghavan SA, Bainbridge MN, Haricharan S. Molecular portraits of cell cycle checkpoint kinases in cancer evolution, progression, and treatment responsiveness. SCIENCE ADVANCES 2023; 9:eadf2860. [PMID: 37390209 PMCID: PMC10313178 DOI: 10.1126/sciadv.adf2860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 05/26/2023] [Indexed: 07/02/2023]
Abstract
Cell cycle dysregulation is prerequisite for cancer formation. However, it is unknown whether the mode of dysregulation affects disease characteristics. Here, we conduct comprehensive analyses of cell cycle checkpoint dysregulation using patient data and experimental investigations. We find that ATM mutation predisposes the diagnosis of primary estrogen receptor (ER)+/human epidermal growth factor (HER)2- cancer in older women. Conversely, CHK2 dysregulation induces formation of metastatic, premenopausal ER+/HER2- breast cancer (P = 0.001) that is treatment-resistant (HR = 6.15, P = 0.01). Lastly, while mutations in ATR alone are rare, ATR/TP53 co-mutation is 12-fold enriched over expected in ER+/HER2- disease (P = 0.002) and associates with metastatic progression (HR = 2.01, P = 0.006). Concordantly, ATR dysregulation induces metastatic phenotypes in TP53 mutant, not wild-type, cells. Overall, we identify mode of cell cycle dysregulation as a distinct event that determines subtype, metastatic potential, and treatment responsiveness, providing rationale for reconsidering diagnostic classification through the lens of the mode of cell cycle dysregulation..
Collapse
Affiliation(s)
- Elena Oropeza
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sinem Seker
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sabrina Carrel
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Aloran Mazumder
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Daniel Lozano
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Athena Jimenez
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | | | - Nindo B. Punturi
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jonathan T. Lei
- Lester and Sue Smith Breast Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bora Lim
- Lester and Sue Smith Breast Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Oncology/Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
- Research Service, Cincinnati Veteran's Affairs Medical Center, 3200 Vine St., Cincinnati, OH, USA
| | | | | | - Svasti Haricharan
- Aging and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
2
|
Hasan A, Rizvi SF, Parveen S, Mir SS. Molecular chaperones in DNA repair mechanisms: Role in genomic instability and proteostasis in cancer. Life Sci 2022; 306:120852. [DOI: 10.1016/j.lfs.2022.120852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 01/09/2023]
|
3
|
Shadfar S, Brocardo M, Atkin JD. The Complex Mechanisms by Which Neurons Die Following DNA Damage in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23052484. [PMID: 35269632 PMCID: PMC8910227 DOI: 10.3390/ijms23052484] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/12/2022] [Accepted: 02/17/2022] [Indexed: 01/18/2023] Open
Abstract
Human cells are exposed to numerous exogenous and endogenous insults every day. Unlike other molecules, DNA cannot be replaced by resynthesis, hence damage to DNA can have major consequences for the cell. The DNA damage response contains overlapping signalling networks that repair DNA and hence maintain genomic integrity, and aberrant DNA damage responses are increasingly described in neurodegenerative diseases. Furthermore, DNA repair declines during aging, which is the biggest risk factor for these conditions. If unrepaired, the accumulation of DNA damage results in death to eliminate cells with defective genomes. This is particularly important for postmitotic neurons because they have a limited capacity to proliferate, thus they must be maintained for life. Neuronal death is thus an important process in neurodegenerative disorders. In addition, the inability of neurons to divide renders them susceptible to senescence or re-entry to the cell cycle. The field of cell death has expanded significantly in recent years, and many new mechanisms have been described in various cell types, including neurons. Several of these mechanisms are linked to DNA damage. In this review, we provide an overview of the cell death pathways induced by DNA damage that are relevant to neurons and discuss the possible involvement of these mechanisms in neurodegenerative conditions.
Collapse
Affiliation(s)
- Sina Shadfar
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia; (S.S.); (M.B.)
| | - Mariana Brocardo
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia; (S.S.); (M.B.)
| | - Julie D. Atkin
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia; (S.S.); (M.B.)
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC 3086, Australia
- Correspondence:
| |
Collapse
|
4
|
Kwok M, Agathanggelou A, Davies N, Stankovic T. Targeting the p53 Pathway in CLL: State of the Art and Future Perspectives. Cancers (Basel) 2021; 13:4681. [PMID: 34572908 PMCID: PMC8468925 DOI: 10.3390/cancers13184681] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 12/20/2022] Open
Abstract
The p53 pathway is a desirable therapeutic target, owing to its critical role in the maintenance of genome integrity. This is exemplified in chronic lymphocytic leukemia (CLL), one of the most common adult hematologic malignancies, in which functional loss of p53 arising from genomic aberrations are frequently associated with clonal evolution, disease progression, and therapeutic resistance, even in the contemporary era of CLL targeted therapy and immunotherapy. Targeting the 'undruggable' p53 pathway therefore arguably represents the holy grail of cancer research. In recent years, several strategies have been proposed to exploit p53 pathway defects for cancer treatment. Such strategies include upregulating wild-type p53, restoring tumor suppressive function in mutant p53, inducing synthetic lethality by targeting collateral genome maintenance pathways, and harnessing the immunogenicity of p53 pathway aberrations. In this review, we will examine the biological and clinical implications of p53 pathway defects, as well as our progress towards development of therapeutic approaches targeting the p53 pathway, specifically within the context of CLL. We will appraise the opportunities and pitfalls associated with these therapeutic strategies, and evaluate their place amongst the array of new biological therapies for CLL.
Collapse
Affiliation(s)
- Marwan Kwok
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK; (A.A.); (N.D.)
- Centre for Clinical Haematology, Queen Elizabeth Hospital Birmingham, Birmingham B15 2SY, UK
| | - Angelo Agathanggelou
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK; (A.A.); (N.D.)
| | - Nicholas Davies
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK; (A.A.); (N.D.)
| | - Tatjana Stankovic
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2SY, UK; (A.A.); (N.D.)
| |
Collapse
|
5
|
Magin S, Meher PK, Iliakis G. Nucleoside Analogs Radiosensitize G0 Cells by Activating DNA End Resection and Alternative End-Joining. Radiat Res 2021; 195:412-426. [PMID: 33755161 DOI: 10.1667/rade-20-00195.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/22/2021] [Indexed: 11/03/2022]
Abstract
Alternative end-joining (alt-EJ) is a DNA end resection-dependent, error-prone pathway utilized by vertebrate cells to repair DNA double-strand breaks (DSBs), but its engagement is linked to chromosomal translocations and genomic instability. Here, we report that when proliferating cells are exposed to ionizing radiation, treatment with nucleoside analogs (NAs) causes strong radiosensitization by increasing engagement of alt-EJ, while at the same time suppressing homologous recombination (HR) in S- and G2phase cells. This NA-mediated pathway shift may reflect a passive compensatory engagement of alt-EJ following HR suppression that is specific for S- and G2-phase cells, and/or the direct activation of alt-EJ throughout the cell cycle. To distinguish between these possibilities, we utilize here a cell culture model that exploits genetic and cell cycle-dependent inactivation of DSB repair pathways, to exclusively study alt-EJ and its modulation by NAs in murine and human cell lines. To this end, we allow LIG4-/--deficient cells to accumulate in G1/G0 phase by transfer to serum-deprived media and obtain cells deficient in c-NHEJ owing to the genetic LIG4 knockout, deficient in HR owing to the absence of S- or G2-phase cells, and compromised in their ability to carry out alt-EJ owing to their accumulation in G0. We find that in these cells irradiation and treatment with the NA, β-arabinofuranosyladenine (araA), and to a lesser degree with other NAs, promptly activates suppressed alt-EJ that now functions at levels approximating those of c-NHEJ in wild-type cells. Results at high dose (20 Gy) generated using pulsed-field gel electrophoresis (PFGE) are corroborated by results at low dose (1 Gy) generated by scoring 53BP1 foci. Strikingly, araA treatment activates a normally undetectable DNA-end-resection at DSBs, which requires ATR activity, but proceeds unimpeded after CtIP knockdown. Treatment with araA increases the formation of chromosomal aberrations and enhances radiation-induced cell killing. The results support direct stimulation of resection by NAs and alt-EJ as a mechanism of their documented radiosensitizing potential. We propose that this stimulation also occurs in repair-proficient cells and that it occurs throughout the cell cycle. It may therefore be harnessed to develop protocols combining NAs with radiation to treat human cancer.
