166 results on '"Kastan MB"'
Search Results
2. Frequent detection of tumor cells in hematopoietic grafts in neuroblastoma and Ewing’s sarcoma
- Author
-
Leung, W, Chen, AR, Klann, RC, Moss, TJ, Davis, JM, Noga, SJ, Cohen, KJ, Friedman, AD, Small, D, Schwartz, CL, Borowitz, MJ, Wharam, MD, Paidas, CN, Long, CA, Karandish, S, McMannis, JD, Kastan, MB, and Civin, CI
- Published
- 1998
- Full Text
- View/download PDF
3. A Novel Dual ATM/DNA-PK Inhibitor, XRD-0394, Potently Radiosensitizes and Potentiates PARP and Topoisomerase I Inhibitors.
- Author
-
Gilmer TM, Lai CH, Guo K, Deland K, Ashcraft KA, Stewart AE, Wang Y, Fu J, Wood KC, Kirsch DG, and Kastan MB
- Subjects
- Humans, Animals, Mice, Xenograft Model Antitumor Assays, Cell Line, Tumor, Female, Drug Synergism, Topoisomerase I Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Ataxia Telangiectasia Mutated Proteins antagonists & inhibitors, Ataxia Telangiectasia Mutated Proteins metabolism, Radiation-Sensitizing Agents pharmacology, DNA-Activated Protein Kinase antagonists & inhibitors, DNA-Activated Protein Kinase metabolism
- Abstract
A majority of patients with cancer receive radiotherapy as part of their treatment regimens whether using external beam therapy or locally-delivered radioisotopes. While often effective, some tumors are inadequately controlled with radiation and radiotherapy has significant short-term and long-term toxicities for cancer survivors. Insights into molecular mechanisms involved in cellular responses to DNA breaks introduced by radiation or other cancer therapies have been gained in recent years and approaches to manipulate these responses to enhance tumor cell killing or reduce normal tissue toxicity are of great interest. Here, we report the identification and initial characterization of XRD-0394, a potent and specific dual inhibitor of two DNA damage response kinases, ATM and DNA-PKcs. This orally bioavailable molecule demonstrates significantly enhanced tumor cell kill in the setting of therapeutic ionizing irradiation in vitro and in vivo. XRD-0394 also potentiates the effectiveness of topoisomerase I inhibitors in vitro. In addition, in cells lacking BRCA1/2 XRD-0394 shows single-agent activity and synergy in combination with PARP inhibitors. A phase Ia clinical trial (NCT05002140) with XRD-0394 in combination with radiotherapy has completed. These results provide a rationale for future clinical trials with XRD-0394 in combination with radiotherapy, PARP inhibitors, and targeted delivery of topoisomerase I inhibitors., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
4. Participation of ATM, SMG1, and DDX5 in a DNA Damage-Induced Alternative Splicing Pathway.
- Author
-
McCann JJ, Fleenor DE, Chen J, Lai CH, Bass TE, and Kastan MB
- Subjects
- Humans, Protein Serine-Threonine Kinases genetics, RNA Precursors genetics, RNA Precursors metabolism, DNA Damage, DEAD-box RNA Helicases genetics, DEAD-box RNA Helicases metabolism, Ataxia Telangiectasia Mutated Proteins metabolism, Alternative Splicing, Neoplasms
- Abstract
Altered cellular responses to DNA damage can contribute to cancer development, progression, and therapeutic resistance. Mutations in key DNA damage response factors occur across many cancer types, and the DNA damage-responsive gene, TP53, is frequently mutated in a high percentage of cancers. We recently reported that an alternative splicing pathway induced by DNA damage regulates alternative splicing of TP53 RNA and further modulates cellular stress responses. Through damage-induced inhibition of the SMG1 kinase, TP53 pre-mRNA is alternatively spliced to generate TP53b mRNA and p53b protein is required for optimal induction of cellular senescence after ionizing radiation-induced DNA damage. Herein, we confirmed and extended these observations by demonstrating that the ATM protein kinase is required for repression of SMG1 kinase activity after ionizing radiation. We found that the RNA helicase and splicing factor, DDX5, interacts with SMG1, is required for alternative splicing of TP53 pre-mRNA to TP53b and TP53c mRNAs after DNA damage, and contributes to radiation-induced cellular senescence. Interestingly, the role of SMG1 in alternative splicing of p53 appears to be distinguishable from its role in regulating nonsense-mediated RNA decay. Thus, ATM, SMG1, and DDX5 participate in a DNA damage-induced alternative splicing pathway that regulates TP53 splicing and modulates radiation-induced cellular senescence., (©2023 by Radiation Research Society. All rights of reproduction in any form reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. ATM Regulation of the Cohesin Complex Is Required for Repression of DNA Replication and Transcription in the Vicinity of DNA Double-Strand Breaks.
- Author
-
Bass TE, Fleenor DE, Burrell PE, and Kastan MB
- Subjects
- Humans, Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, DNA metabolism, Phosphorylation, Cohesins, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, DNA Replication
- Abstract
Implications: Multiple members of the cohesin complex are involved in the regulation of DNA replication and transcription in the vicinity of DNA double-strand breaks and their role(s) are regulated by the ATM kinase., (©2022 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
6. DNA-Damage-Induced Alternative Splicing of p53.
- Author
-
Chen J, Zhang D, Qin X, Owzar K, McCann JJ, and Kastan MB
- Abstract
Cellular responses to DNA damage and other stresses are important determinants of mutagenesis and impact the development of a wide range of human diseases. TP53 is highly mutated in human cancers and plays an essential role in stress responses and cell fate determination. A central dogma of p53 induction after DNA damage has been that the induction results from a transient increase in the half-life of the p53 protein. Our laboratory recently demonstrated that this long-standing paradigm is an incomplete picture of p53 regulation by uncovering a critical role for protein translational regulation in p53 induction after DNA damage. These investigations led to the discovery of a DNA-damage-induced alternative splicing (AS) pathway that affects p53 and other gene products. The damage-induced AS of p53 pre-mRNA generates the beta isoform of p53 (p53β) RNA and protein, which is specifically required for the induction of cellular senescence markers after ionizing irradiation (IR). In an attempt to elucidate the mechanisms behind the differential regulation and apparent functional divergence between full-length (FL) p53 and the p53β isoform (apoptosis versus senescence, respectively), we identified the differential transcriptome and protein interactome between these two proteins that may result from the unique 10-amino-acid tail in p53β protein.
- Published
- 2021
- Full Text
- View/download PDF
7. Retrospective Diagnosis of Ataxia-Telangiectasia in an Adolescent Patient With a Remote History of T-Cell Leukemia.
- Author
-
Sze SK, Lederman HM, Crawford TO, Wangler MF, Lewis AM, Kastan MB, Dibra HK, Taylor AMR, and Wechsler DS
- Subjects
- Adolescent, Adult, Ataxia Telangiectasia etiology, Combined Modality Therapy, Female, Humans, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma therapy, Prognosis, Retrospective Studies, Ataxia Telangiectasia diagnosis, Ataxia Telangiectasia Mutated Proteins genetics, Mutation, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma complications
- Abstract
Ataxia-telangiectasia (A-T) is a rare autosomal recessive disorder characterized by progressive cerebellar degeneration that is typically diagnosed in early childhood. A-T is associated with a predisposition to malignancies, particularly lymphoid tumors in childhood and early adulthood. An adolescent girl with minimal neurologic symptoms was diagnosed with A-T 8 years after completing therapy for T-cell acute lymphoblastic leukemia, following a diagnosis of ATM-mutated breast cancer in her mother. We highlight the importance of recognizing ATM mutations in T-cell acute lymphoblastic leukemia, appreciating the phenotypic heterogeneity of A-T, and defining optimal cancer screening in A-T patients.
- Published
- 2021
- Full Text
- View/download PDF
8. Impaired endoplasmic reticulum-mitochondrial signaling in ataxia-telangiectasia.
- Author
-
Yeo AJ, Chong KL, Gatei M, Zou D, Stewart R, Withey S, Wolvetang E, Parton RG, Brown AD, Kastan MB, Coman D, and Lavin MF
- Abstract
There is evidence that ATM mutated in ataxia-telangiectasia (A-T) plays a key role in protecting against mitochondrial dysfunction, the mechanism for which remains unresolved. We demonstrate here that ATM-deficient cells are exquisitely sensitive to nutrient deprivation, which can be explained by defective cross talk between the endoplasmic reticulum (ER) and the mitochondrion. Tethering between these two organelles in response to stress was reduced in cells lacking ATM, and consistent with this, Ca
2+ release and transfer between ER and mitochondria was reduced dramatically when compared with control cells. The impact of this on mitochondrial function was evident from an increase in oxygen consumption rates and a defect in mitophagy in ATM-deficient cells. Our findings reveal that ER-mitochondrial connectivity through IP3R1-GRP75-VDAC1, to maintain Ca2+ homeostasis, as well as an abnormality in mitochondrial fusion defective in response to nutrient stress, can account for at least part of the mitochondrial dysfunction observed in A-T cells., Competing Interests: None of the authors have a conflict of interest. This study was not funded by any commercial entity and has not attracted any intellectual property protection or patents., (© 2020 The Authors.)- Published
- 2020
- Full Text
- View/download PDF
9. Low dose chloroquine decreases insulin resistance in human metabolic syndrome but does not reduce carotid intima-media thickness.
- Author
-
McGill JB, Johnson M, Hurst S, Cade WT, Yarasheski KE, Ostlund RE, Schechtman KB, Razani B, Kastan MB, McClain DA, de las Fuentes L, Davila-Roman VG, Ory DS, Wickline SA, and Semenkovich CF
- Abstract
Background: Metabolic syndrome, an obesity-related condition associated with insulin resistance and low-grade inflammation, leads to diabetes, cardiovascular diseases, cancer, osteoarthritis, and other disorders. Optimal therapy is unknown. The antimalarial drug chloroquine activates the kinase ataxia telangiectasia mutated (ATM), improves metabolic syndrome and reduces atherosclerosis in mice. To translate this observation to humans, we conducted two clinical trials of chloroquine in people with the metabolic syndrome., Methods: Eligibility included adults with at least 3 criteria of metabolic syndrome but who did not have diabetes. Subjects were studied in the setting of a single academic health center. The specific hypothesis: chloroquine improves insulin sensitivity and decreases atherosclerosis. In Trial 1, the intervention was chloroquine dose escalations in 3-week intervals followed by hyperinsulinemic euglycemic clamps. Trial 2 was a parallel design randomized clinical trial, and the intervention was chloroquine, 80 mg/day, or placebo for 1 year. The primary outcomes were clamp determined-insulin sensitivity for Trial 1, and carotid intima-media thickness (CIMT) for Trial 2. For Trial 2, subjects were allocated based on a randomization sequence using a protocol in blocks of 8. Participants, care givers, and those assessing outcomes were blinded to group assignment., Results: For Trial 1, 25 patients were studied. Chloroquine increased hepatic insulin sensitivity without affecting glucose disposal, and improved serum lipids. For Trial 2, 116 patients were randomized, 59 to chloroquine (56 analyzed) and 57 to placebo (51 analyzed). Chloroquine had no effect on CIMT or carotid contrast enhancement by MRI, a pre-specified secondary outcome. The pre-specified secondary outcomes of blood pressure, lipids, and activation of JNK (a stress kinase implicated in diabetes and atherosclerosis) were decreased by chloroquine. Adverse events were similar between groups., Conclusions: These findings suggest that low dose chloroquine, which improves the metabolic syndrome through ATM-dependent mechanisms in mice, modestly improves components of the metabolic syndrome in humans but is unlikely to be clinically useful in this setting. Trial registration ClinicalTrials.gov (NCT00455325, NCT00455403), both posted 03 April 2007., Competing Interests: Competing interestsThe authors declare that they have no competing interests.
