Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sergei Kozlov is active.

Publication


Featured researches published by Sergei Kozlov.


Science | 2010

ATM Activation by Oxidative Stress

Zhi Guo; Sergei Kozlov; Martin F. Lavin; Maria D. Person; Tanya T. Paull

Stress, DNA Damage, and ATM The protein kinase ATM (ataxia-telangiectasia mutated) is a key component of the signaling pathway through which cells are protected from DNA damage. ATM becomes activated within a protein complex at sites of double-stranded breaks in DNA. ATM is also activated in response to increased production of reactive oxygen species (ROS). Such activation was thought to reflect DNA damage caused by ROS, but Guo et al. (p. 517) showed that ATM was in fact directly activated by ROS. A cysteine residue in ATM contributes to the formation of disulfide-linked dimers of activated ATM on exposure to ROS in vitro. Experiments using mutated forms of the enzyme suggested that two distinct mechanisms regulated ATM activity. The protein kinase ATM is a sensor for reactive oxygen species. The ataxia-telangiectasia mutated (ATM) protein kinase is activated by DNA double-strand breaks (DSBs) through the Mre11-Rad50-Nbs1 (MRN) DNA repair complex and orchestrates signaling cascades that initiate the DNA damage response. Cells lacking ATM are also hypersensitive to insults other than DSBs, particularly oxidative stress. We show that oxidation of ATM directly induces ATM activation in the absence of DNA DSBs and the MRN complex. The oxidized form of ATM is a disulfide–cross-linked dimer, and mutation of a critical cysteine residue involved in disulfide bond formation specifically blocked activation through the oxidation pathway. Identification of this pathway explains observations of ATM activation under conditions of oxidative stress and shows that ATM is an important sensor of reactive oxygen species in human cells.


Nature Genetics | 2000

ATM-dependent phosphorylation of nibrin in response to radiation exposure

Magtouf Gatei; David B. Young; Karen Cerosaletti; Ami Desai-Mehta; Kevin Spring; Sergei Kozlov; Martin F. Lavin; Richard A. Gatti; Patrick Concannon; Kum Kum Khanna

Mutations in the gene ATM are responsible for the genetic disorder ataxia-telangiectasia (A-T), which is characterized by cerebellar dysfunction, radiosensitivity, chromosomal instability and cancer predisposition. Both the A-T phenotype and the similarity of the ATM protein to other DNA-damage sensors suggests a role for ATM in biochemical pathways involved in the recognition, signalling and repair of DNA double-strand breaks (DSBs). There are strong parallels between the pattern of radiosensitivity, chromosomal instability and cancer predisposition in A-T patients and that in patients with Nijmegen breakage syndrome (NBS). The protein defective in NBS, nibrin (encoded by NBS1), forms a complex with MRE11 and RAD50 (refs 1,2). This complex localizes to DSBs within 30 minutes after cellular exposure to ionizing radiation (IR) and is observed in brightly staining nuclear foci after a longer period of time. The overlap between clinical and cellular phenotypes in A-T and NBS suggests that ATM and nibrin may function in the same biochemical pathway. Here we demonstrate that nibrin is phosphorylated within one hour of treatment of cells with IR. This response is abrogated in A-T cells that either do not express ATM protein or express near full-length mutant protein. We also show that ATM physically interacts with and phosphorylates nibrin on serine 343 both in vivo and in vitro. Phosphorylation of this site appears to be functionally important because mutated nibrin (S343A) does not completely complement radiosensitivity in NBS cells. ATM phosphorylation of nibrin does not affect nibrin-MRE11-RAD50 association as revealed by radiation-induced foci formation. Our data provide a biochemical explanation for the similarity in phenotype between A-T and NBS.