Collapse
Affiliation(s)
- Simon Magin
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Prabodha Kumar Meher
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| | - George Iliakis
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| |
Collapse
|
6
|
Boudny M, Zemanova J, Khirsariya P, Borsky M, Verner J, Cerna J, Oltova A, Seda V, Mraz M, Jaros J, Jaskova Z, Spunarova M, Brychtova Y, Soucek K, Drapela S, Kasparkova M, Mayer J, Paruch K, Trbusek M. Novel CHK1 inhibitor MU380 exhibits significant single-agent activity in TP53-mutated chronic lymphocytic leukemia cells. Haematologica 2019; 104:2443-2455. [PMID: 30975914 PMCID: PMC6959166 DOI: 10.3324/haematol.2018.203430] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 04/05/2019] [Indexed: 11/09/2022] Open
Abstract
Introduction of small-molecule inhibitors of B-cell receptor signaling and BCL2 protein significantly improves therapeutic options in chronic lymphocytic leukemia. However, some patients suffer from adverse effects mandating treatment discontinuation, and cases with TP53 defects more frequently experience early progression of the disease. Development of alternative therapeutic approaches is, therefore, of critical importance. Here we report details of the anti-chronic lymphocytic leukemia single-agent activity of MU380, our recently identified potent, selective, and metabolically robust inhibitor of checkpoint kinase 1. We also describe a newly developed enantioselective synthesis of MU380, which allows preparation of gram quantities of the substance. Checkpoint kinase 1 is a master regulator of replication operating primarily in intra-S and G2/M cell cycle checkpoints. Initially tested in leukemia and lymphoma cell lines, MU380 significantly potentiated efficacy of gemcitabine, a clinically used inducer of replication stress. Moreover, MU380 manifested substantial single-agent activity in both TP53-wild type and TP53-mutated leukemia and lymphoma cell lines. In chronic lymphocytic leukemia-derived cell lines MEC-1, MEC-2 (both TP53-mut), and OSU-CLL (TP53-wt) the inhibitor impaired cell cycle progression and induced apoptosis. In primary clinical samples, MU380 used as a single-agent noticeably reduced the viability of unstimulated chronic lymphocytic leukemia cells as well as those induced to proliferate by anti-CD40/IL-4 stimuli. In both cases, effects were comparable in samples harboring p53 pathway dysfunction (TP53 mutations or ATM mutations) and TP53-wt/ATM-wt cells. Lastly, MU380 also exhibited significant in vivo activity in a xenotransplant mouse model (immunodeficient strain NOD-scid IL2Rγnull) where it efficiently suppressed growth of subcutaneous tumors generated from MEC-1 cells.
Collapse
Affiliation(s)
- Miroslav Boudny
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Jana Zemanova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Prashant Khirsariya
- Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University.,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital
| | - Marek Borsky
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Jan Verner
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Jana Cerna
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Alexandra Oltova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Vaclav Seda
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University.,Center of Molecular Medicine, Central European Institute of Technology, Masaryk University
| | - Marek Mraz
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University.,Center of Molecular Medicine, Central European Institute of Technology, Masaryk University
| | - Josef Jaros
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University
| | - Zuzana Jaskova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Michaela Spunarova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Yvona Brychtova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Karel Soucek
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital.,Department of Cytokinetics, Institute of Biophysics CAS, v.v.i.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Stanislav Drapela
- Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital.,Department of Cytokinetics, Institute of Biophysics CAS, v.v.i.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Marie Kasparkova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| | - Kamil Paruch
- Department of Chemistry, CZ Openscreen, Faculty of Science, Masaryk University .,Center of Biomolecular and Cellular Engineering, International Clinical Research Center, St. Anne's University Hospital
| | - Martin Trbusek
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University
| |
Collapse
|
7
|
Moreno-Villanueva M, Feiveson AH, Krieger S, Kay Brinda A, von Scheven G, Bürkle A, Crucian B, Wu H. Synergistic Effects of Weightlessness, Isoproterenol, and Radiation on DNA Damage Response and Cytokine Production in Immune Cells. Int J Mol Sci 2018; 19:ijms19113689. [PMID: 30469384 PMCID: PMC6275019 DOI: 10.3390/ijms19113689] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/10/2018] [Accepted: 11/11/2018] [Indexed: 12/15/2022] Open
Abstract
The implementation of rotating-wall vessels (RWVs) for studying the effect of lack of gravity has attracted attention, especially in the fields of stem cells, tissue regeneration, and cancer research. Immune cells incubated in RWVs exhibit several features of immunosuppression including impaired leukocyte proliferation, cytokine responses, and antibody production. Interestingly, stress hormones influence cellular immune pathways affected by microgravity, such as cell proliferation, apoptosis, DNA repair, and T cell activation. These pathways are crucial defense mechanisms that protect the cell from toxins, pathogens, and radiation. Despite the importance of the adrenergic receptor in regulating the immune system, the effect of microgravity on the adrenergic system has been poorly studied. Thus, we elected to investigate the synergistic effects of isoproterenol (a sympathomimetic drug), radiation, and microgravity in nonstimulated immune cells. Peripheral blood mononuclear cells were treated with the sympathomimetic drug isoproterenol, exposed to 0.8 or 2 Gy γ-radiation, and incubated in RWVs. Mixed model regression analyses showed significant synergistic effects on the expression of the β2-adrenergic receptor gene (ADRB2). Radiation alone increased ADRB2 expression, and cells incubated in microgravity had more DNA strand breaks than cells incubated in normal gravity. We observed radiation-induced cytokine production only in microgravity. Prior treatment with isoproterenol clearly prevents most of the microgravity-mediated effects. RWVs may be a useful tool to provide insight into novel regulatory pathways, providing benefit not only to astronauts but also to patients suffering from immune disorders or undergoing radiotherapy.
Collapse
Affiliation(s)
- Maria Moreno-Villanueva
- National Aeronautics and Space Administration (NASA), Johnson Space Center Houston, Houston, TX 77058, USA.
- Molecular Toxicology Group, Department of Biology, Box 628, University of Konstanz, 78457 Konstanz, Germany.
| | - Alan H Feiveson
- National Aeronautics and Space Administration (NASA), Johnson Space Center Houston, Houston, TX 77058, USA.
| | | | - AnneMarie Kay Brinda
- Department of Biomedical Engineering, University of Minnesota, 312 Church Street SE, Minneapolis, MN 55455, USA.
| | - Gudrun von Scheven
- Molecular Toxicology Group, Department of Biology, Box 628, University of Konstanz, 78457 Konstanz, Germany.
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, Box 628, University of Konstanz, 78457 Konstanz, Germany.
| | - Brian Crucian
- National Aeronautics and Space Administration (NASA), Johnson Space Center Houston, Houston, TX 77058, USA.
| | - Honglu Wu
- National Aeronautics and Space Administration (NASA), Johnson Space Center Houston, Houston, TX 77058, USA.
| |
Collapse
|
8
|
Thomas A, Redon CE, Sciuto L, Padiernos E, Ji J, Lee MJ, Yuno A, Lee S, Zhang Y, Tran L, Yutzy W, Rajan A, Guha U, Chen H, Hassan R, Alewine CC, Szabo E, Bates SE, Kinders RJ, Steinberg SM, Doroshow JH, Aladjem MI, Trepel JB, Pommier Y. Phase I Study of ATR Inhibitor M6620 in Combination With Topotecan in Patients With Advanced Solid Tumors. J Clin Oncol 2018; 36:1594-1602. [PMID: 29252124 PMCID: PMC5978471 DOI: 10.1200/jco.2017.76.6915] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose Our preclinical work identified depletion of ATR as a top candidate for topoisomerase 1 (TOP1) inhibitor synthetic lethality and showed that ATR inhibition sensitizes tumors to TOP1 inhibitors. We hypothesized that a combination of selective ATR inhibitor M6620 (previously VX-970) and topotecan, a selective TOP1 inhibitor, would be tolerable and active, particularly in tumors with high replicative stress. Patients and Methods This phase I study tested the combination of M6620 and topotecan in 3-week cycles using 3 + 3 dose escalation. The primary end point was the identification of the maximum tolerated dose of the combination. Efficacy and pharmacodynamics were secondary end points. Results Between September 2016 and February 2017, 21 patients enrolled. The combination was well tolerated, which allowed for dose escalation to the highest planned dose level (topotecan 1.25 mg/m2, days 1 to 5; M6620 210 mg/m2, days 2 and 5). One of six patients at this dose level experienced grade 4 thrombocytopenia that required transfusion, a dose-limiting toxicity. Most common treatment-related grade 3 or 4 toxicities were anemia, leukopenia, and neutropenia (19% each); lymphopenia (14%); and thrombocytopenia (10%). Two partial responses (≥ 18 months, ≥ 7 months) and seven stable disease responses ≥ 3 months (median, 9 months; range, 3 to 12 months) were seen. Three of five patients with small-cell lung cancer, all of whom had platinum-refractory disease, had a partial response or prolonged stable disease (10, ≥ 6, and ≥ 7 months). Pharmacodynamic studies showed preliminary evidence of ATR inhibition and enhanced DNA double-stranded breaks in response to the combination. Conclusion To our knowledge, this report is the first of an ATR inhibitor-chemotherapy combination. The maximum dose of topotecan plus M6620 is tolerable. The combination seems particularly active in platinum-refractory small-cell lung cancer, which tends not to respond to topotecan alone. Phase II studies with biomarker evaluation are ongoing.