- Published
- 2019
- Full Text
- View/download PDF
10. A novel DNA damage-induced alternative splicing pathway that regulates p53 and cellular senescence markers.
- Author
-
Chen J and Kastan MB
- Abstract
Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
11. Identification of a DNA Damage-Induced Alternative Splicing Pathway That Regulates p53 and Cellular Senescence Markers.
- Author
-
Chen J, Crutchley J, Zhang D, Owzar K, and Kastan MB
- Subjects
- Alternative Splicing genetics, Alternative Splicing radiation effects, CRISPR-Cas Systems genetics, Cell Line, Tumor, Cellular Senescence radiation effects, DNA Damage radiation effects, Humans, Phosphatidylinositol 3-Kinases metabolism, Protein Binding radiation effects, Protein Serine-Threonine Kinases, RNA Precursors genetics, RNA Precursors radiation effects, RNA-Binding Proteins genetics, Radiation, Ionizing, Ribosomal Proteins metabolism, Signal Transduction genetics, Signal Transduction radiation effects, Tumor Suppressor Protein p53 metabolism, Cellular Senescence genetics, DNA Damage genetics, Phosphatidylinositol 3-Kinases genetics, Ribosomal Proteins genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Cellular responses to DNA damage are critical determinants of cancer development and aging-associated pathogenesis. Here, we identify and characterize a DNA-damage response (DDR) pathway that regulates alternative splicing of numerous gene products, including the human tumor suppressor TP53 , and controls DNA damage-induced cellular senescence. In brief, ionizing radiation (IR) inhibits the activity of SMG1, a phosphoinositide-3-kinase-like kinase family member, reducing the binding of SMG1 to a specific region near exon 9 of p53 precursor mRNA and promoting the binding of ribosomal protein L26 (RPL26) to p53 pre-mRNA. RPL26, in turn, is required for the recruitment of the serine/arginine-rich splicing factor SRSF7 to p53 pre-mRNA and generation of alternatively spliced p53β RNA. Disruption of this pathway via selective knockout of p53β by CRISPR/Cas9 or downregulation of pathway constituents significantly reduces IR-induced senescence markers, and cells lacking p53β expression fail to transcriptionally repress negative regulators of cellular senescence and aging. Significance: We identified a new component of the DDR pathway that regulates alternative splicing of messenger RNAs, including human TP53 mRNA. Modulation of this regulatory pathway affects DNA-damage induction of cellular senescence markers. Cancer Discov; 7(7); 766-81. ©2017 AACR. This article is highlighted in the In This Issue feature, p. 653 ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
12. Self-inflicted DNA double-strand breaks sustain tumorigenicity and stemness of cancer cells.
- Author
-
Liu X, Li F, Huang Q, Zhang Z, Zhou L, Deng Y, Zhou M, Fleenor DE, Wang H, Kastan MB, and Li CY
- Subjects
- Ataxia Telangiectasia Mutated Proteins genetics, Cell Cycle Proteins genetics, Cytochromes c metabolism, DNA Breaks, Double-Stranded, DNA Damage genetics, DNA Replication genetics, DNA Replication physiology, Humans, NF-kappa B metabolism, STAT3 Transcription Factor metabolism, Cell Cycle Proteins metabolism
- Abstract
DNA double-strand breaks (DSBs) are traditionally associated with cancer through their abilities to cause chromosomal instabilities or gene mutations. Here we report a new class of self-inflicted DNA DSBs that can drive tumor growth irrespective of their effects on genomic stability. We discover a mechanism through which cancer cells cause DSBs in their own genome spontaneously independent of reactive oxygen species or replication stress. In this mechanism, low-level cytochrome c leakage from the mitochondria leads to sublethal activation of apoptotic caspases and nucleases, which causes DNA DSBs. In response to these spontaneous DNA DSBs, ATM, a key factor involved in DNA damage response, is constitutively activated. Activated ATM leads to activation of transcription factors NF-κB and STAT3, known drivers of tumor growth. Moreover, self-inflicted DNA DSB formation and ATM activation are important in sustaining the stemness of patient-derived glioma cells. In human tumor tissues, elevated levels of activated ATM correlate with poor patient survival. Self-inflicted DNA DSBs therefore are functionally important for maintaining the malignancy of cancer cells.
- Published
- 2017
- Full Text
- View/download PDF
13. Commentary on "Participation of p53 Protein in the Cellular Response to DNA Damage".
- Author
-
Kastan MB
- Subjects
- Humans, DNA Damage, Tumor Suppressor Protein p53 genetics
- Published
- 2016
- Full Text
- View/download PDF
14. Perspectives from man's best friend: National Academy of Medicine's Workshop on Comparative Oncology.
- Author
-
LeBlanc AK, Breen M, Choyke P, Dewhirst M, Fan TM, Gustafson DL, Helman LJ, Kastan MB, Knapp DW, Levin WJ, London C, Mason N, Mazcko C, Olson PN, Page R, Teicher BA, Thamm DH, Trent JM, Vail DM, and Khanna C
- Subjects
- Animals, Clinical Trials as Topic, Dogs, Drug Discovery, Humans, Academies and Institutes, Education, Neoplasms drug therapy, Societies, Scientific
- Published
- 2016
- Full Text
- View/download PDF
15. Optimization of a Novel Series of Ataxia-Telangiectasia Mutated Kinase Inhibitors as Potential Radiosensitizing Agents.
- Author
-
Min J, Guo K, Suryadevara PK, Zhu F, Holbrook G, Chen Y, Feau C, Young BM, Lemoff A, Connelly MC, Kastan MB, and Guy RK
- Subjects
- Animals, Colony-Forming Units Assay, Drug Delivery Systems, Drug Design, Female, Humans, In Vitro Techniques, MCF-7 Cells, Mice, Mice, Inbred C57BL, Microsomes, Liver metabolism, Quinazolines chemical synthesis, Quinazolines pharmacology, Radiation-Sensitizing Agents pharmacokinetics, Structure-Activity Relationship, Substrate Specificity, Ataxia Telangiectasia Mutated Proteins antagonists & inhibitors, Radiation-Sensitizing Agents chemical synthesis, Radiation-Sensitizing Agents pharmacology
- Abstract
We previously reported a novel inhibitor of the ataxia-telangiectasia mutated (ATM) kinase, which is a target for novel radiosensitizing drugs. While our initial lead, compound 4, was relatively potent and nontoxic, it exhibited poor stability to oxidative metabolism and relatively poor selectivity against other kinases. The current study focused on balancing potency and selectivity with metabolic stability through structural modification to the metabolized site on the quinazoline core. We performed extensive structure-activity and structure-property relationship studies on this quinazoline ATM kinase inhibitor in order to identify structural variants with enhanced selectivity and metabolic stability. We show that, while the C-7-methoxy group is essential for potency, replacing the C-6-methoxy group considerably improves metabolic stability without affecting potency. Promising analogues 20, 27g, and 27n were selected based on in vitro pharmacology and evaluated in murine pharmacokinetic and tolerability studies. Compound 27g possessed significantly improve pharmacokinetics relative to that of 4. Compound 27g was also significantly more selective against other kinases than 4. Therefore, 27g is a good candidate for further development as a potential radiosensitizer.
- Published
- 2016
- Full Text
- View/download PDF
16. Chromatin perturbations during the DNA damage response in higher eukaryotes.
- Author
-
Bakkenist CJ and Kastan MB
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, DNA metabolism, DNA Breaks, Double-Stranded, Eukaryota metabolism, Humans, Chromatin metabolism, DNA Repair, Eukaryota genetics, Signal Transduction
- Abstract
The DNA damage response is a widely used term that encompasses all signaling initiated at DNA lesions and damaged replication forks as it extends to orchestrate DNA repair, cell cycle checkpoints, cell death and senescence. ATM, an apical DNA damage signaling kinase, is virtually instantaneously activated following the introduction of DNA double-strand breaks (DSBs). The MRE11-RAD50-NBS1 (MRN) complex, which has a catalytic role in DNA repair, and the KAT5 (Tip60) acetyltransferase are required for maximal ATM kinase activation in cells exposed to low doses of ionizing radiation. The sensing of DNA lesions occurs within a highly complex and heterogeneous chromatin environment. Chromatin decondensation and histone eviction at DSBs may be permissive for KAT5 binding to H3K9me3 and H3K36me3, ATM kinase acetylation and activation. Furthermore, chromatin perturbation may be a prerequisite for most DNA repair. Nucleosome disassembly during DNA repair was first reported in the 1970s by Smerdon and colleagues when nucleosome rearrangement was noted during the process of nucleotide excision repair of UV-induced DNA damage in human cells. Recently, the multi-functional protein nucleolin was identified as the relevant histone chaperone required for partial nucleosome disruption at DBSs, the recruitment of repair enzymes and for DNA repair. Notably, ATM kinase is activated by chromatin perturbations induced by a variety of treatments that do not directly cause DSBs, including treatment with histone deacetylase inhibitors. Central to the mechanisms that activate ATR, the second apical DNA damage signaling kinase, outside of a stalled and collapsed replication fork in S-phase, is chromatin decondensation and histone eviction associated with DNA end resection at DSBs. Thus, a stress that is common to both ATM and ATR kinase activation is chromatin perturbations, and we argue that chromatin perturbations are both sufficient and required for induction of the DNA damage response., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