Nature Genetics | 1998

ATM associates with and phosphorylates p53: mapping the region of interaction

Kum Kum Khanna; Katherine Keating; Sergei Kozlov; Shaun P. Scott; Magtouf Gatei; Karen Hobson; Yoichi Taya; Brian Gabrielli; Doug W. Chan; Susan P. Lees-Miller; Martin F. Lavin

The human genetic disorder ataxia-telangiectasia (AT) is characterized by immunodeficiency, progressive cerebellar ataxia, radiosensitivity, cell cycle checkpoint defects and cancer predisposition. The gene mutated in this syndrome, ATM (for AT mutated), encodes a protein containing a phosphatidyl-inositol 3-kinase (PI-3 kinase)-like domain. ATM also contains a proline-rich region and a leucine zipper, both of which implicate this protein in signal transduction. The proline-rich region has been shown to bind to the SH3 domain of c-Abl, which facilitates its phosphorylation and activation by ATM (Refs 4,6). Previous results have demonstrated that AT cells are defective in the G1/S checkpoint activated after radiation damage and that this defect is attributable to a defective p53 signal transduction pathway. We report here direct interaction between ATM and p53 involving two regions in ATM, one at the amino terminus and the other at the carboxy terminus, corresponding to the PI-3 kinase domain. Recombinant ATM protein phosphorylates p53 on serine 15 near the N terminus. Furthermore, ectopic expression of ATM in AT cells restores normal ionizing radiation (IR)-induced phosphorylation of p53, whereas expression of ATM antisense RNA in control cells abrogates the rapid IR-induced phosphorylation of p53 on serine 15. These results demonstrate that ATM can bind p53 directly and is responsible for its serine 15 phosphorylation, thereby contributing to the activation and stabilization of p53 during the IR-induced DNA damage response.


Cell Cycle | 2007

ATM activation and DNA damage response.

Martin F. Lavin; Sergei Kozlov

Well before the gene (ATM) mutated in the human genetic disorder ataxia-telangiectasia (A-T) was described it was evident from the clinical, molecular and cellular phenotype of A-T that this gene would play a central role in the DNA damage response. Mutation of ATM causes defective cell cycle checkpoint activation,a reduced capacity for repair of DNA double strand breaks and abnormal apoptosis, all of which contribute to the major features of A-T including genome instability, increased cancer risk and neurodegeneration. While the exact mechanism of activation remains unknown, it is clear that the Mre11 complex plays an important role both in the recruitment of ATM to the sites of DNA damage and in the efficient activation of ATM. Although ATM responds to agents that produce double strand breaks in DNA, other stimuli are also capable of ATM activation. The description of autophosphorylation on S1981 of ATM and the ensuing transition from an inactive dimer to an active monomer represents a major milestone in our understanding of the activation process. However, it is now evident that more than one autophosphorylation event is required and not surprisingly this process is also attenuated by phosphatases and other modifications such as acetylation are also implicated. This is further complicated by a recent report that autophosphorylation at S1987 (the mouse site corresponding to S1981) is dispensable for Atm activation in an Atm mutant mouse model. Use of cell extracts and in vitro approaches in the reconstruction of activation complexes have shed further light on what it takes to activate ATM. The aim here is to examine the evidence for the involvement of these various steps in ATM activation and attempt to put together a comprehensive picture of the overall process and its significance to DNA damage signalling.


The EMBO Journal | 2006

Involvement of novel autophosphorylation sites in ATM activation.

Sergei Kozlov; Mark E. Graham; Cheng Peng; Philip Chen; Phillip J. Robinson; Martin F. Lavin

ATM kinase plays a central role in signaling DNA double‐strand breaks to cell cycle checkpoints and to the DNA repair machinery. Although the exact mechanism of ATM activation remains unknown, efficient activation requires the Mre11 complex, autophosphorylation on S1981 and the involvement of protein phosphatases and acetylases. We report here the identification of several additional phosphorylation sites on ATM in response to DNA damage, including autophosphorylation on pS367 and pS1893. ATM autophosphorylates all these sites in vitro in response to DNA damage. Antibodies against phosphoserine 1893 revealed rapid and persistent phosphorylation at this site after in vivo activation of ATM kinase by ionizing radiation, paralleling that observed for S1981 phosphorylation. Phosphorylation was dependent on functional ATM and on the Mre11 complex. All three autophosphorylation sites are physiologically important parts of the DNA damage response, as phosphorylation site mutants (S367A, S1893A and S1981A) were each defective in ATM signaling in vivo and each failed to correct radiosensitivity, genome instability and cell cycle checkpoint defects in ataxia‐telangiectasia cells. We conclude that there are at least three functionally important radiation‐induced autophosphorylation events in ATM.