Collapse
Affiliation(s)
- Anish Thomas
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Christophe E. Redon
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Linda Sciuto
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Emerson Padiernos
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Jiuping Ji
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Min-Jung Lee
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Akira Yuno
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Sunmin Lee
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Yiping Zhang
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Lan Tran
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - William Yutzy
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Arun Rajan
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Udayan Guha
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Haobin Chen
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Raffit Hassan
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Christine C. Alewine
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Eva Szabo
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Susan E. Bates
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Robert J. Kinders
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Seth M. Steinberg
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - James H. Doroshow
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Mirit I. Aladjem
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Jane B. Trepel
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| | - Yves Pommier
- Anish Thomas, Christophe E. Redon, Linda Sciuto, Emerson Padiernos, Min-Jung Lee, Akira Yuno, Sunmin Lee, Arun Rajan, Udayan Guha, Haobin Chen, Raffit Hassan, Christine C. Alewine, Eva Szabo, Seth M. Steinberg, James H. Doroshow, Mirit I. Aladjem, Jane B. Trepel, and Yves Pommier, National Cancer Institute, Bethesda; Jiuping Ji, Yiping Zhang, Lan Tran, William Yutzy, and Robert J. Kinders, Frederick National Laboratory for Cancer Research, Frederick, MD; and Susan E. Bates, Columbia University Medical Center, New York, NY
| |
Collapse
|
9
|
Beyaert M, Starczewska E, Pérez ACG, Vanlangendonck N, Saussoy P, Tilman G, De Leener A, Vekemans MC, Van Den Neste E, Bontemps F. Reevaluation of ATR signaling in primary resting chronic lymphocytic leukemia cells: evidence for pro-survival or pro-apoptotic function. Oncotarget 2017; 8:56906-56920. [PMID: 28915641 PMCID: PMC5593612 DOI: 10.18632/oncotarget.18144] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/24/2017] [Indexed: 12/30/2022] Open
Abstract
ATM, primarily activated by DNA double-strand breaks, and ATR, activated by single-stranded DNA, are master regulators of the cellular response to DNA damage. In primary chronic lymphocytic leukemia (CLL) cells, ATR signaling is considered to be switched off due to ATR downregulation. Here, we hypothesized that ATR, though expressed at low protein level, could play a role in primary resting CLL cells after genotoxic stress. By investigating the response of CLL cells to UV-C irradiation, a prototypical activator of ATR, we could detect phosphorylation of ATR at Thr-1989, a marker for ATR activation, and also observed that selective ATR inhibitors markedly decreased UV-C-induced phosphorylation of ATR targets, including H2AX and p53. Similar results were obtained with the purine analogs fludarabine and cladribine that were also shown to activate ATR and induce ATR-dependent phosphorylation of H2AX and p53. In addition, ATR inhibition was found to sensitize primary CLL cells to UV-C by decreasing DNA repair synthesis. Conversely, ATR inhibition rescued CLL cells against purine analogs by reducing expression of the pro-apoptotic genes PUMA and BAX. Collectively, our study indicates that ATR signaling can be activated in resting CLL cells and play a pro-survival or pro-apoptotic role, depending on the genotoxic context.
Collapse
Affiliation(s)
- Maxime Beyaert
- de Duve Institute, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Eliza Starczewska
- de Duve Institute, Université catholique de Louvain, B-1200 Brussels, Belgium
| | | | - Nicolas Vanlangendonck
- Department of Hematology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Pascale Saussoy
- Service de Biologie clinique, Cliniques universitaires Saint-Luc, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Gaëlle Tilman
- Center for Human Genetic, Cliniques universitaires Saint-Luc, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Anne De Leener
- Center for Human Genetic, Cliniques universitaires Saint-Luc, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Marie-Christiane Vekemans
- Department of Hematology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Eric Van Den Neste
- de Duve Institute, Université catholique de Louvain, B-1200 Brussels, Belgium.,Department of Hematology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Françoise Bontemps
- de Duve Institute, Université catholique de Louvain, B-1200 Brussels, Belgium
| |
Collapse
|
10
|
Petibone DM, Mustafa T, Bourdo SE, Lafont A, Ding W, Karmakar A, Nima ZA, Watanabe F, Casciano D, Morris SM, Dobrovolsky VN, Biris AS. p53
-competent cells and p53
-deficient cells display different susceptibility to oxygen functionalized graphene cytotoxicity and genotoxicity. J Appl Toxicol 2017; 37:1333-1345. [DOI: 10.1002/jat.3472] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Dayton M. Petibone
- Division of Genetic and Molecular Toxicology; National Center for Toxicological Research, FDA; Jefferson AR 72079 USA
| | - Thikra Mustafa
- Center for Integrative Nanotechnology Sciences; University of Arkansas at Little Rock; Little Rock AR 72204 USA
- Department of Medical Bioscience; College of Veterinary Medicine, University of Kirkuk; Kirkuk Iraq
| | - Shawn E. Bourdo
- Center for Integrative Nanotechnology Sciences; University of Arkansas at Little Rock; Little Rock AR 72204 USA
| | - Andersen Lafont
- Nanotechnology Core Facility, Office of Scientific Coordination, National Center for Toxicological Research, FDA; Jefferson AR 72079 USA
| | - Wei Ding
- Division of Genetic and Molecular Toxicology; National Center for Toxicological Research, FDA; Jefferson AR 72079 USA
| | - Alokita Karmakar
- Nanotechnology Core Facility, Office of Scientific Coordination, National Center for Toxicological Research, FDA; Jefferson AR 72079 USA
| | - Zeid A. Nima
- Center for Integrative Nanotechnology Sciences; University of Arkansas at Little Rock; Little Rock AR 72204 USA
| | - Fumiya Watanabe
- Center for Integrative Nanotechnology Sciences; University of Arkansas at Little Rock; Little Rock AR 72204 USA
| | - Daniel Casciano
- Center for Integrative Nanotechnology Sciences; University of Arkansas at Little Rock; Little Rock AR 72204 USA
| | - Suzanne M. Morris
- Division of Genetic and Molecular Toxicology; National Center for Toxicological Research, FDA; Jefferson AR 72079 USA
| | - Vasily N. Dobrovolsky
- Division of Genetic and Molecular Toxicology; National Center for Toxicological Research, FDA; Jefferson AR 72079 USA
| | - Alexandru S. Biris
- Center for Integrative Nanotechnology Sciences; University of Arkansas at Little Rock; Little Rock AR 72204 USA
| |
Collapse
|
11
|
Raskova Kafkova L, Navrkalova V, Jarosova M, Loja T, Chovancova J, Kucerova J, Kriegova E, Prochazka V, Novak Z, Simkova D, Pospisilova S, Divoky V. Ability to downregulate the level of cyclin-dependent kinase inhibitor p27 Kip1 after DNA damage is retained in chronic lymphocytic leukemia cells with functional ATM/p53 signaling pathway. Leuk Lymphoma 2016; 58:199-203. [PMID: 27268868 DOI: 10.1080/10428194.2016.1187276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Leona Raskova Kafkova
- a Department of Biology, Faculty of Medicine and Dentistry , Palacky University , Olomouc , Czech Republic
| | - Veronika Navrkalova
- b Center of Molecular Medicine , CEITEC - Central European Institute of Technology, Masaryk University , Brno , Czech Republic
| | - Marie Jarosova
- c Department of Hemato-Oncology, Faculty of Medicine and Dentistry , Palacky University and University Hospital Olomouc , Czech Republic
| | - Tomas Loja
- b Center of Molecular Medicine , CEITEC - Central European Institute of Technology, Masaryk University , Brno , Czech Republic
| | - Jana Chovancova
- b Center of Molecular Medicine , CEITEC - Central European Institute of Technology, Masaryk University , Brno , Czech Republic
| | - Jana Kucerova
- a Department of Biology, Faculty of Medicine and Dentistry , Palacky University , Olomouc , Czech Republic
| | - Eva Kriegova
- d Department of Immunology, Faculty of Medicine and Dentistry , Palacky University , Olomouc , Czech Republic
| | - Vit Prochazka
- c Department of Hemato-Oncology, Faculty of Medicine and Dentistry , Palacky University and University Hospital Olomouc , Czech Republic
| | - Zdenek Novak
- e Department of Surgery , University of Alabama at Birmingham , Birmingham , AL , USA
| | - Dana Simkova
- a Department of Biology, Faculty of Medicine and Dentistry , Palacky University , Olomouc , Czech Republic
| | - Sarka Pospisilova
- b Center of Molecular Medicine , CEITEC - Central European Institute of Technology, Masaryk University , Brno , Czech Republic
| | - Vladimir Divoky
- a Department of Biology, Faculty of Medicine and Dentistry , Palacky University , Olomouc , Czech Republic
| |
Collapse
|
12
|
Cristini A, Park JH, Capranico G, Legube G, Favre G, Sordet O. DNA-PK triggers histone ubiquitination and signaling in response to DNA double-strand breaks produced during the repair of transcription-blocking topoisomerase I lesions. Nucleic Acids Res 2016; 44:1161-78. [PMID: 26578593 PMCID: PMC4756817 DOI: 10.1093/nar/gkv1196] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 12/12/2022] Open
Abstract
Although defective repair of DNA double-strand breaks (DSBs) leads to neurodegenerative diseases, the processes underlying their production and signaling in non-replicating cells are largely unknown. Stabilized topoisomerase I cleavage complexes (Top1cc) by natural compounds or common DNA alterations are transcription-blocking lesions whose repair depends primarily on Top1 proteolysis and excision by tyrosyl-DNA phosphodiesterase-1 (TDP1). We previously reported that stabilized Top1cc produce transcription-dependent DSBs that activate ATM in neurons. Here, we use camptothecin (CPT)-treated serum-starved quiescent cells to induce transcription-blocking Top1cc and show that those DSBs are generated during Top1cc repair from Top1 peptide-linked DNA single-strand breaks generated after Top1 proteolysis and before excision by TDP1. Following DSB induction, ATM activates DNA-PK whose inhibition suppresses H2AX and H2A ubiquitination and the later assembly of activated ATM into nuclear foci. Inhibition of DNA-PK also reduces Top1 ubiquitination and proteolysis as well as resumption of RNA synthesis suggesting that DSB signaling further enhances Top1cc repair. Finally, we show that co-transcriptional DSBs kill quiescent cells. Together, these new findings reveal that DSB production and signaling by transcription-blocking Top1 lesions impact on non-replicating cell fate and provide insights on the molecular pathogenesis of neurodegenerative diseases such as SCAN1 and AT syndromes, which are caused by TDP1 and ATM deficiency, respectively.