17. ATM functions at the peroxisome to induce pexophagy in response to ROS.
- Author
-
Zhang J, Tripathi DN, Jing J, Alexander A, Kim J, Powell RT, Dere R, Tait-Mulder J, Lee JH, Paull TT, Pandita RK, Charaka VK, Pandita TK, Kastan MB, and Walker CL
- Subjects
- Adaptor Proteins, Signal Transducing metabolism, Animals, Ataxia Telangiectasia Mutated Proteins genetics, Blotting, Western, Cells, Cultured, HEK293 Cells, Hep G2 Cells, Humans, Hydrogen Peroxide pharmacology, MCF-7 Cells, Mechanistic Target of Rapamycin Complex 1, Mice, Knockout, Microscopy, Electron, Microscopy, Fluorescence, Multiprotein Complexes metabolism, Mutation, Peroxisome-Targeting Signal 1 Receptor, Peroxisomes ultrastructure, Phagosomes metabolism, Phagosomes ultrastructure, Phosphorylation drug effects, Protein Binding, RNA Interference, Receptors, Cytoplasmic and Nuclear genetics, Reverse Transcriptase Polymerase Chain Reaction, Sequestosome-1 Protein, Serine genetics, Serine metabolism, TOR Serine-Threonine Kinases metabolism, Ataxia Telangiectasia Mutated Proteins metabolism, Autophagy, Peroxisomes metabolism, Reactive Oxygen Species metabolism, Receptors, Cytoplasmic and Nuclear metabolism
- Abstract
Peroxisomes are highly metabolic, autonomously replicating organelles that generate reactive oxygen species (ROS) as a by-product of fatty acid β-oxidation. Consequently, cells must maintain peroxisome homeostasis, or risk pathologies associated with too few peroxisomes, such as peroxisome biogenesis disorders, or too many peroxisomes, inducing oxidative damage and promoting diseases such as cancer. We report that the PEX5 peroxisome import receptor binds ataxia-telangiectasia mutated (ATM) and localizes this kinase to the peroxisome. In response to ROS, ATM signalling activates ULK1 and inhibits mTORC1 to induce autophagy. Specificity for autophagy of peroxisomes (pexophagy) is provided by ATM phosphorylation of PEX5 at Ser 141, which promotes PEX5 monoubiquitylation at Lys 209, and recognition of ubiquitylated PEX5 by the autophagy adaptor protein p62, directing the autophagosome to peroxisomes to induce pexophagy. These data reveal an important new role for ATM in metabolism as a sensor of ROS that regulates pexophagy.
- Published
- 2015
- Full Text
- View/download PDF
18. Repair versus Checkpoint Functions of BRCA1 Are Differentially Regulated by Site of Chromatin Binding.
- Author
-
Goldstein M and Kastan MB
- Subjects
- BRCA1 Protein metabolism, Binding Sites, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Cycle Checkpoints genetics, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Chromatin metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, HeLa Cells, Histone Chaperones, Humans, M Phase Cell Cycle Checkpoints genetics, MCF-7 Cells, Nuclear Proteins genetics, Nuclear Proteins metabolism, S Phase Cell Cycle Checkpoints genetics, Ubiquitin-Protein Ligases, BRCA1 Protein genetics, Chromatin genetics, DNA Breaks, Double-Stranded, DNA End-Joining Repair, Genes, BRCA1, Genes, cdc
- Abstract
The product of the Brca1 tumor-suppressor gene is involved in multiple aspects of the cellular DNA damage response (DDR), including activation of cell-cycle arrests and DNA double-stranded break (DSB) repair by homologous recombination. Prior reports demonstrated that BRCA1 recruitment to areas of DNA breakage depended on RAP80 and the RNF8/RNF168 E3 ubiquitin ligases. Here, we extend these findings by showing that RAP80 is only required for the binding of BRCA1 to regions flanking the DSB, whereas BRCA1 binding directly to DNA breaks requires Nijmegen breakage syndrome 1 (NBS1). These differential recruitment mechanisms differentially affect BRCA1 functions: (i) RAP80-dependent recruitment of BRCA1 to chromatin flanking DNA breaks is required for BRCA1 phosphorylation at serine 1387 and 1423 by ATM and, consequently, for the activation of S and G(2) checkpoints; and (ii) BRCA1 interaction with NBS1 upon DSB induction results in an NBS1-dependent recruitment of BRCA1 directly to the DNA break and is required for nonhomologous end-joining repair. Together, these findings illustrate that spatially distinct fractions of BRCA1 exist at the DSB site, which are recruited by different mechanisms and execute different functions in the DDR., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
19. HIF-1 Alpha Regulates the Response of Primary Sarcomas to Radiation Therapy through a Cell Autonomous Mechanism.
- Author
-
Zhang M, Qiu Q, Li Z, Sachdeva M, Min H, Cardona DM, DeLaney TF, Han T, Ma Y, Luo L, Ilkayeva OR, Lui K, Nichols AG, Newgard CB, Kastan MB, Rathmell JC, Dewhirst MW, and Kirsch DG
- Subjects
- Animals, Cell Line, Tumor, Chemoradiotherapy, Gene Knockdown Techniques, Humans, Hypoxia-Inducible Factor 1, alpha Subunit deficiency, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Mice, Mitochondria metabolism, Mitochondria radiation effects, Mitochondrial Size genetics, Mitochondrial Size radiation effects, Radiation Tolerance genetics, Radiation Tolerance radiation effects, Sarcoma genetics, Sarcoma pathology, Treatment Outcome, Up-Regulation drug effects, Up-Regulation radiation effects, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Sarcoma metabolism, Sarcoma radiotherapy
- Abstract
Hypoxia is a major cause of radiation resistance, which may predispose to local recurrence after radiation therapy. While hypoxia increases tumor cell survival after radiation exposure because there is less oxygen to oxidize damaged DNA, it remains unclear whether signaling pathways triggered by hypoxia contribute to radiation resistance. For example, intratumoral hypoxia can increase hypoxia inducible factor 1 alpha (HIF-1α), which may regulate pathways that contribute to radiation sensitization or radiation resistance. To clarify the role of HIF-1α in regulating tumor response to radiation, we generated a novel genetically engineered mouse model of soft tissue sarcoma with an intact or deleted HIF-1α. Deletion of HIF-1α sensitized primary sarcomas to radiation exposure in vivo. Moreover, cell lines derived from primary sarcomas lacking HIF-1α, or in which HIF-1α was knocked down, had decreased clonogenic survival in vitro, demonstrating that HIF-1α can promote radiation resistance in a cell autonomous manner. In HIF-1α-intact and -deleted sarcoma cells, radiation-induced reactive oxygen species, DNA damage repair and activation of autophagy were similar. However, sarcoma cells lacking HIF-1α had impaired mitochondrial biogenesis and metabolic response after irradiation, which might contribute to radiation resistance. These results show that HIF-1α promotes radiation resistance in a cell autonomous manner.
- Published
- 2015
- Full Text
- View/download PDF
20. The DNA damage response: implications for tumor responses to radiation and chemotherapy.
- Author
-
Goldstein M and Kastan MB
- Subjects
- Antineoplastic Agents pharmacology, DNA Damage drug effects, DNA Damage radiation effects, DNA Methylation drug effects, DNA Methylation genetics, DNA Methylation radiation effects, DNA Repair drug effects, DNA Repair radiation effects, Humans, Neoplasms therapy, Antineoplastic Agents therapeutic use, DNA Damage genetics, DNA Repair genetics, Neoplasms genetics, Radiotherapy
- Abstract
Cellular responses to DNA damage are important determinants of both cancer development and cancer outcome following radiation therapy and chemotherapy. Identification of molecular pathways governing DNA damage signaling and DNA repair in response to different types of DNA lesions allows for a better understanding of the effects of radiation and chemotherapy on normal and tumor cells. Although dysregulation of the DNA damage response (DDR) is associated with predisposition to cancer development, it can also result in hypersensitivity or resistance of tumors to therapy and can be exploited for improvement of cancer treatment. We highlight the DDR pathways that are activated after treatment with radiation and different classes of chemotherapeutic drugs and describe mechanisms determining tumor sensitivity and resistance to these agents. Further, we discuss approaches to enhance tumor sensitivity to radiation and chemotherapy by modulating the DDR with a goal of enhancing the effectiveness of cancer therapies.
- Published
- 2015
- Full Text
- View/download PDF
21. The scaffold protein WRAP53β orchestrates the ubiquitin response critical for DNA double-strand break repair.
- Author
-
Henriksson S, Rassoolzadeh H, Hedström E, Coucoravas C, Julner A, Goldstein M, Imreh G, Zhivotovsky B, Kastan MB, Helleday T, and Farnebo M
- Subjects
- Adaptor Proteins, Signal Transducing, Ataxia Telangiectasia Mutated Proteins metabolism, Cell Cycle Proteins, Cell Line, Tumor, Cells, Cultured, DNA Breaks, Double-Stranded, DNA Repair genetics, DNA-Binding Proteins metabolism, HeLa Cells, Histones metabolism, Humans, Molecular Chaperones, Nuclear Proteins metabolism, Phosphorylation, Protein Binding, Protein Structure, Tertiary, Telomerase genetics, Trans-Activators metabolism, Ubiquitin-Protein Ligases, DNA Repair physiology, Telomerase metabolism, Ubiquitin metabolism
- Abstract
The WD40 domain-containing protein WRAP53β (WD40 encoding RNA antisense to p53; also referred to as WDR79/TCAB1) controls trafficking of splicing factors and the telomerase enzyme to Cajal bodies, and its functional loss has been linked to carcinogenesis, premature aging, and neurodegeneration. Here, we identify WRAP53β as an essential regulator of DNA double-strand break (DSB) repair. WRAP53β rapidly localizes to DSBs in an ATM-, H2AX-, and MDC1-dependent manner. We show that WRAP53β targets the E3 ligase RNF8 to DNA lesions by facilitating the interaction between RNF8 and its upstream partner, MDC1, in response to DNA damage. Simultaneous binding of MDC1 and RNF8 to the highly conserved WD40 scaffold domain of WRAP53β facilitates their interaction and accumulation of RNF8 at DSBs. In this manner, WRAP53β controls proper ubiquitylation at DNA damage sites and the downstream assembly of 53BP1, BRCA1, and RAD51. Furthermore, we reveal that knockdown of WRAP53β impairs DSB repair by both homologous recombination (HR) and nonhomologous end-joining (NHEJ), causes accumulation of spontaneous DNA breaks, and delays recovery from radiation-induced cell cycle arrest. Our findings establish WRAP53β as a novel regulator of DSB repair by providing a scaffold for DNA repair factors., (© 2014 Henriksson et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2014
- Full Text
- View/download PDF
22. Inactivation of the human papillomavirus E6 or E7 gene in cervical carcinoma cells by using a bacterial CRISPR/Cas RNA-guided endonuclease.