Journal of Biological Chemistry | 2007

Ataxia Telangiectasia Mutated (ATM) Signaling Network Is Modulated by a Novel Poly(ADP-ribose)-dependent Pathway in the Early Response to DNA-damaging Agents

Jean François Haince; Sergei Kozlov; Valina L. Dawson; Ted M. Dawson; Michael J. Hendzel; Martin F. Lavin; Guy G. Poirier

Poly(ADP-ribosyl)ation is a post-translational modification that is instantly stimulated by DNA strand breaks creating a unique signal for the modulation of protein functions in DNA repair and cell cycle checkpoint pathways. Here we report that lack of poly(ADP-ribose) synthesis leads to a compromised response to DNA damage. Deficiency in poly(ADP-ribosyl)ation metabolism induces profound cellular sensitivity to DNA-damaging agents, particularly in cells deficient for the protein kinase ataxia telangiectasia mutated (ATM). At the biochemical level, we examined the significance of poly(ADP-ribose) synthesis on the regulation of early DNA damage-induced signaling cascade initiated by ATM. Using potent PARP inhibitors and PARP-1 knock-out cells, we demonstrate a functional interplay between ATM and poly(ADP-ribose) that is important for the phosphorylation of p53, SMC1, and H2AX. For the first time, we demonstrate a functional and physical interaction between the major DSB signaling kinase, ATM and poly(ADP-ribosyl)ation by PARP-1, a key enzyme of chromatin remodeling. This study suggests that poly(ADP-ribose) might serve as a DNA damage sensory molecule that is critical for early DNA damage signaling.


Oncogene | 1997

Cellular localisation of the ataxia-telangiectasia (ATM) gene product and discrimination between mutated and normal forms.

Dianne Watters; Kum Kum Khanna; Heather Beamish; Geoffrey Birrell; Kevin Spring; Padmini Kedar; Magtouf Gatei; Deborah Stenzel; Karen Hobson; Sergei Kozlov; Ning Zhang; Aine Farrell; Jonathan Ramsay; Richard A. Gatti; Martin F. Lavin

The recently cloned gene (ATM) mutated in the human genetic disorder ataxia-telangiectasia (A-T) is involved in DNA damage response at different cell cycle checkpoints and also appears to have a wider role in signal transduction. Antibodies prepared against peptides from the predicted protein sequence detected a ∼ 350 kDa protein corresponding to the open reading frame, which was absent in 13/23 A-T homozygotes. Subcellular fractionation, immunoelectronmicroscopy and immunofluorescence showed that the ATM protein is present in the nucleus and cytoplasmic vesicles. This distribution did not change after irradiation. We also provide evidence that ATM protein binds to p53 and this association is defective in A-T cells compatible with the defective p53 response in these cells. These results provide further support for a role for the ATM protein as a sensor of DNA damage and in a more general role in cell signalling, compatible with the broader phenotype of the syndrome.


Journal of Biological Chemistry | 2011

Autophosphorylation and ATM activation: Additional sites add to the complexity

Sergei Kozlov; Mark E. Graham; Burkhard Jakob; Frank Tobias; Amanda W. Kijas; Marcel Tanuji; Philip Chen; Phillip J. Robinson; Gisela Taucher-Scholz; Keiji Suzuki; Sairai So; David J. Chen; Martin F. Lavin