Collapse
Affiliation(s)
- Agnese Cristini
- Cancer Research Center of Toulouse, INSERM UMR1037, Toulouse 31037, France
| | - Joon-Hyung Park
- Cancer Research Center of Toulouse, INSERM UMR1037, Toulouse 31037, France
| | - Giovanni Capranico
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Gaëlle Legube
- Université de Toulouse, UPS, LBCMCP, 31062 Toulouse, France CNRS, LBCMCP, 31062 Toulouse, France
| | - Gilles Favre
- Cancer Research Center of Toulouse, INSERM UMR1037, Toulouse 31037, France
| | - Olivier Sordet
- Cancer Research Center of Toulouse, INSERM UMR1037, Toulouse 31037, France
| |
Collapse
|
13
|
Kwok M, Davies N, Agathanggelou A, Smith E, Oldreive C, Petermann E, Stewart G, Brown J, Lau A, Pratt G, Parry H, Taylor M, Moss P, Hillmen P, Stankovic T. ATR inhibition induces synthetic lethality and overcomes chemoresistance in TP53- or ATM-defective chronic lymphocytic leukemia cells. Blood 2016; 127:582-95. [PMID: 26563132 DOI: 10.1182/blood-2015-05-644872] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 10/31/2015] [Indexed: 12/31/2022] Open
Abstract
TP53 and ataxia telangiectasia mutated (ATM) defects are associated with genomic instability, clonal evolution, and chemoresistance in chronic lymphocytic leukemia (CLL). Currently, therapies capable of providing durable remissions in relapsed/refractory TP53- or ATM-defective CLL are lacking. Ataxia telangiectasia and Rad3-related (ATR) mediates response to replication stress, the absence of which leads to collapse of stalled replication forks into chromatid fragments that require resolution through the ATM/p53 pathway. Here, using AZD6738, a novel ATR kinase inhibitor, we investigated ATR inhibition as a synthetically lethal strategy to target CLL cells with TP53 or ATM defects. Irrespective of TP53 or ATM status, induction of CLL cell proliferation upregulated ATR protein, which then became activated in response to replication stress. In TP53- or ATM-defective CLL cells, inhibition of ATR signaling by AZD6738 led to an accumulation of unrepaired DNA damage, which was carried through into mitosis because of defective cell cycle checkpoints, resulting in cell death by mitotic catastrophe. Consequently, AZD6738 was selectively cytotoxic to both TP53- and ATM-defective CLL cell lines and primary cells. This was confirmed in vivo using primary xenograft models of TP53- or ATM-defective CLL, where treatment with AZD6738 resulted in decreased tumor load and reduction in the proportion of CLL cells with such defects. Moreover, AZD6738 sensitized TP53- or ATM-defective primary CLL cells to chemotherapy and ibrutinib. Our findings suggest that ATR is a promising therapeutic target for TP53- or ATM-defective CLL that warrants clinical investigation.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Animals
- Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors
- Ataxia Telangiectasia Mutated Proteins/genetics
- Ataxia Telangiectasia Mutated Proteins/metabolism
- DNA Damage/drug effects
- Drug Resistance, Neoplasm/drug effects
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Mice, Inbred NOD
- Piperidines
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Pyrazoles/pharmacology
- Pyrazoles/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Marwan Kwok
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom; Centre for Clinical Haematology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Nicholas Davies
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Edward Smith
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Ceri Oldreive
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Eva Petermann
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Grant Stewart
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jeff Brown
- Oncology iMed, AstraZeneca Pharmaceuticals, Waltham, MA
| | - Alan Lau
- R&D Oncology iMed, AstraZeneca Pharmaceuticals, Alderley Park, United Kingdom
| | - Guy Pratt
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom; Birmingham Heartlands Hospital, Birmingham, United Kingdom; and
| | - Helen Parry
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom; Centre for Clinical Haematology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Malcolm Taylor
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Moss
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom; Centre for Clinical Haematology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Peter Hillmen
- Section of Experimental Haematology, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Tatjana Stankovic
- School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom; Centre for Clinical Haematology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| |
Collapse
|
14
|
Bakkenist CJ, Czambel RK, Hershberger PA, Tawbi H, Beumer JH, Schmitz JC. A quasi-quantitative dual multiplexed immunoblot method to simultaneously analyze ATM and H2AX Phosphorylation in human peripheral blood mononuclear cells. Oncoscience 2015; 2:542-54. [PMID: 26097887 PMCID: PMC4468340 DOI: 10.18632/oncoscience.162] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/05/2015] [Indexed: 12/12/2022] Open
Abstract
Pharmacologic inhibition of DNA repair may increase the efficacy of many cytotoxic cancer agents. Inhibitors of DNA repair enzymes including APE1, ATM, ATR, DNA-PK and PARP have been developed and the PARP inhibitor olaparib is the first-in-class approved in Europe and the USA for the treatment of advanced BRCA-mutated ovarian cancer. Sensitive pharmacodynamic (PD) biomarkers are needed to further evaluate the efficacy of inhibitors of DNA repair enzymes in clinical trials. ATM is a protein kinase that mediates cell-cycle checkpoint activation and DNA double-strand break repair. ATM kinase activation at DNA double-strand breaks (DSBs) is associated with intermolecular autophosphorylation on serine-1981. Exquisite sensitivity and high stoichiometry as well as facile extraction suggest that ATM serine-1981 phosphorylation may be a highly dynamic PD biomarker for both ATM kinase inhibitors and radiation- and chemotherapy-induced DSBs. Here we report the pre-clinical analytical validation and fit-for-purpose biomarker method validation of a quasi-quantitative dual multiplexed immunoblot method to simultaneously analyze ATM and H2AX phosphorylation in human peripheral blood mononuclear cells (PBMCs). We explore the dynamics of these phosphorylations in PBMCs exposed to chemotherapeutic agents and DNA repair inhibitors in vitro, and show that ATM serine-1981 phosphorylation is increased in PBMCs in sarcoma patients treated with DNA damaging chemotherapy.
Collapse
Affiliation(s)
| | - R Kenneth Czambel
- Medicine, University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA 15213-1863
| | - Pamela A Hershberger
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263
| | - Hussein Tawbi
- Medicine, University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA 15213-1863
| | - Jan H Beumer
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, 5117 Centre Avenue, Pittsburgh, PA 15213-1863
| | - John C Schmitz
- Medicine, University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA 15213-1863
| |
Collapse
|
15
|
Chen T, Middleton FK, Falcon S, Reaper PM, Pollard JR, Curtin NJ. Development of pharmacodynamic biomarkers for ATR inhibitors. Mol Oncol 2014; 9:463-72. [PMID: 25459351 DOI: 10.1016/j.molonc.2014.09.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/29/2014] [Accepted: 09/30/2014] [Indexed: 10/24/2022] Open
Abstract
BACKGROUND ATR, which signals DNA damage to S/G2 cell cycle checkpoints and for repair, is an attractive target in cancer therapy. ATR inhibitors are being developed and a pharmacodynamic assay is needed to support clinical studies. METHODS Phosphorylation of ATR targets, Chk1 and H2AX, was evaluated in MCF7 and K562 cells, human volunteer PBMCs and whole blood by Western blot, immunofluorescence microscopy and flow cytometry after DNA damage. The effect of cell cycle phase, ATR knockdown and inhibition on these phosphorylation events was determined. RESULTS Hydroxyurea, UV and 4NQO induced Chk1 and H2AX phosphorylation in MCF7 and K562 cells. UV/4NQO activation of ATR was detectable in non-cycling cells. Chk1 phosphorylation was reduced by ATR knockdown and reflects ATR activity for 3 h, H2AX phosphorylation after UV/4NQO is ATR-dependent for 1 h but increasingly ATM and DNA-PK-dependent at later time points. In isolated PBMCs both phospho-targets were detectable after UV/4NQO but in PBMCs from whole blood treated with 4NQO only H2AX was detectable. CONCLUSION PhosphoChk1 and H2AX are useful biomarkers for ATR inhibition using a variety of immuno-detection methods, but timing may be critical. Importantly, ATR activity is detectable in non-cycling PBMCs allowing them to be used as a surrogate tissue for biomarker measurement. In PBMCs from whole blood treated with 4NQO phosphoH2AX was the most useful biomarker of ATR activity and a clinically viable pharmacodynamic assay for ATR inhibitors has been developed.
Collapse
Affiliation(s)
- Tao Chen
- Newcastle University, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle upon Tyne, NE2 4HH, UK
| | - Fiona K Middleton
- Newcastle University, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle upon Tyne, NE2 4HH, UK
| | - Susanna Falcon
- Vertex Pharmaceuticals (Europe) Limited, 86-88 Jubilee Avenue, Milton Park, Abingdon, Oxfordshire, OX14 4RW, UK
| | - Philip M Reaper
- Vertex Pharmaceuticals (Europe) Limited, 86-88 Jubilee Avenue, Milton Park, Abingdon, Oxfordshire, OX14 4RW, UK
| | - John R Pollard
- Vertex Pharmaceuticals (Europe) Limited, 86-88 Jubilee Avenue, Milton Park, Abingdon, Oxfordshire, OX14 4RW, UK
| | - Nicola J Curtin
- Newcastle University, Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
16
|
Lin K, Farahani M, Yang Y, Johnson GG, Oates M, Atherton M, Douglas A, Kalakonda N, Pettitt AR. Loss of MIR15A and MIR16-1 at 13q14 is associated with increased TP53 mRNA, de-repression of BCL2 and adverse outcome in chronic lymphocytic leukaemia. Br J Haematol 2014; 167:346-55. [PMID: 25040181 DOI: 10.1111/bjh.13043] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/16/2014] [Indexed: 12/29/2022]
Abstract
This study was conducted to investigate the possibility that TP53 mRNA is variably expressed in chronic lymphocytic leukaemia (CLL) and that under-expression is associated with TP53 dysfunction and adverse outcome. Although TP53 mRNA levels did indeed vary among the 104 CLL samples examined, this variability resulted primarily from over-expression of TP53 mRNA in 18 samples, all of which lacked TP53 deletion/mutation. These patients had higher lymphocyte counts and shorter overall and treatment-free survival times compared to cases with low TP53 mRNA expression and no TP53 deletion/mutation. Furthermore, TP53 mRNA levels did not correlate with levels of TP53 protein or its transcriptional target CDKN1A. We speculated that the adverse outcome associated with TP53 mRNA over-expression might reflect variation in levels of MIR15A and MIR16-1, which are encoded on chromosome 13q14 and target TP53 and some oncogenes including BCL2. In keeping with our hypothesis, 13q14 copy number and levels of MIR15A/MIR16-1 correlated positively with one another but negatively with levels of TP53 mRNA and BCL2 mRNA. Our findings support a model in which loss of MIR15A/MIR16-1 at chromosome 13q14 results in adverse outcome due to de-repression of oncogenes such as BCL2, and up-regulation of TP53 mRNA as a bystander effect.
Collapse
Affiliation(s)
- Ke Lin
- Department of Haematology, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK; Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Bakkenist CJ, Czambel RK, Clump DA, Greenberger JS, Beumer JH, Schmitz JC. Radiation therapy induces the DNA damage response in peripheral blood. Oncotarget 2014; 4:1143-8. [PMID: 23900392 PMCID: PMC3787146 DOI: 10.18632/oncotarget.1084] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Stereotactic body radiation therapy (SBRT) is a radiotherapy modality that delivers highly conformal, ablative doses to a well-defined target. Here, using a semiquantitative multiplexed assay to analyze ATM and H2AX phosphorylation, we show that ATM kinase activity in peripheral blood mononuclear cells is induced following SBRT. This observation of a systemic ATM kinase-dependent DNA damage response in the peripheral blood is unprecedented and promotes the use of ATM serine-1981 phosphorylation as a predictive biomarker for DNA damaging modalities and ATM inhibitors.
Collapse
Affiliation(s)
- Christopher J Bakkenist
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
18
|
Poly(ADP-ribose) polymerase inhibitor CEP-8983 synergizes with bendamustine in chronic lymphocytic leukemia cells in vitro. Leuk Res 2013; 38:411-7. [PMID: 24439051 DOI: 10.1016/j.leukres.2013.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/16/2013] [Accepted: 12/18/2013] [Indexed: 12/19/2022]
Abstract
DNA repair aberrations and associated chromosomal instability is a feature of chronic lymphocytic leukemia (CLL). To evaluate if DNA repair insufficiencies are related to methylation changes, we examined the methylation of nine promoter regions of DNA repair proteins by bisulfide sequencing in 26 CLL primary samples and performed quantitative PCR on a subset of samples to examine BRCA1 expression. We also investigated if changes in cytogenetic or expression level of DNA repair proteins led to changes in sensitivity to a novel PARP inhibitor, CEP-8983, alone and in combination with bendamustine. No changes in promoter methylation were identified in BRCA1, BRCA2, FANC-C, FANC-F, FANC-L, ATM, MGMT, hMLH1 and H2AX except for two cases of minor BRCA1 hypermethylation. CLL samples appeared to have reduced BRCA1 mRNA expression uniformly in comparison to non-malignant lymphocytes irrespective of promoter hypermethylation. CEP-8983 displayed single agent cytotoxicity and the combination with bendamustine demonstrated synergistic cytotoxicity in the majority of CLL samples. These results were consistent across cytogenetic subgroups, including 17p deleted and previously treated patients. Our results provide rationale for further exploration of the combination of a PARP inhibitor and DNA damaging agents as a novel therapeutic strategy in CLL.
Collapse
|
19
|
Navrkalova V, Sebejova L, Zemanova J, Jaskova Z, Trbusek M. The p53 pathway induction is not primarily dependent on Ataxia Telangiectasia Mutated (ATM) gene activity after fludarabine treatment in chronic lymphocytic leukemia cells. Leuk Lymphoma 2013; 54:1840-3. [PMID: 23808769 DOI: 10.3109/10428194.2013.796056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The prognostic role of ATM defects is well documented in chronic lymphocytic leukemia. However, the predictive value of ATM inactivation is much less understood, even in response to common drugs like fludarabine. It has been demonstrated that CLL cells having inactive ATM exhibit defective phosphorylation of its downstream targets after fludarabine treatment. We performed alternative analysis focusing on fludarabine-induced p53 accumulation and induction of p53-downstream genes after artificial ATM inhibition and, in parallel, using cells with endogenous ATM inactivation. We show that after 24h fludarabine exposure: (i) 5 out of 8 ATM-deficient samples (63%) normally accumulated p53 protein, and (ii) all analyzed ATM-deficient samples (n = 7) manifested clear induction of p21, PUMA, BAX, and GADD45 genes. In all experiments, doxorubicin was used as a confined ATM inductor and confirmed effective ATM inactivation. In conclusion, CLL cells lacking functional ATM appear to have normal response to fludarabine regarding the p53 pathway activation.
Collapse
Affiliation(s)
- Veronika Navrkalova
- Department of Molecular Medicine, CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | | | | | | | | |
Collapse
|
20
|
de Viron E, Michaux L, Put N, Bontemps F, Van Den Neste E. Present status and perspectives in functional analysis of p53 in chronic lymphocytic leukemia. Leuk Lymphoma 2012; 53:1445-51. [PMID: 22280536 DOI: 10.3109/10428194.2012.660630] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Aberrations of TP53 (mutations and/or deletions) are associated with a dismal prognosis in chronic lymphocytic leukemia (CLL). Complete loss of ATM is another mechanism of failed DNA damage response and also associated with poorer prognosis in CLL. However, p53 dysfunction may arise through alternative mechanisms unrelated to structural aberrations (deletion and/or mutation) of TP53 or ATM, and thus be undetectable by traditional DNA-directed approaches (fluorescence in situ hybridization [FISH], sequencing, karyotyping). In order to address the latter changes, and also to better understand the consequences of TP53/ATM aberrations, p53 functional assays have recently been developed. The purpose of dynamic assessment of p53 response in CLL is to carry out a comprehensive analysis of all mechanisms causing p53-deficient phenotype, including those unrelated to genomic aberrations of TP53 and ATM. The present review focuses on the current knowledge of p53 function assays in CLL, including important features such as technical issues, correlation with structural aberrations and clinical value.
Collapse
Affiliation(s)
- Emeline de Viron
- De Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | | | | | | |
Collapse
|
21
|
Genetic diversity and population structure of the endangered marsupial Sarcophilus harrisii (Tasmanian devil). Proc Natl Acad Sci U S A 2011; 108:12348-53. [PMID: 21709235 DOI: 10.1073/pnas.1102838108] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The Tasmanian devil (Sarcophilus harrisii) is threatened with extinction because of a contagious cancer known as Devil Facial Tumor Disease. The inability to mount an immune response and to reject these tumors might be caused by a lack of genetic diversity within a dwindling population. Here we report a whole-genome analysis of two animals originating from extreme northwest and southeast Tasmania, the maximal geographic spread, together with the genome from a tumor taken from one of them. A 3.3-Gb de novo assembly of the sequence data from two complementary next-generation sequencing platforms was used to identify 1 million polymorphic genomic positions, roughly one-quarter of the number observed between two genetically distant human genomes. Analysis of 14 complete mitochondrial genomes from current and museum specimens, as well as mitochondrial and nuclear SNP markers in 175 animals, suggests that the observed low genetic diversity in today's population preceded the Devil Facial Tumor Disease disease outbreak by at least 100 y. Using a genetically characterized breeding stock based on the genome sequence will enable preservation of the extant genetic diversity in future Tasmanian devil populations.
Collapse
|
22
|
Warmerdam DO, Kanaar R, Smits VAJ. Differential Dynamics of ATR-Mediated Checkpoint Regulators. J Nucleic Acids 2010; 2010. [PMID: 20847938 PMCID: PMC2933903 DOI: 10.4061/2010/319142] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 06/28/2010] [Indexed: 12/18/2022] Open
Abstract
The ATR-Chk1 checkpoint pathway is activated by UV-induced DNA lesions and replication stress. Little was known about the spatio and temporal behaviour of the proteins involved, and we, therefore, examined the behaviour of the ATRIP-ATR and Rad9-Rad1-Hus1 putative DNA damage sensor complexes and the downstream effector kinase Chk1. We developed assays for the generation and validation of stable cell lines expressing GFP-fusion proteins. Photobleaching experiments in living cells expressing these fusions indicated that after UV-induced DNA damage, ATRIP associates more transiently with damaged chromatin than members of the Rad9-Rad1-Hus1 complex. Interestingly, ATRIP directly associated with locally induced UV damage, whereas Rad9 bound in a cooperative manner, which can be explained by the Rad17-dependent loading of Rad9 onto damaged chromatin. Although Chk1 dissociates from the chromatin upon UV damage, no change in the mobility of GFP-Chk1 was observed, supporting the notion that Chk1 is a highly dynamic protein.
Collapse
Affiliation(s)
- Daniël O Warmerdam
- Department of Cell Biology and Genetics, Cancer Genome Center, Erasmus MC, Dr. Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | | | |
Collapse
|
23
|
Degerman S, Siwicki JK, Osterman P, Lafferty-Whyte K, Keith WN, Roos G. Telomerase upregulation is a postcrisis event during senescence bypass and immortalization of two Nijmegen breakage syndrome T cell cultures. Aging Cell 2010; 9:220-35. [PMID: 20089118 DOI: 10.1111/j.1474-9726.2010.00550.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Our knowledge on immortalization and telomere biology is mainly based on genetically manipulated cells analyzed before and many population doublings post growth crisis. The general view is that growth crisis is telomere length (TL) dependent and that escape from crisis is coupled to increased expression of the telomerase reverse transcriptase (hTERT) gene, telomerase activity upregulation and TL stabilization. Here we have analyzed the process of spontaneous immortalization of human T cells, regarding pathways involved in senescence and telomerase regulation. Two Nijmegen breakage syndrome (NBS) T cell cultures (S3R and S4) showed gradual telomere attrition until a period of growth crisis followed by the outgrowth of immortalized cells. Whole genome expression analysis indicated differences between pre-, early post- and late postcrisis cells. Early postcrisis cells demonstrated a logarithmic growth curve, very short telomeres and, notably, no increase in hTERT or telomerase activity despite downregulation of several negative hTERT regulators (e.g. FOS, JUN D, SMAD3, RUNX2, TNF-a and TGFb-R2). Thereafter, cMYC mRNA increased in parallel with increased hTERT expression, telomerase activity and elongation of short telomeres, indicating a step-wise activation of hTERT transcription involving reduction of negative regulators followed by activation of positive regulator(s). Gene expression analysis indicated that cells escaped growth crisis by deregulated DNA damage response and senescence controlling genes, including downregulation of ATM, CDKN1B (p27), CDKN2D (p19) and ASF1A and upregulation of CDK4, TWIST1, TP73L (p63) and SYK. Telomerase upregulation was thus found to be uncoupled to escape of growth crisis but rather a later event in the immortalization process of NBS T cell cultures.
Collapse
Affiliation(s)
- Sofie Degerman
- Department of Medical Biosciences, Pathology, Umeå University, SE-90185 Umeå, Sweden
| | | | | | | | | | | |
Collapse
|
24
|
Ouillette P, Fossum S, Parkin B, Ding L, Bockenstedt P, Al-Zoubi A, Shedden K, Malek SN. Aggressive chronic lymphocytic leukemia with elevated genomic complexity is associated with multiple gene defects in the response to DNA double-strand breaks. Clin Cancer Res 2010; 16:835-47. [PMID: 20086003 DOI: 10.1158/1078-0432.ccr-09-2534] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Genomic complexity is present in approximately 15% to 30% of all chronic lymphocytic leukemia (CLL) and has emerged as a strong independent predictor of rapid disease progression and short remission duration in CLL. We conducted this study to advance our understanding of the causes of genomic complexity in CLL. EXPERIMENTAL DESIGN We have obtained quantitative measurements of radiation-induced apoptosis and radiation-induced ATM autophosphorylation in purified CLL cells from 158 and 140 patients, respectively, and have used multivariate analysis to identify independent contributions of various biological variables on genomic complexity in CLL. RESULTS Here, we identify a strong independent effect of radiation resistance on elevated genomic complexity in CLL and describe radiation resistance as a predictor for shortened CLL survival. Furthermore, using multivariate analysis, we identify del17p/p53 aberrations, del11q, del13q14 type II (invariably resulting in Rb loss), and CD38 expression as independent predictors of genomic complexity in CLL, with aberrant p53 as a predictor of approximately 50% of genomic complexity in CLL. Focusing on del11q, we determined that normalized ATM activity was a modest predictor of genomic complexity but was not independent of del11q. Through single nucleotide polymorphism array-based fine mapping of del11q, we identified frequent monoallelic loss of Mre11 and H2AFX in addition to ATM, indicative of compound del11q-resident gene defects in the DNA double-strand break response. CONCLUSIONS Our quantitative analysis links multiple molecular defects, including for the first time del11q and large 13q14 deletions (type II), to elevated genomic complexity in CLL, thereby suggesting mechanisms for the observed clinical aggressiveness of CLL in patients with unstable genomes.
Collapse
Affiliation(s)
- Peter Ouillette
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Sordet O, Nakamura AJ, Redon CE, Pommier Y. DNA double-strand breaks and ATM activation by transcription-blocking DNA lesions. Cell Cycle 2010; 9:274-8. [PMID: 20023421 DOI: 10.4161/cc.9.2.10506] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
A taxia telangiectasia mutated (ATM), the deficiency of which causes a severe neurodegenerative disease, is a crucial mediator for the DNA double-strand break (DSB) response. We recently showed that transcription-blocking topoisomerase I cleavage complexes (TOP1cc) produce DSBs related to R-loop formation and activate ATM in post-mitotic neurons and lymphocytes. Here we discuss how TOP1cc can produce transcription arrest with R-loop formation and generate DSBs that activate ATM, as well as data suggesting that those transcription-dependent DSBs tend to form at the IgH locus and at specific genomic sites. We also address the potential roles of ATM in response to transcription-blocking TOP1cc.
Collapse
Affiliation(s)
- Olivier Sordet
- Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
26
|
de Viron E, Knoops L, Connerotte T, Smal C, Michaux L, Saussoy P, Vannuffel P, Beert E, Vekemans MC, Hermans C, Bontemps F, Van Den Neste E. Impaired up-regulation of polo-like kinase 2 in B-cell chronic lymphocytic leukaemia lymphocytes resistant to fludarabine and 2-chlorodeoxyadenosine: a potential marker of defective damage response. Br J Haematol 2009; 147:641-52. [PMID: 19764992 DOI: 10.1111/j.1365-2141.2009.07900.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The functional evaluation of ataxia telangiectasia mutated (ATM) and p53 was recently developed in B-cell chronic lymphocytic leukaemia (B-CLL), a disease in which the response to DNA damage is frequently altered. We identified a novel biomarker of chemosensitivity based on the induction of DNA damage by the purine nucleoside analogues (PNA) fludarabine and 2-chlorodeoxyadenosine (CdA). Using genome-wide expression profiling, it was observed that, in chemosensitive samples, PNA predominantly increased the expression of p53-dependent genes, among which PLK2 was the most highly activated at early time points. Conversely, in chemoresistant samples, p53-dependent and PLK2 responses were abolished. Using a quantitative real time polymerase chain reaction, we confirmed that PNA dose- and time-dependently increased PLK2 expression in chemosensitive but not chemoresistant B-CLL samples. Analysis of a larger cohort of B-CLL patients showed that cytotoxicity induced by PNA correlated well with PLK2 mRNA induction. Interestingly, we observed that failure to up-regulate PLK2 following PNA and chemoresistance were not strictly correlated with structural alterations in the TP53 gene. In conclusion, we propose that testing PLK2 activation after a 24-h incubation with PNA could be used to investigate the functional integrity of DNA damage-response pathways in B-CLL cells, and predict clinical sensitivity to these drugs.
Collapse
Affiliation(s)
- Emeline de Viron
- De Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ataxia telangiectasia mutated activation by transcription- and topoisomerase I-induced DNA double-strand breaks. EMBO Rep 2009; 10:887-93. [PMID: 19557000 DOI: 10.1038/embor.2009.97] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Revised: 03/30/2009] [Accepted: 04/09/2009] [Indexed: 12/22/2022] Open
Abstract
Ataxia telangiectasia mutated (ATM), the deficiency of which causes a severe neurodegenerative disease, is a crucial mediator for the DNA damage response (DDR). As neurons have high rates of transcription that require topoisomerase I (TOP1), we investigated whether TOP1 cleavage complexes (TOP1cc)-which are potent transcription-blocking lesions-also produce transcription-dependent DNA double-strand breaks (DSBs) with ATM activation. We show the induction of DSBs and DDR activation in post-mitotic primary neurons and lymphocytes treated with camptothecin, with the induction of nuclear DDR foci containing activated ATM, gamma-H2AX (phosphorylated histone H2AX), activated CHK2 (checkpoint kinase 2), MDC1 (mediator of DNA damage checkpoint 1) and 53BP1 (p53 binding protein 1). The DSB-ATM-DDR pathway was suppressed by inhibiting transcription and gamma-H2AX signals were reduced by RNase H1 transfection, which removes transcription-mediated R-loops. Thus, we propose that Top1cc produce transcription arrests with R-loop formation and generate DSBs that activate ATM in post-mitotic cells.
Collapse
|
28
|
Tsai CY, Ray AS, Tumas DB, Keating MJ, Reiser H, Plunkett W. Targeting DNA Repair in Chronic Lymphocytic Leukemia Cells with a Novel Acyclic Nucleotide Analogue, GS-9219. Clin Cancer Res 2009; 15:3760-9. [DOI: 10.1158/1078-0432.ccr-08-2848] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Porcedda P, Turinetto V, Brusco A, Cavalieri S, Lantelme E, Orlando L, Ricardi U, Amoroso A, Gregori D, Giachino C. A rapid flow cytometry test based on histone H2AX phosphorylation for the sensitive and specific diagnosis of ataxia telangiectasia. Cytometry A 2008; 73:508-16. [PMID: 18431795 DOI: 10.1002/cyto.a.20566] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ataxia telangiectasia (A-T) is a progressive neurodegenerative disease with onset in early childhood, caused by mutations in the ATM (ataxia-telangiectasia mutated) gene. Diagnosis relies on laboratory tests showing high levels of serum alphafetoprotein, cell sensitivity to ionizing radiation (IR) and absence or reduced levels of ATM protein. Many tests, however, are not sufficiently sensitive or specific for A-T, have long turnaround times, or require large blood samples. This prompted us to develop a new flow cytometry method for the diagnosis of A-T based on the measurement of histone H2AX phosphorylation. We established normal ranges of histone H2AX phosphorylation after 2 Gy IR by testing T-cell lines, lymphoblastoid cell lines (LCLs) and/or peripheral blood mononuclear cells (PBMCs) or both from 20 genetically proven A-T and 46 control donors. To further evaluate the specificity and sensitivity of the test, we analyzed cells from 19 patients suspected of having A-T, and from one Friedreich Ataxia, one Ataxia with Oculomotor Apraxia type 2, and one Nijmegen Breakage Syndrome patients. Phosphorylated histone H2AX mean fluorescence intensity of irradiated A-T cells was significantly lower than that of healthy donors. The intrastaining, intraassay, and interassay imprecisions were <or=13.22%. Sensitivity and specificity were virtually 100% when the test was performed on PBMCs. Screening of 19 consecutive new patients with suspected A-T classified 15 patients as non-A-T and four as A-T; diagnosis of the latter four was subsequently confirmed by DNA sequencing to identify ATM mutations. The Friedreich Ataxia patient, the Ataxia with Oculomotor Apraxia type 2 patient and the Nijmegen Breakage Syndrome patient were classified as non-A-T. This flow cytometry test is very sensitive, specific and rapid, and requires only 2 ml of blood. It may thus be proposed for the early differential diagnosis of A-T as an alternative to methods requiring the production of LCLs.
Collapse
Affiliation(s)
- Paola Porcedda
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Entezam A, Usdin K. ATR protects the genome against CGG.CCG-repeat expansion in Fragile X premutation mice. Nucleic Acids Res 2007; 36:1050-6. [PMID: 18160412 PMCID: PMC2241920 DOI: 10.1093/nar/gkm1136] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fragile X mental retardation syndrome is a repeat expansion disease caused by expansion of a CGG.CCG-repeat tract in the 5' UTR of the FMR1 gene. In humans, small expansions occur more frequently on paternal transmission while large expansions are exclusively maternal in origin. It has been suggested that expansion is the result of aberrant DNA replication, repair or recombination. To distinguish amongst these possibilities we crossed mice containing 120 CGG.CCG-repeats in the 5' UTR of the mouse Fmr1 gene to mice with mutations in ATR, a protein important in the cellular response to stalled replication forks and bulky DNA lesions. We show here that ATR heterozygosity results in increased expansion rates of maternally, but not paternally, transmitted alleles. In addition, age-related somatic expansions occurred in mice of both genders that were not seen in ATR wild-type animals. Some ATR-sensitive expansion occurs in postmitotic cells including haploid gametes suggesting that aberrant DNA repair is responsible. Our data suggest that two mechanisms of repeat expansion exist that may explain the small and large expansions seen in humans. In addition, our data provide an explanation for the maternal bias of large expansions in humans and the lower incidence of these expansions in mice.
Collapse
Affiliation(s)
- Ali Entezam
- Section on Gene Structure and Disease, Laboratory of Molecular and Cellular Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0830, USA
| | | |
Collapse
|
31
|
Cotter FE, Auer RL. Genetic alteration associated with chronic lymphocytic leukemia. Cytogenet Genome Res 2007; 118:310-9. [PMID: 18000385 DOI: 10.1159/000108315] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2007] [Accepted: 03/14/2007] [Indexed: 12/19/2022] Open
Abstract
The genetics of B-cell chronic lymphocytic leukemia (B-CLL) differ considerably from most other forms of hematologic malignancy which are usually characterized by chromosome translocations. B-CLL typically contains chromosomal deletions and chromosomes 13q14 and 11q22-->q23 are the most common. These two regions appear to share a common ancestral origin (Auer et al., 2007b). Overall, chromosomal abnormalities can be found in the majority of patients with B-CLL when using sensitive techniques (Dohneret al., 2000) and possibly reflects an underlying predisposition, with a small but significant number of familial cases. Although single and consistent abnormalities are most common, multiple rearrangements can occur, often with disease progression (Feganetal., 1995; Dohner et al., 2000). Regions of recurrent deletion suggest the presence of tumor suppressor genes if following Knudson's theoretical 2-hit model. However, despite extensive sequencing analysis over the last decade and lack of pathogenic mutations identified, there has been a move away from this suggested hypothesis and alternative mechanisms of gene inactivation involving epigenetic silencing or haploinsufficiency may be considered as more likely in this disease. This review focuses on the common genetic abnormalities in B-CLL and relates them to some of the more recent hypotheses on inactivation of genes within these regions of deletion.
Collapse
Affiliation(s)
- F E Cotter
- Centre for Haematology, Institute of Cell and Molecular Sciences, Barts and the London Queen Mary School of Medicine, London, UK.
| | | |
Collapse
|
32
|
Hsp90 inhibition has opposing effects on wild-type and mutant p53 and induces p21 expression and cytotoxicity irrespective of p53/ATM status in chronic lymphocytic leukaemia cells. Oncogene 2007; 27:2445-55. [PMID: 17982489 DOI: 10.1038/sj.onc.1210893] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In chronic lymphocytic leukaemia (CLL), mutation/deletion of TP53 is strongly associated with early disease progression, resistance to chemotherapy and short patient survival. Consequently, there is a pressing need to develop novel treatment protocols for this high-risk patient group. The present study was performed to evaluate Hsp90 inhibition as a possible therapeutic approach for such patients. Primary CLL cells of defined ataxia telangiectasia mutated (ATM)/p53 status were incubated with the Hsp90 inhibitor geldanamycin (GA) and analysed by western blotting for the expression of p53, p21, MDM2 and Akt. GA downregulated overexpressed mutant p53 protein (an oncogene) and upregulated wild-type (wt) p53 (a tumour suppressor). The upregulation of wt p53 by GA was independent of ATM and was accompanied by downregulation of Akt and the active form of MDM2, indicating a possible mechanism. GA also produced a p53/ATM-independent increase in the levels of p21-a potent inducer of cell-cycle arrest. In-vitro cytotoxicity studies showed that GA killed cultured CLL cells in a dose- and time-dependent fashion irrespective of their p53/ATM status and more effectively than normal blood mononuclear cells. In summary, our findings reveal important consequences of inhibiting Hsp90 in CLL cells and strongly support the therapeutic evaluation of Hsp90 inhibitors in poor-prognosis patients with p53 defects.
Collapse
|
33
|
Austen B, Skowronska A, Baker C, Powell JE, Gardiner A, Oscier D, Majid A, Dyer M, Siebert R, Taylor AM, Moss PA, Stankovic T. Mutation status of the residual ATM allele is an important determinant of the cellular response to chemotherapy and survival in patients with chronic lymphocytic leukemia containing an 11q deletion. J Clin Oncol 2007; 25:5448-57. [PMID: 17968022 DOI: 10.1200/jco.2007.11.2649] [Citation(s) in RCA: 180] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE The ataxia telangiectasia mutated (ATM) gene is located on chromosome 11q and loss of this region is common in B-cell chronic lymphocytic leukemia (CLL). Our aim was to determine if CLL tumors with a chromosome 11q deletion might be divided into two subgroups based on the status of the remaining ATM allele. METHODS The sequence of the residual ATM allele was determined in 72 CLLs with an 11q deletion. This was related to the cellular response to irradiation or cytotoxic drug exposure in vitro and clinical outcome. RESULTS We show that the residual ATM allele is mutated in 36% of CLLs with an 11q deletion and that these leukemias demonstrate an impaired cellular response to irradiation or cytotoxic drug exposure in vitro. Inactivation of the second ATM allele was associated with a reduction in patient survival beyond that already dictated by the presence of an 11q deletion (P = .0283). Furthermore, we demonstrate that ATM mutations may arise during the evolution of an 11q deleted subclone and are associated with its expansion. CONCLUSION CLL with 11q deletion can be divided into two subgroups based on the integrity of the residual ATM allele. Patients with complete loss of ATM function, due to biallelic ATM defects, have defective responses to cytotoxic chemotherapeutics in vitro and a poorer clinical outcome. ATM mutant subclones can develop during an individual's disease course and give rise to additional expansion of the 11q deleted subclone.
Collapse
Affiliation(s)
- Belinda Austen
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Zimmerman ES, Sherman MP, Blackett JL, Neidleman JA, Kreis C, Mundt P, Williams SA, Warmerdam M, Kahn J, Hecht FM, Grant RM, de Noronha CMC, Weyrich AS, Greene WC, Planelles V. Human immunodeficiency virus type 1 Vpr induces DNA replication stress in vitro and in vivo. J Virol 2006; 80:10407-18. [PMID: 16956949 PMCID: PMC1641771 DOI: 10.1128/jvi.01212-06] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) viral protein R (Vpr) causes cell cycle arrest in G2. Vpr-expressing cells display the hallmarks of certain forms of DNA damage, specifically activation of the ataxia telangiectasia mutated and Rad3-related kinase, ATR. However, evidence that Vpr function is relevant in vivo or in the context of viral infection is still lacking. In the present study, we demonstrate that HIV-1 infection of primary, human CD4+ lymphocytes causes G2 arrest in a Vpr-dependent manner and that this response requires ATR, as shown by RNA interference. The event leading to ATR activation in CD4+ lymphocytes is the accumulation of replication protein A in nuclear foci, an indication that Vpr likely induces stalling of replication forks. Primary macrophages are refractory to ATR activation by Vpr, a finding that is consistent with the lack of detectable ATR, Rad17, and Chk1 protein expression in these nondividing cells. These observations begin to explain the remarkable resilience of macrophages to HIV-1-induced cytopathicity. To study the in vivo consequences of Vpr function, we isolated CD4+ lymphocytes from HIV-1-infected individuals and interrogated the cell cycle status of anti-p24Gag-immunoreactive cells. We report that infected cells in vivo display an aberrant cell cycle profile whereby a majority of cells have a 4N DNA content, consistent with the onset of G2 arrest.
Collapse
Affiliation(s)
- Erik S Zimmerman
- Division of Cellular Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Porcedda P, Turinetto V, Lantelme E, Fontanella E, Chrzanowska K, Ragona R, De Marchi M, Delia D, Giachino C. Impaired elimination of DNA double-strand break-containing lymphocytes in ataxia telangiectasia and Nijmegen breakage syndrome. DNA Repair (Amst) 2006; 5:904-13. [PMID: 16765653 DOI: 10.1016/j.dnarep.2006.05.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Accepted: 05/09/2006] [Indexed: 01/10/2023]
Abstract
The repair of DNA double-strand breaks is critical for genome integrity and tumor suppression. Here we show that following treatment with the DNA-intercalating agent actinomycin D (ActD), normal quiescent T cells accumulate double-strand breaks and die, whereas T cells from ataxia telangiectasia (AT) and Nijmegen breakage syndrome (NBS) patients are resistant to this death pathway despite a comparable amount of DNA damage. We demonstrate that the ActD-induced death pathway in quiescent T lymphocytes follows DNA damage and H2AX phosphorylation, is ATM- and NBS1-dependent and due to p53-mediated cellular apoptosis. In response to genotoxic 2-Gy gamma-irradiation, on the other hand, quiescent T cells from normal donors survive following complete resolution of the damage thus induced. T cells from AT and NBS patients also survive, but retain foci of phosphorylated H2AX due to a subtle double-strand break (DSB) repair defect. A common consequence of these two genetic defects in the DSB response is the apparent tolerance of cells containing DNA breaks. We suggest that this tolerance makes a major contribution to the oncogenic risk of patients with chromosome instability syndromes.
Collapse
Affiliation(s)
- Paola Porcedda
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Cuadrado M, Martinez-Pastor B, Fernandez-Capetillo O. "ATR activation in response to ionizing radiation: still ATM territory". Cell Div 2006; 1:7. [PMID: 16759429 PMCID: PMC1475580 DOI: 10.1186/1747-1028-1-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Accepted: 05/17/2006] [Indexed: 11/12/2022] Open
Abstract
Unrepaired DNA double-strand breaks (DSBs) are a major cause for genomic instability. Therefore, upon detection of a DSB a rapid response must be assembled to coordinate the proper repair/signaling of the lesion or the elimination of cells with unsustainable amounts of DNA damage. Three members of the PIKK family of protein kinases -ATM, ATR and DNA-PKcs- take the lead and initiate the signaling cascade emanating from DSB sites. Whereas DNA-PKcs activity seems to be restricted to the phosphorylation of targets involved in DNA repair, ATM and ATR phosphorylate a broad spectrum of cell cycle regulators and DNA repair proteins. In the canonical model, ATM and ATR are activated by two different types of lesions and signal through two independent and alternate pathways. Specifically, ATR is activated by various forms of DNA damage, including DSBs, arising at stalled replication forks ("replication stress"), and ATM is responsible for the signaling of DSBs that are not associated with the replication machinery throughout the cell cycle. Recent evidence suggests that this model might be oversimplified and that coordinated crosstalk between ATM and ATR activation routes goes on at the core of the DNA damage response.
Collapse
Affiliation(s)
- Myriam Cuadrado
- Genomic Instability Group, Spanish National Cancer Center, Madrid, Spain
| | | | | |
Collapse
|
37
|
Sivozhelezov V, Giacomelli L, Tripathi S, Nicolini C. Gene expression in the cell cycle of human T lymphocytes: I. Predicted gene and protein networks. J Cell Biochem 2006; 97:1137-50. [PMID: 16315318 DOI: 10.1002/jcb.20693] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The key genes involved in the cell cycle of human T lymphocytes were identified by iterative searches of gene-related databases, as derived also from DNA microarray experimentation, revealing and predicting interactions between those genes, assigning scores to each of the genes according to numbers of interaction for each gene weighted by significance of each interaction, and finally applying several types of clustering algorithms to genes basing on the assigned scores. All clustering algorithms applied, both hierarchical and K-means, invariably selected the same six "leader" genes involved in controlling the cell cycle of human T lymphocytes. Relations of the six genes to experimental data describing switching between stages of cell cycle of human T lymphocytes are discussed.
Collapse
Affiliation(s)
- V Sivozhelezov
- Fondazione Elba, Via delle Testuggini snc, 00100 Rome, Italy
| | | | | | | |
Collapse
|