- Author
-
Kennedy EM, Kornepati AV, Goldstein M, Bogerd HP, Poling BC, Whisnant AW, Kastan MB, and Cullen BR
- Subjects
- Alphapapillomavirus isolation & purification, Base Sequence, Cell Line, Tumor, DNA, Viral genetics, Female, Humans, Molecular Sequence Data, Uterine Cervical Neoplasms virology, Alphapapillomavirus genetics, Clustered Regularly Interspaced Short Palindromic Repeats, DNA-Binding Proteins genetics, Endonucleases metabolism, Oncogene Proteins, Viral genetics, Papillomavirus E7 Proteins genetics, Repressor Proteins genetics, Uterine Cervical Neoplasms genetics
- Abstract
High-risk human papillomaviruses (HPVs), including HPV-16 and HPV-18, are the causative agents of cervical carcinomas and are linked to several other tumors of the anogenital and oropharyngeal regions. The majority of HPV-induced tumors contain integrated copies of the normally episomal HPV genome that invariably retain intact forms of the two HPV oncogenes E6 and E7. E6 induces degradation of the cellular tumor suppressor p53, while E7 destabilizes the retinoblastoma (Rb) protein. Previous work has shown that loss of E6 function in cervical cancer cells induces p53 expression as well as downstream effectors that induce apoptosis and cell cycle arrest. Similarly, loss of E7 allows increased Rb expression, leading to cell cycle arrest and senescence. Here, we demonstrate that expression of a bacterial Cas9 RNA-guided endonuclease, together with single guide RNAs (sgRNAs) specific for E6 or E7, is able to induce cleavage of the HPV genome, resulting in the introduction of inactivating deletion and insertion mutations into the E6 or E7 gene. This results in the induction of p53 or Rb, leading to cell cycle arrest and eventual cell death. Both HPV-16- and HPV-18-transformed cells were found to be responsive to targeted HPV genome-specific DNA cleavage. These data provide a proof of principle for the idea that vector-delivered Cas9/sgRNA combinations could represent effective treatment modalities for HPV-induced cancers. Importance: Human papillomaviruses (HPVs) are the causative agents of almost all cervical carcinomas and many other tumors, including many head and neck cancers. In these cancer cells, the HPV DNA genome is integrated into the cellular genome, where it expresses high levels of two viral oncogenes, called E6 and E7, that are required for cancer cell growth and viability. Here, we demonstrate that the recently described bacterial CRISPR/Cas RNA-guided endonuclease can be reprogrammed to target and destroy the E6 or E7 gene in cervical carcinoma cells transformed by HPV, resulting in cell cycle arrest, leading to cancer cell death. We propose that viral vectors designed to deliver E6- and/or E7-specific CRISPR/Cas to tumor cells could represent a novel and highly effective tool to treat and eliminate HPV-induced cancers., (Copyright © 2014, American Society for Microbiology. All Rights Reserved.)
- Published
- 2014
- Full Text
- View/download PDF
23. Rad17 recruits the MRE11-RAD50-NBS1 complex to regulate the cellular response to DNA double-strand breaks.
- Author
-
Wang Q, Goldstein M, Alexander P, Wakeman TP, Sun T, Feng J, Lou Z, Kastan MB, and Wang XF
- Subjects
- Acid Anhydride Hydrolases, Ataxia Telangiectasia Mutated Proteins metabolism, Cell Line, Humans, MRE11 Homologue Protein, Phosphorylation, Protein Binding, Protein Processing, Post-Translational, Cell Cycle Proteins metabolism, DNA Breaks, Double-Stranded, DNA Repair Enzymes metabolism, DNA-Binding Proteins metabolism, Nuclear Proteins metabolism, Protein Multimerization, Signal Transduction
- Abstract
The MRE11-RAD50-NBS1 (MRN) complex is essential for the detection of DNA double-strand breaks (DSBs) and initiation of DNA damage signaling. Here, we show that Rad17, a replication checkpoint protein, is required for the early recruitment of the MRN complex to the DSB site that is independent of MDC1 and contributes to ATM activation. Mechanistically, Rad17 is phosphorylated by ATM at a novel Thr622 site resulting in a direct interaction of Rad17 with NBS1, facilitating recruitment of the MRN complex and ATM to the DSB, thereby enhancing ATM signaling. Repetition of these events creates a positive feedback for Rad17-dependent activation of MRN/ATM signaling which appears to be a requisite for the activation of MDC1-dependent MRN complex recruitment. A point mutation of the Thr622 residue of Rad17 leads to a significant reduction in MRN/ATM signaling and homologous recombination repair, suggesting that Thr622 phosphorylation is important for regulation of the MRN/ATM signaling by Rad17. These findings suggest that Rad17 plays a critical role in the cellular response to DNA damage via regulation of the MRN/ATM pathway.
- Published
- 2014
- Full Text
- View/download PDF
24. Development of a cell-based, high-throughput screening assay for ATM kinase inhibitors.
- Author
-
Guo K, Shelat AA, Guy RK, and Kastan MB
- Subjects
- Antineoplastic Agents, Phytogenic pharmacology, Ataxia Telangiectasia Mutated Proteins metabolism, DNA Damage drug effects, Drug Discovery, Drug Screening Assays, Antitumor, Enzyme Activation drug effects, Etoposide pharmacology, Humans, Reproducibility of Results, Signal Transduction drug effects, Small Molecule Libraries, Ataxia Telangiectasia Mutated Proteins antagonists & inhibitors, High-Throughput Screening Assays, Protein Kinase Inhibitors pharmacology
- Abstract
The ATM (ataxia-telangiectasia, mutated) protein kinase is a major regulator of cellular responses to DNA double-strand breaks (DSBs), DNA lesions that can be caused by ionizing irradiation (IR), oxidative damage, or exposure to certain chemical agents. In response to DSBs, the ATM kinase is activated and subsequently phosphorylates numerous downstream substrates, including p53, Chk2, BRCA1, and KAP1, which affect processes such as cell cycle progression and DNA repair. Numerous studies have demonstrated that loss of ATM function results in enhanced sensitivity to ionizing irradiation in clinically relevant dose ranges, suggesting that ATM kinase is an attractive therapeutic target for enhancing tumor cell kill with radiotherapy. Previously identified small-molecule ATM kinase inhibitors, such as CP466722 and Ku55933, were identified using in vitro kinase assays carried out with recombinant ATM kinase isolated from mammalian cells. Since it has not been feasible to express full-length recombinant ATM in bacterial or baculovirus systems, a robust in vitro screening tool has been lacking. We have developed a cell-based assay that is robust, straightforward, and sensitive. Using this high-throughput assay, we screened more than 7000 compounds and discovered additional small molecules that inhibit the ATM kinase and further validated these hits by secondary assays.
- Published
- 2014
- Full Text
- View/download PDF
25. Pilot study of modified LMB-based therapy for children with ataxia-telangiectasia and advanced stage high grade mature B-cell malignancies.
- Author
-
Sandlund JT, Hudson MM, Kennedy W, Onciu M, and Kastan MB
- Subjects
- Adult, Ataxia Telangiectasia complications, Child, Cyclophosphamide therapeutic use, Cytarabine therapeutic use, Doxorubicin therapeutic use, Etoposide therapeutic use, Female, Follow-Up Studies, Humans, Hydrocortisone therapeutic use, Leucovorin therapeutic use, Lymphoma, B-Cell complications, Male, Methotrexate therapeutic use, Neoplasm Staging, Pilot Projects, Prednisone therapeutic use, Prognosis, Prospective Studies, Vincristine therapeutic use, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Ataxia Telangiectasia drug therapy, Lymphoma, B-Cell drug therapy
- Abstract
Children with ataxia-telangiectasia (A-T) and cancer have a poorer prognosis due in part to increased treatment-related toxicity. We piloted a curative intent approach in five children with A-T who presented with advanced stage (III, n = 2; IV, n = 3) B-NHL (diffuse large B-cell lymphoma, n = 4; Burkitt leukemia, n = 1) using a modified LMB-based protocol. Two achieved sustained CCR (one, CCR at 6 years; one, pulmonary death after 3 years in CCR). Two died from toxicity during induction and 1 failed induction with progressive disease. Novel therapeutic approaches which overcome drug resistance and are less toxic are needed for children with A-T and B-NHL., (Copyright © 2013 Wiley Periodicals, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
26. Nucleolin mediates nucleosome disruption critical for DNA double-strand break repair.
- Author
-
Goldstein M, Derheimer FA, Tait-Mulder J, and Kastan MB
- Subjects
- Acid Anhydride Hydrolases, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Line, Tumor, DNA Repair Enzymes genetics, DNA Repair Enzymes metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Histones genetics, Histones metabolism, Humans, MRE11 Homologue Protein, Nuclear Proteins genetics, Nuclear Proteins metabolism, Nucleosomes genetics, Phosphoproteins genetics, RNA-Binding Proteins genetics, Replication Protein A genetics, Replication Protein A metabolism, Nucleolin, DNA Breaks, Double-Stranded, G1 Phase Cell Cycle Checkpoints, Nucleosomes metabolism, Phosphoproteins metabolism, Protein Multimerization, RNA-Binding Proteins metabolism, Recombinational DNA Repair
- Abstract
Recruitment of DNA repair factors and modulation of chromatin structure at sites of DNA double-strand breaks (DSBs) is a complex and highly orchestrated process. We developed a system that can induce DSBs rapidly at defined endogenous sites in mammalian genomes and enables direct assessment of repair and monitoring of protein recruitment, egress, and modification at DSBs. The tight regulation of the system also permits assessments of relative kinetics and dependencies of events associated with cellular responses to DNA breakage. Distinct advantages of this system over focus formation/disappearance assays for assessing DSB repair are demonstrated. Using ChIP, we found that nucleosomes are partially disassembled around DSBs during nonhomologous end-joining repair in G1-arrested mammalian cells, characterized by a transient loss of the H2A/H2B histone dimer. Nucleolin, a protein with histone chaperone activity, interacts with RAD50 via its arginine-glycine rich domain and is recruited to DSBs rapidly in an MRE11-NBS1-RAD50 complex-dependent manner. Down-regulation of nucleolin abrogates the nucleosome disruption, the recruitment of repair factors, and the repair of the DSB, demonstrating the functional importance of nucleosome disruption in DSB repair and identifying a chromatin-remodeling protein required for the process. Interestingly, the nucleosome disruption that occurs during DSB repair in cycling cells differs in that both H2A/H2B and H3/H4 histone dimers are removed. This complete nucleosome disruption is also dependent on nucleolin and is required for recruitment of replication protein A to DSBs, a marker of DSB processing that is a requisite for homologous recombination repair.
- Published
- 2013
- Full Text
- View/download PDF
27. A tuberous sclerosis complex signalling node at the peroxisome regulates mTORC1 and autophagy in response to ROS.
- Author
-
Zhang J, Kim J, Alexander A, Cai S, Tripathi DN, Dere R, Tee AR, Tait-Mulder J, Di Nardo A, Han JM, Kwiatkowski E, Dunlop EA, Dodd KM, Folkerth RD, Faust PL, Kastan MB, Sahin M, and Walker CL
- Subjects
- Animals, Cell Line, HEK293 Cells, Humans, MCF-7 Cells, Mechanistic Target of Rapamycin Complex 1, Membrane Proteins metabolism, Mice, Multiprotein Complexes metabolism, Protein Binding, Rats, TOR Serine-Threonine Kinases metabolism, Tuberous Sclerosis Complex 1 Protein, Tuberous Sclerosis Complex 2 Protein, Tumor Suppressor Proteins metabolism, Autophagy, Gene Expression Regulation, Enzymologic, Multiprotein Complexes genetics, Peroxisomes metabolism, Reactive Oxygen Species, Signal Transduction, TOR Serine-Threonine Kinases genetics
- Abstract
Subcellular localization is emerging as an important mechanism for mTORC1 regulation. We report that the tuberous sclerosis complex (TSC) signalling node, TSC1, TSC2 and Rheb, localizes to peroxisomes, where it regulates mTORC1 in response to reactive oxygen species (ROS). TSC1 and TSC2 were bound by peroxisomal biogenesis factors 19 and 5 (PEX19 and PEX5), respectively, and peroxisome-localized TSC functioned as a Rheb GTPase-activating protein (GAP) to suppress mTORC1 and induce autophagy. Naturally occurring pathogenic mutations in TSC2 decreased PEX5 binding, and abrogated peroxisome localization, Rheb GAP activity and suppression of mTORC1 by ROS. Cells lacking peroxisomes were deficient in mTORC1 repression by ROS, and peroxisome-localization-deficient TSC2 mutants caused polarity defects and formation of multiple axons in neurons. These data identify a role for the TSC in responding to ROS at the peroxisome, and identify the peroxisome as a signalling organelle involved in regulation of mTORC1.
- Published
- 2013
- Full Text
- View/download PDF
28. Strategies for optimizing the response of cancer and normal tissues to radiation.
- Author
-
Moding EJ, Kastan MB, and Kirsch DG
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents therapeutic use, Chemoradiotherapy adverse effects, Chemoradiotherapy methods, Dose-Response Relationship, Radiation, Humans, Neoplasms metabolism, Neoplasms pathology, Radiation Dosage, Radiation-Protective Agents therapeutic use, Radiation-Sensitizing Agents administration & dosage, Radiation-Sensitizing Agents therapeutic use, Neoplasms radiotherapy, Radiotherapy, Image-Guided adverse effects, Radiotherapy, Image-Guided methods, Radiotherapy, Intensity-Modulated adverse effects, Radiotherapy, Intensity-Modulated methods
- Abstract
Approximately 50% of all patients with cancer receive radiation therapy at some point during the course of their treatment, and the majority of these patients are treated with curative intent. Despite recent advances in the planning of radiation treatment and the delivery of image-guided radiation therapy, acute toxicity and potential long-term side effects often limit the ability to deliver a sufficient dose of radiation to control tumours locally. In the past two decades, a better understanding of the hallmarks of cancer and the discovery of specific signalling pathways by which cells respond to radiation have provided new opportunities to design molecularly targeted therapies to increase the therapeutic window of radiation therapy. Here, we review efforts to develop approaches that could improve outcomes with radiation therapy by increasing the probability of tumour cure or by decreasing normal tissue toxicity.
- Published
- 2013
- Full Text
- View/download PDF
29. A message from the Editor-in-Chief.
- Author
-
Kastan MB
- Subjects
- Humans, Publications, Translational Research, Biomedical, Medical Oncology, Neoplasms
- Published
- 2012
- Full Text
- View/download PDF
30. Chloroquine improves survival and hematopoietic recovery after lethal low-dose-rate radiation.
- Author
-
Lim Y, Hedayati M, Merchant AA, Zhang Y, Yu HH, Kastan MB, Matsui W, and Deweese TL
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Bone Marrow Transplantation methods, Cell Cycle Proteins genetics, Chloroquine administration & dosage, DNA-Binding Proteins genetics, Flow Cytometry methods, Male, Mice, Mice, Inbred C57BL, Protein Serine-Threonine Kinases genetics, Radiation Chimera, Radiation Injuries, Experimental mortality, Radiation-Protective Agents administration & dosage, Survival Rate, Tumor Suppressor Proteins genetics, Bone Marrow Cells radiation effects, Cell Cycle Proteins metabolism, Chloroquine pharmacology, DNA-Binding Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Radiation Injuries, Experimental drug therapy, Radiation-Protective Agents pharmacology, Tumor Suppressor Proteins metabolism
- Abstract
Purpose: We have previously shown that the antimalarial agent chloroquine can abrogate the lethal cellular effects of low-dose-rate (LDR) radiation in vitro, most likely by activating the ataxia-telangiectasia mutated (ATM) protein. Here, we demonstrate that chloroquine treatment also protects against lethal doses of LDR radiation in vivo., Methods and Materials: C57BL/6 mice were irradiated with a total of 12.8 Gy delivered at 9.4 cGy/hour. ATM null mice from the same background were used to determine the influence of ATM. Chloroquine was administered by two intraperitoneal injections of 59.4 μg per 17 g of body weight, 24 hours and 4 hours before irradiation. Bone marrow cells isolated from tibia, fibula, and vertebral bones were transplanted into lethally irradiated CD45 congenic recipient mice by retroorbital injection. Chimerism was assessed by flow cytometry. In vitro methylcellulose colony-forming assay of whole bone marrow cells and fluorescence activated cell sorting analysis of lineage depleted cells were used to assess the effect of chloroquine on progenitor cells., Results: Mice pretreated with chloroquine before radiation exhibited a significantly higher survival rate than did mice treated with radiation alone (80% vs. 31%, p = 0.0026). Chloroquine administration before radiation did not affect the survival of ATM null mice (p = 0.86). Chloroquine also had a significant effect on the early engraftment of bone marrow cells from the irradiated donor mice 6 weeks after transplantation (4.2% vs. 0.4%, p = 0.015)., Conclusion: Chloroquine administration before radiation had a significant effect on the survival of normal but not ATM null mice, strongly suggesting that the in vivo effect, like the in vitro effect, is also ATM dependent. Chloroquine improved the early engraftment of bone marrow cells from LDR-irradiated mice, presumably by protecting the progenitor cells from radiation injury. Chloroquine thus could serve as a very useful drug for protection against the harmful effects of LDR radiation., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
31. Interactions of nucleolin and ribosomal protein L26 (RPL26) in translational control of human p53 mRNA.
- Author
-
Chen J, Guo K, and Kastan MB
- Subjects
- Cell Line, Tumor, Humans, Models, Biological, Phosphoproteins genetics, Point Mutation, Protein Multimerization physiology, RNA, Double-Stranded genetics, RNA, Double-Stranded metabolism, RNA-Binding Proteins genetics, Ribosomal Proteins genetics, Stress, Physiological physiology, Tumor Suppressor Protein p53 genetics, Nucleolin, 3' Untranslated Regions physiology, DNA Damage physiology, Phosphoproteins metabolism, Protein Biosynthesis physiology, RNA-Binding Proteins metabolism, Ribosomal Proteins metabolism, Tumor Suppressor Protein p53 biosynthesis
- Abstract
Ribosomal protein RPL26 enhances p53 translation after DNA damage, and this regulation depends upon interactions between the 5'- and 3'-UTRs of human p53 mRNA (Takagi, M., Absalon, M. J., McLure, K. G., and Kastan, M. B. (2005) Cell 123, 49-63; Chen, J., and Kastan, M. B. (2010) Genes Dev. 24, 2146-2156). In contrast, nucleolin (NCL) suppresses the translation of p53 mRNA and its induction after DNA damage. We confirmed reports that RPL26 and NCL interact with each other and then explored the potential role of this interaction in the translational control of p53 after stress. NCL repression of p53 translation utilizes both the 5'- and 3'-UTRs of p53 mRNA, and NCL binds to the same 5'-3'-UTR interaction region that is critical for the recruitment of RPL26 to p53 mRNA after DNA damage. We also found that NCL is able to oligomerize, consistent with a model in which NCL stabilizes this double-stranded RNA structure. We found that the RNA-binding domain of NCL participates in binding to p53 mRNA, is required for both NCL dimerization and NCL-mediated translational repression, and is the domain of NCL that interacts with RPL26. Excessive RPL26 disrupts NCL dimerization, and point mutations in the NCL-interacting region of RPL26 reduce NCL-RPL26 interactions and attenuate both RPL26 binding to human p53 mRNA and p53 induction by RPL26. These observations suggest a model in which the base pairings in the p53 UTR interaction regions are critical for both translational repression and stress induction of p53 by NCL and RPL26, respectively, and that disruption of a NCL-NCL homodimer by RPL26 may be the switch between translational repression and activation after stress.
- Published
- 2012
- Full Text
- View/download PDF
32. A new role for ATM: regulating mitochondrial function and mitophagy.
- Author
-
Valentin-Vega YA and Kastan MB
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins deficiency, DNA-Binding Proteins deficiency, Fibroblasts enzymology, Fibroblasts pathology, Humans, Membrane Potential, Mitochondrial, Mice, Oxidative Stress, Protein Serine-Threonine Kinases deficiency, Tumor Suppressor Proteins deficiency, Ubiquitin-Protein Ligases metabolism, Autophagy, Cell Cycle Proteins metabolism, DNA-Binding Proteins metabolism, Mitochondria metabolism, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Proteins metabolism
- Abstract
The various pathologies in ataxia telangiectasia (A-T) patients including T-cell lymphomagenesis have been attributed to defects in the DNA damage response pathway because ATM, the gene mutated in this disease, is a key mediator of this process. Analysis of Atm-deficient thymocytes in mice reveals that the absence of this gene results in altered mitochondrial homeostasis, a phenomenon that appears to result from abnormal mitophagy engagement. Interestingly, allelic loss of the autophagic gene Becn1 delays tumorigenesis in Atm-null mice presumably by reversing the mitochondrial abnormalities and not by improving the DNA damage response (DDR) pathway. Thus, ATM plays a critical role in modulating mitochondrial homeostasis perhaps by regulating mitophagy.
- Published
- 2012
- Full Text
- View/download PDF
33. Autophagy links inflammasomes to atherosclerotic progression.
- Author
-
Razani B, Feng C, Coleman T, Emanuel R, Wen H, Hwang S, Ting JP, Virgin HW, Kastan MB, and Semenkovich CF
- Subjects
- Animals, Apoptosis Regulatory Proteins metabolism, Beclin-1, Cholesterol pharmacology, Disease Models, Animal, Haploinsufficiency drug effects, Heterozygote, Inflammation pathology, Macrophage Activation drug effects, Macrophages drug effects, Macrophages metabolism, Macrophages pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Models, Biological, Plaque, Atherosclerotic metabolism, Plaque, Atherosclerotic pathology, Atherosclerosis metabolism, Atherosclerosis pathology, Autophagy drug effects, Disease Progression, Inflammasomes metabolism
- Abstract
We investigated the role of autophagy in atherosclerosis. During plaque formation in mice, autophagic markers colocalized predominantly with macrophages (mφ). Atherosclerotic aortas had elevated levels of p62, suggesting that dysfunctional autophagy is characteristic of plaques. To determine whether autophagy directly influences atherogenesis, we characterized Beclin-1 heterozygous-null and mφ-specific ATG5-null (ATG5-mφKO) mice, commonly used models of autophagy haploinsufficiency and deficiency, respectively. Haploinsufficent Beclin-1 mice had no atherosclerotic phenotype, but ATG5-mφKO mice had increased plaques, suggesting an essential role for basal levels of autophagy in atheroprotection. Defective autophagy is associated with proatherogenic inflammasome activation. Classic inflammasome markers were robustly induced in ATG5-null mφ, especially when coincubated with cholesterol crystals. Moreover, cholesterol crystals appear to be increased in ATG5-mφKO plaques, suggesting a potentially vicious cycle of crystal formation and inflammasome activation in autophagy-deficient plaques. These results show that autophagy becomes dysfunctional in atherosclerosis and its deficiency promotes atherosclerosis in part through inflammasome hyperactivation., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
34. Mitochondrial dysfunction in ataxia-telangiectasia.
- Author
-
Valentin-Vega YA, Maclean KH, Tait-Mulder J, Milasta S, Steeves M, Dorsey FC, Cleveland JL, Green DR, and Kastan MB
- Subjects
- Adenosine Triphosphate metabolism, Animals, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Ataxia Telangiectasia genetics, Ataxia Telangiectasia metabolism, Ataxia Telangiectasia physiopathology, Ataxia Telangiectasia Mutated Proteins, Autophagy, Beclin-1, Cell Cycle Proteins genetics, Cells, Cultured, DNA-Binding Proteins genetics, Fibroblasts cytology, Fibroblasts metabolism, Gene Expression, Humans, Immunoblotting, Kaplan-Meier Estimate, Lymphoma, T-Cell genetics, Lymphoma, T-Cell metabolism, Membrane Potential, Mitochondrial, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Electron, Transmission, Mitochondria genetics, Mitochondria physiology, Oxygen Consumption, Protein Serine-Threonine Kinases genetics, RNA Interference, Reactive Oxygen Species metabolism, Reverse Transcriptase Polymerase Chain Reaction, Thymocytes metabolism, Thymocytes ultrastructure, Tumor Suppressor Proteins genetics, Cell Cycle Proteins metabolism, DNA-Binding Proteins metabolism, Mitochondria metabolism, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Ataxia-telangiectasia mutated (ATM) plays a central role in DNA damage responses, and its loss leads to development of T-cell malignancies. Here, we show that ATM loss also leads to intrinsic mitochondrial abnormalities in thymocytes, including elevated reactive oxygen species, increased aberrant mitochondria, high cellular respiratory capacity, and decreased mitophagy. A fraction of ATM protein is localized in mitochondria, and it is rapidly activated by mitochondrial dysfunction. Unexpectedly, allelic loss of the autophagy regulator Beclin-1 significantly delayed tumor development in ATM-null mice. This effect was not associated with rescue of DNA damage signaling but rather with a significant reversal of the mitochondrial abnormalities. These data support a model in which ATM plays direct roles in modulating mitochondrial homeostasis and suggest that mitochondrial dysfunction and associated increases in mitochondrial reactive oxygen species contribute to the cancer-prone phenotype observed in organisms lacking ATM. Thus, ataxia-telangiectasia should be considered, at least in part, as a mitochondrial disease.
- Published
- 2012
- Full Text
- View/download PDF
35. 5'-3'-UTR interactions regulate p53 mRNA translation and provide a target for modulating p53 induction after DNA damage.
- Author
-
Chen J and Kastan MB
- Subjects
- Humans, Oligonucleotides pharmacology, Protein Binding, RNA, Double-Stranded metabolism, Ribosomal Proteins metabolism, Stress, Physiological drug effects, 3' Untranslated Regions, 5' Untranslated Regions, DNA Damage physiology, Gene Expression Regulation, RNA, Messenger metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Optimal induction of p53 protein after DNA damage requires RPL26-mediated increases in p53 mRNA translation. We report here the existence of a dsRNA region containing complementary sequences of the 5'- and 3'-untranslated regions (UTRs) of human p53 mRNA that is critical for its translational regulation by RPL26. Mutating as few as 3 bases in either of the two complementary UTR sequences abrogates the ability of RPL26 to bind to p53 mRNA and stimulate p53 translation, while compensatory mutations restore this binding and regulation. Short, single-strand oligonucleotides that target this 5'-3'-UTR base-pairing region blunt the binding of RPL26 to p53 mRNA in cells and reduce p53 induction and p53-mediated cell death after several different types of DNA damage and cellular stress. The ability to reduce stress induction of p53 with oligonucleotides or other small molecules has numerous potential therapeutic uses.
- Published
- 2010
- Full Text
- View/download PDF
36. Multiple roles of ATM in monitoring and maintaining DNA integrity.
- Author
-
Derheimer FA and Kastan MB
- Subjects
- Cell Cycle genetics, DNA Damage genetics, DNA Repair genetics, Genetic Diseases, Inborn genetics, Genomic Instability genetics, Humans, Protein Serine-Threonine Kinases genetics, Transcriptional Activation genetics, Ataxia Telangiectasia genetics
- Abstract
The ability of our cells to maintain genomic integrity is fundamental for protection from cancer development. Central to this process is the ability of cells to recognize and repair DNA damage and progress through the cell cycle in a regulated and orderly manner. In addition, protection of chromosome ends through the proper assembly of telomeres prevents loss of genetic information and aberrant chromosome fusions. Cells derived from patients with ataxia-telangiectasia (A-T) show defects in cell cycle regulation, abnormal responses to DNA breakage, and chromosomal end-to-end fusions. The identification and characterization of the ATM (ataxia-telangiectasia, mutated) gene product has provided an essential tool for researchers in elucidating cellular mechanisms involved in cell cycle control, DNA repair, and chromosomal stability., (Copyright 2010 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
37. Deficient innate immunity, thymopoiesis, and gene expression response to radiation in survivors of childhood acute lymphoblastic leukemia.
- Author
-
Leung W, Neale G, Behm F, Iyengar R, Finkelstein D, Kastan MB, and Pui CH
- Subjects
- Adolescent, Adult, Child, DNA Damage, Humans, Lymphopenia complications, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Thymus Gland cytology, Thymus Gland immunology, Time, Young Adult, Gene Expression radiation effects, Immunity, Innate, Precursor Cell Lymphoblastic Leukemia-Lymphoma immunology, Survivors, T-Lymphocytes immunology
- Abstract
Background: Survivors of childhood acute lymphoblastic leukemia (ALL) are at an increased risk of developing secondary malignant neoplasms. Radiation and chemotherapy can cause mutations and cytogenetic abnormalities and induce genomic instability. Host immunity and appropriate DNA damage responses are critical inhibitors of carcinogenesis. Therefore, we sought to determine the long-term effects of ALL treatment on immune function and response to DNA damage., Methods: Comparative studies on 14 survivors in first complete remission and 16 siblings were conducted., Results: In comparison to siblings on the cells that were involved in adaptive immunity, the patients had either higher numbers (CD19+ B cells and CD4+CD25+ T regulatory cells) or similar numbers (alphabetaT cells and CD45RO+/RA- memory T cells) in the blood. In contrast, patients had lower numbers of all lymphocyte subsets involved in innate immunity (gammadeltaT cells and all NK subsets, including KIR2DL1+ cells, KIR2DL2/L3+ cells, and CD16+ cells), and lower natural cytotoxicity against K562 leukemia cells. Thymopoiesis was lower in patients, as demonstrated by less CD45RO-/RA+ naïve T cell and less SjTREC levels in the blood, whereas the Vbeta spectratype complexity score was similar. Array of gene expression response to low-dose radiation showed that about 70% of the probesets had a reduced response in patients. One of these genes, SCHIP-1, was also among the top-ranked single nucleotide polymorphisms (SNPs) during the whole-genome scanning by SNP microarray analysis., Conclusion: ALL survivors were deficient in innate immunity, thymopoiesis, and DNA damage responses to radiation. These defects may contribute to their increased likelihood of second malignancy., (Copyright (c) 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
38. The NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis.
- Author
-
Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M, and Kanneganti TD
- Subjects
- Animals, Apoptosis Regulatory Proteins, CARD Signaling Adaptor Proteins, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Proliferation, Colitis chemically induced, Colitis microbiology, Cytoskeletal Proteins immunology, Dextran Sulfate, Disease Models, Animal, Epithelial Cells cytology, Interleukin-18 biosynthesis, Interleukin-18 immunology, Intestinal Absorption, Mice, Mice, Inbred C57BL, Mice, Knockout, NLR Family, Pyrin Domain-Containing 3 Protein, Signal Transduction, Carrier Proteins immunology, Colitis immunology, Colitis pathology, Epithelial Cells immunology
- Abstract
Decreased expression of the Nlrp3 protein is associated with susceptibility to Crohn's disease. However, the role of Nlrp3 in colitis has not been characterized. Nlrp3 interacts with the adaptor protein ASC to activate caspase-1 in inflammasomes, which are protein complexes responsible for the maturation and secretion of interleukin-1beta (IL-1beta) and IL-18. Here, we showed that mice deficient for Nlrp3 or ASC and caspase-1 were highly susceptible to dextran sodium sulfate (DSS)-induced colitis. Defective inflammasome activation led to loss of epithelial integrity, resulting in systemic dispersion of commensal bacteria, massive leukocyte infiltration, and increased chemokine production in the colon. This process was a consequence of a decrease in IL-18 in mice lacking components of the Nlrp3 inflammasome, resulting in higher mortality rates. Thus, the Nlrp3 inflammasome is critically involved in the maintenance of intestinal homeostasis and protection against colitis.
- Published
- 2010
- Full Text
- View/download PDF
39. ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS.
- Author
-
Alexander A, Cai SL, Kim J, Nanez A, Sahin M, MacLean KH, Inoki K, Guan KL, Shen J, Person MD, Kusewitt D, Mills GB, Kastan MB, and Walker CL
- Subjects
- Adenylate Kinase metabolism, Animals, Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins genetics, Cell Line, DNA-Binding Proteins genetics, Humans, Mechanistic Target of Rapamycin Complex 1, Mice, Mice, Transgenic, Multiprotein Complexes, Oxidative Stress, Phosphorylation, Protein Serine-Threonine Kinases genetics, Proteins, TOR Serine-Threonine Kinases, Tuberous Sclerosis Complex 2 Protein, Tumor Suppressor Proteins genetics, Cell Cycle Proteins metabolism, Cytoplasm metabolism, DNA-Binding Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Reactive Oxygen Species metabolism, Signal Transduction, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Ataxia-telangiectasia mutated (ATM) is a cellular damage sensor that coordinates the cell cycle with damage-response checkpoints and DNA repair to preserve genomic integrity. However, ATM also has been implicated in metabolic regulation, and ATM deficiency is associated with elevated reactive oxygen species (ROS). ROS has a central role in many physiological and pathophysiological processes including inflammation and chronic diseases such as atherosclerosis and cancer, underscoring the importance of cellular pathways involved in redox homeostasis. We have identified a cytoplasmic function for ATM that participates in the cellular damage response to ROS. We show that in response to elevated ROS, ATM activates the TSC2 tumor suppressor via the LKB1/AMPK metabolic pathway in the cytoplasm to repress mTORC1 and induce autophagy. Importantly, elevated ROS and dysregulation of mTORC1 in ATM-deficient cells is inhibited by rapamycin, which also rescues lymphomagenesis in Atm-deficient mice. Our results identify a cytoplasmic pathway for ROS-induced ATM activation of TSC2 to regulate mTORC1 signaling and autophagy, identifying an integration node for the cellular damage response with key pathways involved in metabolism, protein synthesis, and cell survival.
- Published
- 2010
- Full Text
- View/download PDF
40. Mdm2 regulates p53 mRNA translation through inhibitory interactions with ribosomal protein L26.
- Author
-
Ofir-Rosenfeld Y, Boggs K, Michael D, Kastan MB, and Oren M
- Subjects
- Animals, Cell Line, Feedback, Physiological, Humans, Mice, Models, Genetic, Proteasome Endopeptidase Complex metabolism, Ubiquitination, Gene Expression Regulation, Protein Biosynthesis, Proto-Oncogene Proteins c-mdm2 physiology, RNA, Messenger metabolism, Ribosomal Proteins metabolism, Tumor Suppressor Protein p53 genetics
- Abstract
Mdm2 regulates the p53 tumor suppressor by promoting its proteasome-mediated degradation. Mdm2 and p53 engage in an autoregulatory feedback loop that maintains low p53 activity in nonstressed cells. We now report that Mdm2 regulates p53 levels also by targeting ribosomal protein L26. L26 binds p53 mRNA and augments its translation. Mdm2 binds L26 and drives its polyubiquitylation and proteasomal degradation. In addition, the binding of Mdm2 to L26 attenuates the association of L26 with p53 mRNA and represses L26-mediated augmentation of p53 protein synthesis. Under nonstressed conditions, both mechanisms help maintain low cellular p53 levels by constitutively tuning down p53 translation. In response to genotoxic stress, the inhibitory effect of Mdm2 on L26 is attenuated, enabling a rapid increase in p53 synthesis. The Mdm2-L26 interaction thus represents an additional important component of the autoregulatory feedback loop that dictates cellular p53 levels and activity.
- Published
- 2008
- Full Text
- View/download PDF
41. Transient inhibition of ATM kinase is sufficient to enhance cellular sensitivity to ionizing radiation.
- Author
-
Rainey MD, Charlton ME, Stanton RV, and Kastan MB
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Cell Cycle drug effects, HeLa Cells, Humans, Infrared Rays, Mice, Phosphoinositide-3 Kinase Inhibitors, Proto-Oncogene Proteins c-abl antagonists & inhibitors, Quinazolines pharmacology, Radiation Tolerance drug effects, Signal Transduction, Triazoles pharmacology, Cell Cycle Proteins antagonists & inhibitors, DNA Damage, DNA, Neoplasm radiation effects, DNA-Binding Proteins antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Radiation Tolerance physiology, Tumor Suppressor Proteins antagonists & inhibitors
- Abstract
In response to DNA damage, the ATM protein kinase activates signal transduction pathways essential for coordinating cell cycle progression with DNA repair. In the human disease ataxia-telangiectasia, mutation of the ATM gene results in multiple cellular defects, including enhanced sensitivity to ionizing radiation (IR). This phenotype highlights ATM as a potential target for novel inhibitors that could be used to enhance tumor cell sensitivity to radiotherapy. A targeted compound library was screened for potential inhibitors of the ATM kinase, and CP466722 was identified. The compound is nontoxic and does not inhibit phosphatidylinositol 3-kinase (PI3K) or PI3K-like protein kinase family members in cells. CP466722 inhibited cellular ATM-dependent phosphorylation events and disruption of ATM function resulted in characteristic cell cycle checkpoint defects. Inhibition of cellular ATM kinase activity was rapidly and completely reversed by removing CP466722. Interestingly, clonogenic survival assays showed that transient inhibition of ATM is sufficient to sensitize cells to IR and suggests that therapeutic radiosensitization may only require ATM inhibition for short periods of time. The ability of CP466722 to rapidly and reversibly regulate ATM activity provides a new tool to ask questions about ATM function that could not easily be addressed using genetic models or RNA interference technologies.
- Published
- 2008
- Full Text
- View/download PDF
42. Human T-cell leukemia virus type 1 tax attenuates the ATM-mediated cellular DNA damage response.
- Author
-
Chandhasin C, Ducu RI, Berkovich E, Kastan MB, and Marriott SJ
- Subjects
- Adaptor Proteins, Signal Transducing, Ataxia Telangiectasia Mutated Proteins, Cell Line, DNA Replication, Humans, Nuclear Proteins metabolism, T-Lymphocytes radiation effects, T-Lymphocytes virology, Trans-Activators metabolism, Cell Cycle Proteins antagonists & inhibitors, DNA Repair physiology, DNA-Binding Proteins antagonists & inhibitors, Gene Products, tax metabolism, Human T-lymphotropic virus 1 physiology, Protein Serine-Threonine Kinases antagonists & inhibitors, Tumor Suppressor Proteins antagonists & inhibitors
- Abstract
Genomic instability, a hallmark of leukemic cells, is associated with malfunctioning cellular responses to DNA damage caused by defective cell cycle checkpoints and/or DNA repair. Adult T-cell leukemia, which can result from infection with human T-cell leukemia virus type 1 (HTLV-1), is associated with extensive genomic instability that has been attributed to the viral oncoprotein Tax. How Tax influences cellular responses to DNA damage to mediate genomic instability, however, remains unclear. Therefore, we investigated the effect of Tax on cellular pathways involved in recognition and repair of DNA double-strand breaks. Premature attenuation of ATM kinase activity and reduced association of MDC1 with repair foci were observed in Tax-expressing cells. Following ionizing radiation-induced S-phase checkpoint activation, Tax-expressing cells progressed more rapidly than non-Tax-expressing cells toward DNA replication. These results demonstrate that Tax expression may allow premature DNA replication in the presence of genomic lesions. Attempts to replicate in the presence of these lesions would result in gradual accumulation of mutations, leading to genome instability and cellular transformation.
- Published
- 2008
- Full Text
- View/download PDF
43. A novel ATM-dependent pathway regulates protein phosphatase 1 in response to DNA damage.
- Author
-
Tang X, Hui ZG, Cui XL, Garg R, Kastan MB, and Xu B
- Subjects
- Amino Acid Sequence, Animals, Ataxia Telangiectasia Mutated Proteins, Cell Cycle, Cell Cycle Proteins genetics, Cell Line, DNA-Binding Proteins genetics, Enzyme Activation, Humans, Molecular Sequence Data, Phosphorylation radiation effects, Protein Binding, Protein Phosphatase 1 genetics, Protein Serine-Threonine Kinases genetics, Sequence Alignment, Sequence Homology, Amino Acid, Tumor Suppressor Proteins genetics, Cell Cycle Proteins metabolism, DNA Damage genetics, DNA-Binding Proteins metabolism, Protein Phosphatase 1 metabolism, Protein Serine-Threonine Kinases metabolism, Signal Transduction, Tumor Suppressor Proteins metabolism
- Abstract
Protein phosphatase 1 (PP1), a major protein phosphatase important for a variety of cellular responses, is activated in response to ionizing irradiation (IR)-induced DNA damage. Here, we report that IR induces the rapid dissociation of PP1 from its regulatory subunit inhibitor-2 (I-2) and that the process requires ataxia-telangiectasia mutated (ATM), a protein kinase central to DNA damage responses. In response to IR, ATM phosphorylates I-2 on serine 43, leading to the dissociation of the PP1-I-2 complex and the activation of PP1. Furthermore, ATM-mediated I-2 phosphorylation results in the inhibition of the Aurora-B kinase, the down-regulation of histone H3 serine 10 phosphorylation, and the activation of the G(2)/M checkpoint. Collectively, the results of these studies demonstrate a novel pathway that links ATM, PP1, and I-2 in the cellular response to DNA damage.
- Published
- 2008
- Full Text
- View/download PDF
44. DNA damage responses: mechanisms and roles in human disease: 2007 G.H.A. Clowes Memorial Award Lecture.
- Author
-
Kastan MB
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Awards and Prizes, Cell Cycle Proteins metabolism, DNA-Binding Proteins metabolism, Humans, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Proteins metabolism, DNA Damage, Disease
- Abstract
Significant progress has been made in recent years in elucidating the molecular controls of cellular responses to DNA damage in mammalian cells. Much of our understanding of the mechanisms involved in cellular DNA damage response pathways has come from studies of human cancer susceptibility syndromes that are altered in DNA damage responses. Ataxia-telangiectasia mutated (ATM), the gene mutated in the disorder ataxia-telangiectasia, codes for a protein kinase that is a central mediator of responses to DNA double-strand breaks (DSB) in cells. Once activated, ATM phosphorylates numerous substrates in the cell that modulate the response of the cell to the DNA damage. We recently developed a novel system to create DNA DSBs at defined endogenous sites in the human genome and used this system to detect protein recruitment and loss at and around these breaks by chromatin immunoprecipitation. Results from this system showed the functional importance of ATM kinase activity and phosphorylation in the response to DSBs and supported a model in which ordered chromatin structure changes that occur after DNA breakage and that depend on functional NBS1 and ATM facilitate DNA DSB repair. Insights about these pathways provide us with opportunities to develop new approaches to benefit patients. Examples and opportunities for developing inhibitors that act as sensitizers to chemotherapy or radiation therapy or activators that could improve responses to cellular stresses, such as oxidative damage, are discussed. Relevant to the latter, we have shown benefits of an ATM activator in disease settings ranging from metabolic syndrome to cancer prevention.
- Published
- 2008
- Full Text
- View/download PDF
45. Assessment of protein dynamics and DNA repair following generation of DNA double-strand breaks at defined genomic sites.
- Author
-
Berkovich E, Monnat RJ Jr, and Kastan MB
- Subjects
- Chromatin Immunoprecipitation, DNA Repair genetics, Endodeoxyribonucleases genetics, Endodeoxyribonucleases metabolism, Genetic Vectors genetics, Humans, DNA Breaks, Double-Stranded, DNA Repair physiology, Genome, Human genetics, Molecular Probe Techniques
- Abstract
The formation of protein aggregates (foci) at sites of DNA double-strand breaks (DSBs) is mainly studied by immunostaining and is hence limited by the low resolution of light microscopy and the availability of appropriate and selective antibodies. Here, we describe a system using enzymatic creation of site-specific DNA DSBs within the human genome combined with chromatin immunoprecipitation (ChIP) that enables molecular probing of a DSB. Following induction of the I-PpoI enzyme and generation of DSBs, cellular DNA and proteins are crosslinked and analyzed by ChIP for specific proteins at the site of the break. The system allows the direct detection of protein and chromatin dynamics at the site of the break with high resolution, as well as direct measurement of DNA repair defects in human cells. Starting with fragmented chromatin, results can be achieved in 2-3 d.
- Published
- 2008
- Full Text
- View/download PDF
46. Targeting lysosomal degradation induces p53-dependent cell death and prevents cancer in mouse models of lymphomagenesis.
- Author
-
Maclean KH, Dorsey FC, Cleveland JL, and Kastan MB
- Subjects
- Animals, Antirheumatic Agents therapeutic use, Apoptosis genetics, Ataxia Telangiectasia genetics, Ataxia Telangiectasia metabolism, Ataxia Telangiectasia pathology, Ataxia Telangiectasia Mutated Proteins, Autophagy genetics, B-Lymphocytes metabolism, B-Lymphocytes pathology, Burkitt Lymphoma genetics, Burkitt Lymphoma metabolism, Burkitt Lymphoma pathology, Caspases genetics, Caspases metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Cells, Cultured, Chloroquine therapeutic use, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Embryo, Mammalian metabolism, Embryo, Mammalian pathology, Female, Fibroblasts metabolism, Fibroblasts pathology, Humans, Lysosomes pathology, Male, Mice, Mice, Mutant Strains, Neoplasms, Experimental genetics, Neoplasms, Experimental metabolism, Neoplasms, Experimental pathology, Neoplasms, Experimental prevention & control, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, bcl-2 Homologous Antagonist-Killer Protein genetics, bcl-2 Homologous Antagonist-Killer Protein metabolism, bcl-2-Associated X Protein genetics, bcl-2-Associated X Protein metabolism, Antirheumatic Agents pharmacology, Apoptosis drug effects, Ataxia Telangiectasia prevention & control, Autophagy drug effects, Burkitt Lymphoma prevention & control, Cell Transformation, Neoplastic metabolism, Chloroquine pharmacology, Lysosomes metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Despite great interest in cancer chemoprevention, effective agents are few. Here we show that chloroquine, a drug that activates the stress-responsive Atm-p53 tumor-suppressor pathway, preferentially enhances the death of Myc oncogene-overexpressing primary mouse B cells and mouse embryonic fibroblasts (MEFs) and impairs Myc-induced lymphomagenesis in a transgenic mouse model of human Burkitt lymphoma. Chloroquine-induced cell death in primary MEFs and human colorectal cancer cells was dependent upon p53, but not upon the p53 modulators Atm or Arf. Accordingly, chloroquine impaired spontaneous lymphoma development in Atm-deficient mice, a mouse model of ataxia telangiectasia, but not in p53-deficient mice. Chloroquine treatment enhanced markers of both macroautophagy and apoptosis in MEFs but ultimately impaired lysosomal protein degradation. Interestingly, chloroquine-induced cell death was not dependent on caspase-mediated apoptosis, as neither overexpression of the antiapoptotic protein Bcl-2 nor deletion of the proapoptotic Bax and Bak affected chloroquine-induced MEF death. However, when both apoptotic and autophagic pathways were blocked simultaneously, chloroquine-induced killing of Myc-overexpressing cells was blunted. Thus chloroquine induces lysosomal stress and provokes a p53-dependent cell death that does not require caspase-mediated apoptosis. These findings specifically demonstrate that intermittent chloroquine use effectively prevents cancer in mouse models of 2 genetically distinct human cancer syndromes, Burkitt lymphoma and ataxia telangiectasia, suggesting that agents targeting lysosome-mediated degradation may be effective in cancer prevention.
- Published
- 2008
- Full Text
- View/download PDF
47. Ataxia telangiectasia-mutated and p53 are potential mediators of chloroquine-induced resistance to mammary carcinogenesis.
- Author
-
Loehberg CR, Thompson T, Kastan MB, Maclean KH, Edwards DG, Kittrell FS, Medina D, Conneely OM, and O'Malley BW
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Breast cytology, Breast physiology, Cell Cycle Proteins physiology, DNA-Binding Proteins physiology, Drug Resistance, Neoplasm, Epithelial Cells cytology, Epithelial Cells physiology, Female, Humans, Mice, Mice, Inbred BALB C, Mice, Knockout, Protein Serine-Threonine Kinases physiology, Tumor Suppressor Proteins physiology, Cell Cycle Proteins genetics, Chloroquine pharmacology, DNA-Binding Proteins genetics, Genes, p53, Mammary Neoplasms, Animal genetics, Mammary Neoplasms, Animal prevention & control, Protein Serine-Threonine Kinases genetics, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins genetics
- Abstract
The use of agents to prevent the onset of and/or the progression to breast cancer has the potential to lower breast cancer risk. We have previously shown that the tumor-suppressor gene p53 is a potential mediator of hormone (estrogen/progesterone)-induced protection against chemical carcinogen-induced mammary carcinogenesis in animal models. Here, we show for the first time a breast cancer-protective effect of chloroquine in an animal model. Chloroquine significantly reduced the incidence of N-methyl-N-nitrosourea-induced mammary tumors in our animal model similar to estrogen/progesterone treatment. No protection was seen in our BALB/c p53-null mammary epithelium model, indicating a p53 dependency for the chloroquine effect. Using a human nontumorigenic mammary gland epithelial cell line, MCF10A, we confirm that in the absence of detectable DNA damage, chloroquine activates the tumor-suppressor p53 and the p53 downstream target gene p21, resulting in G(1) cell cycle arrest. p53 activation occurs at a posttranslational level via chloroquine-dependent phosphorylation of the checkpoint protein kinase, ataxia telangiectasia-mutated (ATM), leading to ATM-dependent phosphorylation of p53. In primary mammary gland epithelial cells isolated from p53-null mice, chloroquine does not induce G(1) cell cycle arrest compared with cells isolated from wild-type mice, also indicating a p53 dependency. Our results indicate that a short prior exposure to chloroquine may have a preventative application for mammary carcinogenesis.
- Published
- 2007
- Full Text
- View/download PDF
48. Our cells get stressed too! Implications for human disease.
- Author
-
Kastan MB
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Humans, Neoplasms etiology, Signal Transduction, Cell Cycle Proteins physiology, DNA Damage, DNA-Binding Proteins physiology, Disease Susceptibility, Protein Serine-Threonine Kinases physiology, Tumor Suppressor Proteins physiology
- Abstract
Significant progress has been made in recent years elucidating the molecular controls of cellular responses to DNA damage in mammalian cells. Many of the insights that we have gained into the mechanisms involved in cellular DNA damage response pathways have come from studies of human cancer susceptibility syndromes that are altered in DNA damage responses. ATM, the gene mutated in the cancer-prone disorder, ataxia telangiectasia, is a protein kinase that is a central mediator of responses to DNA double strand breaks in cells. Such insights provide us with opportunities to develop new approaches to benefit patients. For example, inhibitors of the ATM pathway have the potential to act as sensitizers to chemotherapy or radiation therapy and could even have anti-neoplastic effects on their own. Conversely, activators of ATM could improve responses to cellular stresses such as oxidative damage. The potential benefits of ATM modulation in disease settings ranging from metabolic syndrome to cancer will be discussed.
- Published
- 2007
- Full Text
- View/download PDF
49. Atm deficiency affects both apoptosis and proliferation to augment Myc-induced lymphomagenesis.
- Author
-
Maclean KH, Kastan MB, and Cleveland JL
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins, Cell Proliferation radiation effects, DNA Damage, Fibroblasts metabolism, Fibroblasts radiation effects, Humans, Mice, Phosphorylation radiation effects, Radiation, Ionizing, Signal Transduction radiation effects, Tumor Suppressor Protein p53 metabolism, Apoptosis radiation effects, DNA-Binding Proteins deficiency, Lymphoma metabolism, Lymphoma pathology, Protein Serine-Threonine Kinases deficiency, Proto-Oncogene Proteins c-myc metabolism, Tumor Suppressor Proteins deficiency
- Abstract
Myc oncoproteins are commonly activated in malignancies and are sufficient to provoke many types of cancer. However, the critical mechanisms by which Myc contributes to malignant transformation are not clear. DNA damage seems to be an important initiating event in tumorigenesis. Here, we show that although Myc does not directly induce double-stranded DNA breaks, it does augment activation of the Atm/p53 DNA damage response pathway, suggesting that Atm may function as a guardian against Myc-induced transformation. Indeed, we show that Atm loss augments Myc-induced lymphomagenesis and impairs Myc-induced apoptosis, which normally harnesses Myc-driven tumorigenesis. Surprisingly, Atm loss also augments the proliferative response induced by Myc, and this augmentation is associated with enhanced suppression of the expression of the cyclin-dependent kinase inhibitor p27(Kip1). Therefore, regulation of cell proliferation and p27(Kip1) seems to be a contributing mechanism by which Atm holds tumor formation in check.
- Published
- 2007
- Full Text
- View/download PDF
50. Roles of ATM and NBS1 in chromatin structure modulation and DNA double-strand break repair.
- Author
-
Berkovich E, Monnat RJ Jr, and Kastan MB
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins genetics, Cell Line, Tumor, Chromatin metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Humans, Nuclear Proteins genetics, Nucleosomes genetics, Nucleosomes metabolism, Phosphorylation, Protein Serine-Threonine Kinases genetics, Tumor Suppressor Proteins genetics, Cell Cycle Proteins physiology, Chromatin genetics, DNA genetics, DNA Breaks, Double-Stranded, DNA Repair genetics, DNA-Binding Proteins physiology, Nuclear Proteins physiology, Protein Serine-Threonine Kinases physiology, Tumor Suppressor Proteins physiology
- Abstract
We developed a novel system to create DNA double-strand breaks (DSBs) at defined endogenous sites in the human genome, and used this system to detect protein recruitment and loss at and around these breaks by chromatin immunoprecipitation (ChIP). The detection of human ATM protein at site-specific DSBs required functional NBS1 protein, ATM kinase activity and ATM autophosphorylation on Ser 1981. DSB formation led to the localized disruption of nucleosomes, a process that depended on both functional NBS1 and ATM. These two proteins were also required for efficient recruitment of the repair cofactor XRCC4 to DSBs, and for efficient DSB repair. These results demonstrate the functional importance of ATM kinase activity and phosphorylation in the response to DSBs, and support a model in which ordered chromatin structure changes that occur after DNA breakage depend on functional NBS1 and ATM, and facilitate DNA DSB repair.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.