The recognition and signaling of DNA double strand breaks involves the participation of multiple proteins, including the protein kinase ATM (mutated in ataxia-telangiectasia). ATM kinase is activated in the vicinity of the break and is recruited to the break site by the Mre11-Rad50-Nbs1 complex, where it is fully activated. In human cells, the activation process involves autophosphorylation on three sites (Ser367, Ser1893, and Ser1981) and acetylation on Lys3016. We now describe the identification of a new ATM phosphorylation site, Thr(P)1885 and an additional autophosphorylation site, Ser(P)2996, that is highly DNA damage-inducible. We also confirm that human and murine ATM share five identical phosphorylation sites. We targeted the ATM phosphorylation sites, Ser367 and Ser2996, for further study by generating phosphospecific antibodies against these sites and demonstrated that phosphorylation of both was rapidly induced by radiation. These phosphorylations were abolished by a specific inhibitor of ATM and were dependent on ATM and the Mre11-Rad50-Nbs1 complex. As found for Ser(P)1981, ATM phosphorylated at Ser367 and Ser2996 localized to sites of DNA damage induced by radiation, but ATM recruitment was not dependent on phosphorylation at these sites. Phosphorylation at Ser367 and Ser2996 was functionally important because mutant forms of ATM were defective in correcting the S phase checkpoint defect and restoring radioresistance in ataxia-telangiectasia cells. These data provide further support for the importance of autophosphorylation in the activation and function of ATM in vivo.


Cellular and Molecular Life Sciences | 2011

ATM protein kinase: the linchpin of cellular defenses to stress

Shahzad Bhatti; Sergei Kozlov; Ammad Ahmad Farooqi; Ali Naqi; Martin F. Lavin; Kum Kum Khanna

ATM is the most significant molecule involved in monitoring the genomic integrity of the cell. Any damage done to DNA relentlessly challenges the cellular machinery involved in recognition, processing and repair of these insults. ATM kinase is activated early to detect and signal lesions in DNA, arrest the cell cycle, establish DNA repair signaling and faithfully restore the damaged chromatin. ATM activation plays an important role as a barrier to tumorigenesis, metabolic syndrome and neurodegeneration. Therefore, studies of ATM-dependent DNA damage signaling pathways hold promise for treatment of a variety of debilitating diseases through the development of new therapeutics capable of modulating cellular responses to stress. In this review, we have tried to untangle the complex web of ATM signaling pathways with the purpose of pinpointing multiple roles of ATM underlying the complex phenotypes observed in AT patients.


Stem Cells Translational Medicine | 2012

Induced Pluripotent Stem Cells from Ataxia-Telangiectasia Recapitulate the Cellular Phenotype

Sam P. Nayler; Magtouf Gatei; Sergei Kozlov; Richard A. Gatti; Jessica C. Mar; Christine A. Wells; Martin F. Lavin; Ernst J. Wolvetang

Pluripotent stem cells can differentiate into every cell type of the human body. Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) therefore provides an opportunity to gain insight into the molecular and cellular basis of disease. Because the cellular DNA damage response poses a barrier to reprogramming, generation of iPSCs from patients with chromosomal instability syndromes has thus far proven to be difficult. Here we demonstrate that fibroblasts from patients with ataxia‐telangiectasia (A‐T), a disorder characterized by chromosomal instability, progressive neurodegeneration, high risk of cancer, and immunodeficiency, can be reprogrammed to bona fide iPSCs, albeit at a reduced efficiency. A‐T iPSCs display defective radiation‐induced signaling, radiosensitivity, and cell cycle checkpoint defects. Bioinformatic analysis of gene expression in the A‐T iPSCs identifies abnormalities in DNA damage signaling pathways, as well as changes in mitochondrial and pentose phosphate pathways. A‐T iPSCs can be differentiated into functional neurons and thus represent a suitable model system to investigate A‐T‐associated neurodegeneration. Collectively, our data show that iPSCs can be generated from a chromosomal instability syndrome and that these cells can be used to discover early developmental consequences of ATM deficiency, such as altered mitochondrial function, that may be relevant to A‐T pathogenesis and amenable to therapeutic intervention.

Collaboration


Dive into the Sergei Kozlov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Magtouf Gatei

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Shaun P. Scott

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Nuri Gueven

University of Tasmania

View shared research outputs
Top Co-Authors

Avatar

Tara L. Roberts

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Yi Chieh Lim

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Amanda W. Kijas

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Kum Kum Khanna

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Philip Chen

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Dianne Watters

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge