1
|
Fujimaki K, Jambhekar A, Lahav G. DNA damage checkpoints balance a tradeoff between diploid- and polyploid-derived arrest failures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638318. [PMID: 39990415 PMCID: PMC11844538 DOI: 10.1101/2025.02.14.638318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The DNA damage checkpoint system ensures genomic integrity by preventing the division of damaged cells. This system operates primarily through theG 1 ∕ S andG 2 ∕ M checkpoints, which are susceptible to failure; how these checkpoints coordinate quantitatively to ensure optimal cellular outcomes remains unclear. In this study, we exposed non-cancerous human cells to exogenous DNA damage and used single-cell imaging to monitor spontaneous arrest failure. We discovered that cells fail to arrest in two major paths, resulting in two types with distinct characteristics, including ploidy, nuclear morphology, and micronuclei composition. Computational simulations and experiments revealed strengthening one checkpoint reduced one mode of arrest failure but increased the other, leading to a critical tradeoff for optimizing total arrest failure rates. Our findings suggest optimal checkpoint strengths for minimizing total error are inherently suboptimal for any single failure type, elucidating the systemic cause of genomic instability and tetraploid-like cells in response to DNA damage.
Collapse
Affiliation(s)
- Kotaro Fujimaki
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
- Ludwig Center at Harvard Medical School
| | - Galit Lahav
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
- Ludwig Center at Harvard Medical School
| |
Collapse
|
2
|
Malik MZ, Dashti M, Jangid A, Channanath A, Elsa John S, Singh RKB, Al-Mulla F, Alphonse Thanaraj T. Complex p53 dynamics regulated by miR-125b in cellular responses to reactive oxidative stress and DNA damage. Brief Bioinform 2024; 26:bbae706. [PMID: 39820247 PMCID: PMC11736902 DOI: 10.1093/bib/bbae706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/27/2024] [Accepted: 12/28/2024] [Indexed: 01/19/2025] Open
Abstract
In response to distinct cellular stresses, the p53 exhibits distinct dynamics. These p53 dynamics subsequently control cell fate. However, different stresses can generate the same p53 dynamics with different cell fate outcomes, suggesting that the integration of dynamic information from other pathways is important for cell fate regulation. The interactions between miRNA-125b, p53, and reactive oxygen species (ROS) are significant in the context of cellular stress responses and apoptosis. However, the regulating mechanism of miR-125b with p53 is not fully studied. The dynamics of p53 and its response to the miR-125b regulation are still open questions. In the present study, we try to answer some of these fundamental questions based on basic model built from available experimental reports. The miR-125b-p53 regulatory network is modeled using a set of 11 molecular species variables. The biochemical network of miR-125b-p53, described by 22 reaction channels, is represented by coupled ordinary differential equations (ODEs) using the mass action law of chemical kinetics. These ODEs are solved numerically using the standard fourth-order Runge-Kutta method to analyze the dynamical behavior of the system. The biochemical network model we designed is based on both experimental and theoretical reported data. The p53 dynamics driven by miR-125b exhibit five distinct dynamical states: first and second stable states, first and second dynamical states, and a sustained oscillation state. These different p53 dynamical states may correspond to various cellular conditions. If the stress induced by miR-125b is weak, the system will be weakly activated, favoring a return to normal functioning. However, if the stress is significantly strong, the system will move to an active state. To sustain this active state, which is far from equilibrium with little scope for returning to normal conditions, the system may transition to an apoptotic state by crossing through other intermediate states, as it is unlikely to regain normal functioning. The p53 dynamical states show a multifractal nature, contributed by both short- and long-range correlations. The networks illustrated from these dynamical states follow hierarchical scale-free features, exhibiting an assortative nature with an absence of the centrality-lethality rule. Furthermore, the active dynamical state is generally closer to hierarchical characteristics and is self-organized. Our research study reveals that significant activity of miR-125b on the p53 regulatory network and its dynamics can only be observed when the system is slightly activated by ROS. However, this process does not necessarily require the direct study of ROS activity. These findings elucidate the mechanisms by which cells integrate signaling pathways with distinct temporal activity patterns to encode stress specificity and direct diverse cell fate decisions.
Collapse
Affiliation(s)
- Md Zubbair Malik
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait City, Kuwait
| | - Mohammed Dashti
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait City, Kuwait
| | - Amit Jangid
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Arshad Channanath
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait City, Kuwait
| | - Sumi Elsa John
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait City, Kuwait
| | - R K Brojen Singh
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Fahd Al-Mulla
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait City, Kuwait
| | | |
Collapse
|
3
|
Rizzotto D, Vigorito V, Rieder P, Gallob F, Moretta GM, Soratroi C, Riley JS, Bellutti F, Veli SL, Mattivi A, Lohmüller M, Herzog S, Bornhauser BC, Jacotot ED, Villunger A, Fava LL. Caspase-2 kills cells with extra centrosomes. SCIENCE ADVANCES 2024; 10:eado6607. [PMID: 39475598 PMCID: PMC11524169 DOI: 10.1126/sciadv.ado6607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024]
Abstract
Centrosomes are membrane-less organelles that orchestrate a wide array of biological functions by acting as microtubule organizing centers. Here, we report that caspase-2-driven apoptosis is elicited in blood cells failing cytokinesis and that extra centrosomes are necessary to trigger this cell death. Activation of caspase-2 depends on the PIDDosome multi-protein complex, and priming of PIDD1 at extra centrosomes is necessary for pathway activation. Accordingly, loss of its centrosomal adapter, ANKRD26, allows for cell survival and unrestricted polyploidization in response to cytokinesis failure. Mechanistically, cell death is initiated upstream of mitochondria via caspase-2-mediated processing of the BCL2 family protein BID, driving BAX/BAK-dependent mitochondrial outer membrane permeabilization (MOMP). Remarkably, BID-deficient cells enforce apoptosis by engaging p53-dependent proapoptotic transcriptional responses initiated by caspase-2. Consistently, BID and MDM2 act as shared caspase-2 substrates, with BID being kinetically favored. Our findings document that the centrosome limits its own unscheduled duplication by the induction of PIDDosome-driven mitochondrial apoptosis to avoid potentially pathogenic polyploidization events.
Collapse
Affiliation(s)
- Dario Rizzotto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Vincenza Vigorito
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Patricia Rieder
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Filip Gallob
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Gian Mario Moretta
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Claudia Soratroi
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Joel S. Riley
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Florian Bellutti
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Stefano Li Veli
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Alessia Mattivi
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Michael Lohmüller
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Sebastian Herzog
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Beat C. Bornhauser
- Department of Oncology and Children’s Research Centre, University Children’s Hospital Zürich, 8032 Zürich, Switzerland
| | - Etienne D. Jacotot
- Inserm U1268, Medicinal Chemistry and Translational Research, Paris F-75006, France
- Faculté de Pharmacie, UMR 8038 CiTCoM, Université Paris Cité, Paris F-75006, France
| | - Andreas Villunger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Luca L. Fava
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| |
Collapse
|
4
|
García García OR, Ortiz R, Moreno-Barbosa E, D-Kondo N, Faddegon B, Ramos-Méndez J. TOPAS-Tissue: A Framework for the Simulation of the Biological Response to Ionizing Radiation at the Multi-Cellular Level. Int J Mol Sci 2024; 25:10061. [PMID: 39337547 PMCID: PMC11431975 DOI: 10.3390/ijms251810061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/21/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
This work aims to develop and validate a framework for the multiscale simulation of the biological response to ionizing radiation in a population of cells forming a tissue. We present TOPAS-Tissue, a framework to allow coupling two Monte Carlo (MC) codes: TOPAS with the TOPAS-nBio extension, capable of handling the track-structure simulation and subsequent chemistry, and CompuCell3D, an agent-based model simulator for biological and environmental behavior of a population of cells. We verified the implementation by simulating the experimental conditions for a clonogenic survival assay of a 2-D PC-3 cell culture model (10 cells in 10,000 µm2) irradiated by MV X-rays at several absorbed dose values from 0-8 Gy. The simulation considered cell growth and division, irradiation, DSB induction, DNA repair, and cellular response. The survival was obtained by counting the number of colonies, defined as a surviving primary (or seeded) cell with progeny, at 2.7 simulated days after irradiation. DNA repair was simulated with an MC implementation of the two-lesion kinetic model and the cell response with a p53 protein-pulse model. The simulated survival curve followed the theoretical linear-quadratic response with dose. The fitted coefficients α = 0.280 ± 0.025/Gy and β = 0.042 ± 0.006/Gy2 agreed with published experimental data within two standard deviations. TOPAS-Tissue extends previous works by simulating in an end-to-end way the effects of radiation in a cell population, from irradiation and DNA damage leading to the cell fate. In conclusion, TOPAS-Tissue offers an extensible all-in-one simulation framework that successfully couples Compucell3D and TOPAS for multiscale simulation of the biological response to radiation.
Collapse
Affiliation(s)
- Omar Rodrigo García García
- Facultad de Ciencias Físico Matemáticas, Benemérita Universidad Autónoma de Puebla, Puebla 72000, Mexico; (O.R.G.G.); (E.M.-B.)
| | - Ramon Ortiz
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA 94115, USA; (R.O.); (N.D.-K.); (B.F.)
| | - Eduardo Moreno-Barbosa
- Facultad de Ciencias Físico Matemáticas, Benemérita Universidad Autónoma de Puebla, Puebla 72000, Mexico; (O.R.G.G.); (E.M.-B.)
| | - Naoki D-Kondo
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA 94115, USA; (R.O.); (N.D.-K.); (B.F.)
| | - Bruce Faddegon
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA 94115, USA; (R.O.); (N.D.-K.); (B.F.)
| | - Jose Ramos-Méndez
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA 94115, USA; (R.O.); (N.D.-K.); (B.F.)
| |
Collapse
|
5
|
Fu RZ, Cottrell O, Cutillo L, Rowntree A, Zador Z, Wurdak H, Papalopulu N, Marinopoulou E. Identification of genes with oscillatory expression in glioblastoma: the paradigm of SOX2. Sci Rep 2024; 14:2123. [PMID: 38267500 PMCID: PMC10808450 DOI: 10.1038/s41598-024-51340-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
Quiescence, a reversible state of cell-cycle arrest, is an important state during both normal development and cancer progression. For example, in glioblastoma (GBM) quiescent glioblastoma stem cells (GSCs) play an important role in re-establishing the tumour, leading to relapse. While most studies have focused on identifying differentially expressed genes between proliferative and quiescent cells as potential drivers of this transition, recent studies have shown the importance of protein oscillations in controlling the exit from quiescence of neural stem cells. Here, we have undertaken a genome-wide bioinformatic inference approach to identify genes whose expression oscillates and which may be good candidates for controlling the transition to and from the quiescent cell state in GBM. Our analysis identified, among others, a list of important transcription regulators as potential oscillators, including the stemness gene SOX2, which we verified to oscillate in quiescent GSCs. These findings expand on the way we think about gene regulation and introduce new candidate genes as key regulators of quiescence.
Collapse
Affiliation(s)
- Richard Zhiming Fu
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, University of Manchester, Manchester, M13 9PL, UK
- Department of Neurosurgery, Manchester Centre for Clinical Neurosciences, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford Royal, Stott Lane, Salford, M6 8HD, UK
| | - Oliver Cottrell
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Luisa Cutillo
- School of Mathematics, University of Leeds, Woodhouse, Leeds, LS2 9JT, UK
| | - Andrew Rowntree
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Zsolt Zador
- Division of Neurosurgery, Department of Surgery, St. Michael's Hospital, 36 Queen St E, Toronto, ON, M5B 1W8, Canada
- Department of Surgery, McMaster University, 1280 Mains St W, Hamilton, ON, L8S 4L8, Canada
- Center for Discovery in Cancer Research (CDCR), McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Heiko Wurdak
- Stem Cell and Brain Tumour Group, Leeds Institute of Medical Research at St James's, School of Medicine, University of Leeds, Leeds, LS9 7TF, UK
| | - Nancy Papalopulu
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| | - Elli Marinopoulou
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
6
|
Wang Y, Hu S, Zhang W, Zhang B, Yang Z. Emerging role and therapeutic implications of p53 in intervertebral disc degeneration. Cell Death Discov 2023; 9:433. [PMID: 38040675 PMCID: PMC10692240 DOI: 10.1038/s41420-023-01730-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023] Open
Abstract
Lower back pain (LBP) is a common degenerative musculoskeletal disease that imposes a huge economic burden on both individuals and society. With the aggravation of social aging, the incidence of LBP has increased globally. Intervertebral disc degeneration (IDD) is the primary cause of LBP. Currently, IDD treatment strategies include physiotherapy, medication, and surgery; however, none can address the root cause by ending the degeneration of intervertebral discs (IVDs). However, in recent years, targeted therapy based on specific molecules has brought hope for treating IDD. The tumor suppressor gene p53 produces a transcription factor that regulates cell metabolism and survival. Recently, p53 was shown to play an important role in maintaining IVD microenvironment homeostasis by regulating IVD cell senescence, apoptosis, and metabolism by activating downstream target genes. This study reviews research progress regarding the potential role of p53 in IDD and discusses the challenges of targeting p53 in the treatment of IDD. This review will help to elucidate the pathogenesis of IDD and provide insights for the future development of precision treatments.
Collapse
Affiliation(s)
- Yidian Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Shouye Hu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Weisong Zhang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Binfei Zhang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhi Yang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Gutu N, Binish N, Keilholz U, Herzel H, Granada AE. p53 and p21 dynamics encode single-cell DNA damage levels, fine-tuning proliferation and shaping population heterogeneity. Commun Biol 2023; 6:1196. [PMID: 38001355 PMCID: PMC10673849 DOI: 10.1038/s42003-023-05585-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Cells must accurately and quickly detect DNA damage through a set of checkpoint mechanisms that enable repair and control proliferation. Heterogeneous levels of cellular stress and noisy signaling processes can lead to phenotypic variability but little is known about their role in underlying proliferation heterogeneity. Here we study two previously published single cell datasets and find that cells encode heterogeneous levels of endogenous and exogenous DNA damage to shape proliferation heterogeneity at the population level. Using a comprehensive time series analysis of short- and long-term signaling dynamics of p53 and p21, we show that DNA damage levels are quantitatively translated into p53 and p21 signal parameters in a gradual manner. Analyzing instantaneous proliferation and signaling differences among equally-radiated cells, we identify time-localized changes in the period of p53 pulses that drive cells out of a low proliferative state. Our findings suggest a novel role of the p53-p21 network in quantitatively encoding DNA damage strength and fine-tuning proliferation trajectories.
Collapse
Affiliation(s)
- Nica Gutu
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany
- Humboldt-Universität zu Berlin, Berlin, Germany
| | - Neha Binish
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany
- Hertie Institute for Clinical Brain Research, Center for Neurology, University Medical Center Tübingen, Tübingen, Germany
| | - Ulrich Keilholz
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany
- German Cancer Consortium, Deutschen Konsortiums für Translationale Krebsforschung (DKTK), Berlin, Germany
| | - Hanspeter Herzel
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany
- Humboldt-Universität zu Berlin, Berlin, Germany
- Institute for Theoretical Biology, Berlin, Germany
| | - Adrián E Granada
- Charité Universitätsmedizin, Charité Comprehensive Cancer Center, Berlin, Germany.
- German Cancer Consortium, Deutschen Konsortiums für Translationale Krebsforschung (DKTK), Berlin, Germany.
| |
Collapse
|
8
|
Venkatachalapathy H, Yang Z, Azarin SM, Sarkar CA, Batchelor E. Pulsed stimuli entrain p53 to synchronize single cells and modulate cell-fate determination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563786. [PMID: 37961090 PMCID: PMC10634792 DOI: 10.1101/2023.10.24.563786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Entrainment to an external stimulus enables a synchronized oscillatory response across a population of cells, increasing coherent responses by reducing cell-to-cell heterogeneity. It is unclear whether the property of entrainability extends to systems where responses are intrinsic to the individual cell, rather than dependent on coherence across a population of cells. Using a combination of mathematical modeling, time-lapse fluorescence microscopy, and single-cell tracking, we demonstrated that p53 oscillations triggered by DNA double-strand breaks (DSBs) can be entrained with a periodic damage stimulus, despite such synchrony not known to function in effective DNA damage responses. Surprisingly, p53 oscillations were experimentally entrained over a wider range of DSB frequencies than predicted by an established computational model for the system. We determined that recapitulating the increased range of entrainment frequencies required, non-intuitively, a less robust oscillator and wider steady-state valley on the energy landscape. Further, we show that p53 entrainment can lead to altered expression dynamics of downstream targets responsible for cell fate in a manner dependent on target mRNA stability. Overall, this study demonstrates that entrainment can occur in a biological oscillator despite the apparent lack of an evolutionary advantage conferred through synchronized responses and highlights the potential of externally entraining p53 dynamics to reduce cellular variability and synchronize cell-fate responses for therapeutic outcomes.
Collapse
|
9
|
Peng Q, Shi X, Li D, Guo J, Zhang X, Zhang X, Chen Q. SCML2 contributes to tumor cell resistance to DNA damage through regulating p53 and CHK1 stability. Cell Death Differ 2023; 30:1849-1867. [PMID: 37353627 PMCID: PMC10307790 DOI: 10.1038/s41418-023-01184-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 05/20/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023] Open
Abstract
SCML2 has been found to be highly expressed in various tumors. However, the extent to which SCML2 is involved in tumorigenesis and cancer therapy is yet to be fully understood. In this study, we aimed to investigate the relationship between SCML2 and DNA damage response (DDR). Firstly, DNA damage stabilizes SCML2 through CHK1-mediated phosphorylation at Ser570. Functionally, this increased stability of SCML2 enhances resistance to DNA damage agents in p53-positive, p53-mutant, and p53-negative cells. Notably, SCML2 promotes chemoresistance through distinct mechanisms in p53-positive and p53-negative cancer cells. SCML2 binds to the TRAF domain of USP7, and Ser441 is a critical residue for their interaction. In p53-positive cancer cells, SCML2 competes with p53 for USP7 binding and destabilizes p53, which prevents DNA damage-induced p53 overactivation and increases chemoresistance. In p53-mutant or p53-negative cancer cells, SCML2 promotes CHK1 and p21 stability by inhibiting their ubiquitination, thereby enhancing the resistance to DNA damage agents. Interestingly, we found that SCML2A primarily stabilizes CHK1, while SCML2B regulates the stability of p21. Therefore, we have identified SCML2 as a novel regulator of chemotherapy resistance and uncovered a positive feedback loop between SCML2 and CHK1 after DNA damage, which serves to promote the chemoresistance to DNA damage agents.
Collapse
Affiliation(s)
- Qianqian Peng
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, PR China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, PR China
| | - Xin Shi
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, PR China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, PR China
| | - Dingwei Li
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, PR China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, PR China
| | - Jing Guo
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, PR China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, PR China
| | - Xiaqing Zhang
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, PR China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, PR China
| | - Xiaoyan Zhang
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, PR China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, PR China
| | - Qiang Chen
- Department of Radiation and Medical Oncology, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, PR China.
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, PR China.
| |
Collapse
|
10
|
Anwar F, Al-Abbasi FA, Naqvi S, Sheikh RA, Alhayyani S, Asseri AH, Asar TO, Kumar V. Therapeutic Potential of Nanomedicine in Management of Alzheimer's Disease and Glioma. Int J Nanomedicine 2023; 18:2737-2756. [PMID: 37250469 PMCID: PMC10211371 DOI: 10.2147/ijn.s405454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
Neoplasm (Glioblastoma) and Alzheimer's disease (AD) comprise two of the most chronic psychological ailments. Glioblastoma is one of the aggressive and prevalent malignant diseases characterized by rapid growth and invasion resulting from cell migration and degradation of extracellular matrix. While the latter is characterized by extracellular plaques of amyloid and intracellular tangles of tau proteins. Both possess a high degree of resistance to treatment owing to the restricted transport of corresponding drugs to the brain protected by the blood-brain barrier (BBB). Development of optimized therapies using advanced technologies is a great need of today. One such approach is the designing of nanoparticles (NPs) to facilitate the drug delivery at the target site. The present article elaborates the advances in nanomedicines in treatment of both AD as well as Gliomas. The intention of this review is to provide an overview of different types of NPs with their physical properties emphasizing their importance in traversing the BBB and hitting the target site. Further, we discuss the therapeutic applications of these NPs along with their specific targets. Multiple overlapping factors with a common pathway in development of AD and Glioblastoma are discussed in details that will assist the readers in developing the conceptual approach to target the NP for an aging population in the given circumstances with limitations of currently designed NPs, and the challenges to meet and the future perspectives.
Collapse
Affiliation(s)
- Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salma Naqvi
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Ryan Adnan Sheikh
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sultan Alhayyani
- Department of Chemistry, College of Sciences & Arts, Rabigh King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amer H Asseri
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turky Omar Asar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Vikas Kumar
- Natural Product Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health and Allied Sciences, SHUATS, Prayagraj, India
| |
Collapse
|
11
|
Burnaevskiy N, Oshima J, Mendenhall AR. Rapid emergence of transcriptional heterogeneity upon molecular stress predisposes cells to two distinct states of senescence. GeroScience 2023; 45:1115-1130. [PMID: 36562924 PMCID: PMC9886721 DOI: 10.1007/s11357-022-00709-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022] Open
Abstract
Slowing aging can reduce the risk of chronic diseases. In particular, eliminating senescent cells is a promising approach to slow aging. Previous studies found that both cells from older animals and senescent cells have noisy gene expression. Here, we performed a large-scale single-cell RNA-sequencing time course to understand how transcriptional heterogeneity develops among senescent cells. We found that cells experiencing senescence-inducing oxidative stress rapidly adopt one of two major transcriptional states. One senescent cell state is associated with stress response, and the other is associated with tissue remodeling. We did not observe increased stochastic gene expression. This data is consistent with the idea that reproducible, limited, distinct, and coherent transcriptional states exist in senescent cell populations. These physiologically distinct senescent cell subtypes may each affect the aging process in unique ways and constitute a source of heterogeneity in aging.
Collapse
Affiliation(s)
- Nikolay Burnaevskiy
- Department of Pathology, University of Washington, Seattle, WA, USA
- Present Address: Altius Institute for Biomedical Sciences, Seattle, WA, USA
| | - Junko Oshima
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
12
|
Tran AP, Tralie CJ, Reyes J, Moosmüller C, Belkhatir Z, Kevrekidis IG, Levine AJ, Deasy JO, Tannenbaum AR. Long-term p21 and p53 dynamics regulate the frequency of mitosis events and cell cycle arrest following radiation damage. Cell Death Differ 2023; 30:660-672. [PMID: 36182991 PMCID: PMC9984379 DOI: 10.1038/s41418-022-01069-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/07/2022] Open
Abstract
Radiation exposure of healthy cells can halt cell cycle temporarily or permanently. In this work, we analyze the time evolution of p21 and p53 from two single cell datasets of retinal pigment epithelial cells exposed to several levels of radiation, and in particular, the effect of radiation on cell cycle arrest. Employing various quantification methods from signal processing, we show how p21 levels, and to a lesser extent p53 levels, dictate whether the cells are arrested in their cell cycle and how frequently these mitosis events are likely to occur. We observed that single cells exposed to the same dose of DNA damage exhibit heterogeneity in cellular outcomes and that the frequency of cell division is a more accurate monitor of cell damage rather than just radiation level. Finally, we show how heterogeneity in DNA damage signaling is manifested early in the response to radiation exposure level and has potential to predict long-term fate.
Collapse
Affiliation(s)
- Anh Phong Tran
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher J Tralie
- Department of Mathematics and Computer Science, Ursinus College, Collegeville, PA, USA
| | - José Reyes
- Cancer Biology and Genetics Program and Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Caroline Moosmüller
- Department of Mathematics, University of California, San Diego, La Jolla, CA, USA
| | - Zehor Belkhatir
- School of Engineering and Sustainable Development, De Montfort University, Leicester, UK
| | - Ioannis G Kevrekidis
- Department of Chemical and Biological Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Arnold J Levine
- Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allen R Tannenbaum
- Departments of Computer Science and Applied Mathematics & Statistics, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
13
|
Bock FJ, Riley JS. When cell death goes wrong: inflammatory outcomes of failed apoptosis and mitotic cell death. Cell Death Differ 2023; 30:293-303. [PMID: 36376381 PMCID: PMC9661468 DOI: 10.1038/s41418-022-01082-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Apoptosis is a regulated cellular pathway that ensures that a cell dies in a structured fashion to prevent negative consequences for the tissue or the organism. Dysfunctional apoptosis is a hallmark of numerous pathologies, and treatments for various diseases are successful based on the induction of apoptosis. Under homeostatic conditions, apoptosis is a non-inflammatory event, as the activation of caspases ensures that inflammatory pathways are disabled. However, there is an increasing understanding that under specific conditions, such as caspase inhibition, apoptosis and the apoptotic machinery can be re-wired into a process which is inflammatory. In this review we discuss how the death receptor and mitochondrial pathways of apoptosis can activate inflammation. Furthermore, we will highlight how cell death due to mitotic stress might be a special case when it comes to cell death and the induction of inflammation.
Collapse
Affiliation(s)
- Florian J Bock
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Joel S Riley
- Institute of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
14
|
Krassnig SC, Mäser M, Probst NA, Werner J, Schlett C, Schumann N, von Scheven G, Mangerich A, Bürkle A. Comparative analysis of chlorambucil-induced DNA lesion formation and repair in a spectrum of different human cell systems. Toxicol Rep 2023; 10:171-189. [PMID: 36714466 PMCID: PMC9881385 DOI: 10.1016/j.toxrep.2023.01.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023] Open
Abstract
Chlorambucil (CLB) belongs to the class of nitrogen mustards (NMs), which are highly reactive bifunctional alkylating agents and were the first chemotherapeutic agents developed. They form DNA interstrand crosslinks (ICLs), which cause a blockage of DNA strand separation, inhibiting essential processes in DNA metabolism like replication and transcription. In fast replicating cells, e.g., tumor cells, this can induce cell death. The upregulation of ICL repair is thought to be a key factor for the resistance of tumor cells to ICL-inducing cytostatic agents including NMs. To monitor induction and repair of CLB-induced ICLs, we adjusted the automated reversed fluorometric analysis of alkaline DNA unwinding assay (rFADU) for the detection of ICLs in adherent cells. For the detection of monoalkylated DNA bases we established an LC-MS/MS method. We performed a comparative analysis of adduct formation and removal in five human cell lines and in peripheral blood mononuclear cells (PBMCs) after treatment with CLB. Dose-dependent increases in adduct formation were observed, and suitable treatment concentrations were identified for each cell line, which were then used for monitoring the kinetics of adduct formation. We observed significant differences in the repair kinetics of the cell lines tested. For example, in A2780 cells, hTERT immortalized VH10 cells, and in PBMCs a time-dependent repair of the two main monoalkylated DNA-adducts was confirmed. Regarding ICLs, repair was observed in all cell systems except for PBMCs. In conclusion, LC-MS/MS analyses combined with the rFADU technique are powerful tools to study the molecular mechanisms of NM-induced DNA damage and repair. By applying these methods to a spectrum of human cell systems of different origin and transformation status, we obtained insight into the cell-type specific repair of different CLB-induced DNA lesions, which may help identify novel resistance mechanisms of tumors and define molecular targets for therapeutic interventions.
Collapse
Key Words
- BER, base excision repair
- CLB, chlorambucil
- Chlorambucil
- DNA repair kinetics
- ICL, interstrand crosslink
- Interstrand crosslink
- MS, mass spectrometry
- Mass spectrometry
- Monoalkylated DNA adducts
- NER, nucleotide excision repair
- NM, Nitrogen mustard
- Nitrogen mustard
- PBMCs, peripheral blood mononuclear cells
- PI, propidium iodide
- RPE-1, human retinal pigment epithelial
- SD, standard deviation
- VH10, human foreskin fibroblasts
- dG, 2'-deoxyguanosine
- hTERT, human telomerase reverse transcriptase
- rFADU, reverse fluorometric analysis of alkaline DNA unwinding
Collapse
Affiliation(s)
- Sarah Ceylan Krassnig
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Marina Mäser
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Nicola Anna Probst
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Jens Werner
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Charlotte Schlett
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Nina Schumann
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Gudrun von Scheven
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| | - Aswin Mangerich
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
- Nutritional Toxicology, Institute of Nutritional Science, University of Potsdam, D-14558 Nuthetal, Germany
| | - Alexander Bürkle
- Molecular Toxicology, Department of Biology, University of Konstanz, D-78464 Konstanz, Germany
| |
Collapse
|
15
|
Xiong L, Garfinkel A. A common pathway to cancer: Oncogenic mutations abolish p53 oscillations. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 174:28-40. [PMID: 35752348 DOI: 10.1016/j.pbiomolbio.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
The tumor suppressor p53 oscillates in response to DNA double-strand breaks, a behavior that has been suggested to be essential to its anti-cancer function. Nearly all human cancers have genetic alterations in the p53 pathway; a number of these alterations have been shown to be oncogenic by experiment. These alterations include somatic mutations and copy number variations as well as germline polymorphisms. Intriguingly, they exhibit a mixed pattern of interactions in tumors, such as co-occurrence, mutual exclusivity, and paradoxically, mutual antagonism. Using a differential equation model of p53-Mdm2 dynamics, we employ Hopf bifurcation analysis to show that these alterations have a common mode of action, to abolish the oscillatory competence of p53, thereby, we suggest, impairing its tumor suppressive function. In this analysis, diverse genetic alterations, widely associated with human cancers clinically, have a unified mechanistic explanation of their role in oncogenesis.
Collapse
Affiliation(s)
- Lingyun Xiong
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90007 USA; Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90007, USA; Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, UK
| | - Alan Garfinkel
- Departments of Medicine (Cardiology) and Integrative Biology and Physiology, University of California, Los Angeles, CA, 90095, USA; Newton-Abraham Visiting Professor (2019-2020), Lincoln College and Department of Computer Science, University of Oxford, Oxford, OX1 3DR, UK.
| |
Collapse
|
16
|
Langlois-Lemay L, D’Amours D. Moonlighting at the Poles: Non-Canonical Functions of Centrosomes. Front Cell Dev Biol 2022; 10:930355. [PMID: 35912107 PMCID: PMC9329689 DOI: 10.3389/fcell.2022.930355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Centrosomes are best known as the microtubule organizing centers (MTOCs) of eukaryotic cells. In addition to their classic role in chromosome segregation, centrosomes play diverse roles unrelated to their MTOC activity during cell proliferation and quiescence. Metazoan centrosomes and their functional doppelgängers from lower eukaryotes, the spindle pole bodies (SPBs), act as important structural platforms that orchestrate signaling events essential for cell cycle progression, cellular responses to DNA damage, sensory reception and cell homeostasis. Here, we provide a critical overview of the unconventional and often overlooked roles of centrosomes/SPBs in the life cycle of eukaryotic cells.
Collapse
Affiliation(s)
- Laurence Langlois-Lemay
- Department of Cellular and Molecular Medicine, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | | |
Collapse
|
17
|
PIDD1 in cell cycle control, sterile inflammation and cell death. Biochem Soc Trans 2022; 50:813-824. [PMID: 35343572 PMCID: PMC9162469 DOI: 10.1042/bst20211186] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023]
Abstract
The death fold domain-containing protein PIDD1 has recently attracted renewed attention as a regulator of the orphan cell death-related protease, Caspase-2. Caspase-2 can activate p53 to promote cell cycle arrest in response to centrosome aberrations, and its activation requires formation of the PIDDosome multi-protein complex containing multimers of PIDD1 and the adapter RAIDD/CRADD at its core. However, PIDD1 appears to be able to engage with multiple client proteins to promote an even broader range of biological responses, such as NF-κB activation, translesion DNA synthesis or cell death. PIDD1 shows features of inteins, a class of self-cleaving proteins, to create different polypeptides from a common precursor protein that allow it to serve these diverse functions. This review summarizes structural information and molecular features as well as recent experimental advances that highlight the potential pathophysiological roles of this unique death fold protein to highlight its drug-target potential.
Collapse
|
18
|
Therapeutic strategies to overcome cisplatin resistance in ovarian cancer. Eur J Med Chem 2022; 232:114205. [DOI: 10.1016/j.ejmech.2022.114205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
|
19
|
The role of caspases as executioners of apoptosis. Biochem Soc Trans 2021; 50:33-45. [PMID: 34940803 DOI: 10.1042/bst20210751] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/13/2022]
Abstract
Caspases are a family of cysteine aspartyl proteases mostly involved in the execution of apoptotic cell death and in regulating inflammation. This article focuses primarily on the evolutionarily conserved function of caspases in apoptosis. We summarise which caspases are involved in apoptosis, how they are activated and regulated, and what substrates they target for cleavage to orchestrate programmed cell death by apoptosis.
Collapse
|
20
|
Liu N, Yang H, Yang L. Dual roles of SIRT1 in the BAX switch through the P53 module: A mathematical modeling study. Comput Struct Biotechnol J 2021; 19:5578-5588. [PMID: 34849192 PMCID: PMC8598928 DOI: 10.1016/j.csbj.2021.09.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 01/07/2023] Open
Abstract
SIRT1 is a multifunctional deacetylase that participates in a variety of cellular physiological processes to cope with stress. The anticancer protein P53 is an important target of SIRT1. It has been found that SIRT1 is involved in apoptosis by regulating the activity and intracellular location of P53. Moreover, P53-dependent apoptosis is inseparable from the BCL-2 protein family. Among the members of this family, BAX’s switching dynamics may play a key role in apoptosis. Therefore, a challenging question arises: what effect does SIRT1 have on the BAX switch? To answer this question, we built a small-scale protein network model. Through computer simulation, the properties of SIRT1 that on the one hand promote and on the other inhibit apoptosis are revealed. We found that the opening time of the BAX switch will be delayed in the case of either SIRT1 excess or deficiency. Similarly, the stimulus threshold required for apoptosis will also increase in the above two scenarios. Thereby, we proposed that SIRT1 has an optimal content at which the probability of apoptosis is greatest. In addition, P53 oscillation requires the concentration of SIRT1 to be higher than the optimal value. This work may be helpful both experimentally and clinically.
Collapse
Affiliation(s)
- Nan Liu
- School of Mathematical Sciences, Inner Mongolia University, Hohhot 010021, China
| | - Hongli Yang
- School of Mathematical Sciences, Inner Mongolia University, Hohhot 010021, China
- Corresponding author.
| | - Liangui Yang
- School of Mathematical Sciences, Inner Mongolia University, Hohhot 010021, China
| |
Collapse
|
21
|
|
22
|
Progress and challenges in understanding the regulation and function of p53 dynamics. Biochem Soc Trans 2021; 49:2123-2131. [PMID: 34495325 PMCID: PMC8765192 DOI: 10.1042/bst20210148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 02/01/2023]
Abstract
The dynamics of p53 expression provide a mechanism to increase differentiation between cellular stresses and specificity in appropriate responses. Here, we review recent advances in our understanding of the molecular mechanisms regulating p53 dynamics and the functions of the dynamics in the regulation of p53-dependent cell stress responses. We also compare dynamic encoding in the p53 system with that found in other important cell signaling systems, many of which can interact with the p53 network. Finally, we highlight some of the current challenges in understanding dynamic cell signaling within a larger cellular network context.
Collapse
|
23
|
Rizzotto D, Englmaier L, Villunger A. At a Crossroads to Cancer: How p53-Induced Cell Fate Decisions Secure Genome Integrity. Int J Mol Sci 2021; 22:ijms221910883. [PMID: 34639222 PMCID: PMC8509445 DOI: 10.3390/ijms221910883] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
P53 is known as the most critical tumor suppressor and is often referred to as the guardian of our genome. More than 40 years after its discovery, we are still struggling to understand all molecular details on how this transcription factor prevents oncogenesis or how to leverage current knowledge about its function to improve cancer treatment. Multiple cues, including DNA-damage or mitotic errors, can lead to the stabilization and nuclear translocation of p53, initiating the expression of multiple target genes. These transcriptional programs may be cell-type- and stimulus-specific, as is their outcome that ultimately imposes a barrier to cellular transformation. Cell cycle arrest and cell death are two well-studied consequences of p53 activation, but, while being considered critical, they do not fully explain the consequences of p53 loss-of-function phenotypes in cancer. Here, we discuss how mitotic errors alert the p53 network and give an overview of multiple ways that p53 can trigger cell death. We argue that a comparative analysis of different types of p53 responses, elicited by different triggers in a time-resolved manner in well-defined model systems, is critical to understand the cell-type-specific cell fate induced by p53 upon its activation in order to resolve the remaining mystery of its tumor-suppressive function.
Collapse
Affiliation(s)
- Dario Rizzotto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (D.R.); (L.E.)
| | - Lukas Englmaier
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (D.R.); (L.E.)
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria
| | - Andreas Villunger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (D.R.); (L.E.)
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence:
| |
Collapse
|
24
|
Lees A, Sessler T, McDade S. Dying to Survive-The p53 Paradox. Cancers (Basel) 2021; 13:3257. [PMID: 34209840 PMCID: PMC8268032 DOI: 10.3390/cancers13133257] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
The p53 tumour suppressor is best known for its canonical role as "guardian of the genome", activating cell cycle arrest and DNA repair in response to DNA damage which, if irreparable or sustained, triggers activation of cell death. However, despite an enormous amount of work identifying the breadth of the gene regulatory networks activated directly and indirectly in response to p53 activation, how p53 activation results in different cell fates in response to different stress signals in homeostasis and in response to p53 activating anti-cancer treatments remains relatively poorly understood. This is likely due to the complex interaction between cell death mechanisms in which p53 has been activated, their neighbouring stressed or unstressed cells and the local stromal and immune microenvironment in which they reside. In this review, we evaluate our understanding of the burgeoning number of cell death pathways affected by p53 activation and how these may paradoxically suppress cell death to ensure tissue integrity and organismal survival. We also discuss how these functions may be advantageous to tumours that maintain wild-type p53, the understanding of which may provide novel opportunity to enhance treatment efficacy.
Collapse
Affiliation(s)
- Andrea Lees
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK;
| | | | - Simon McDade
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK;
| |
Collapse
|
25
|
The p53-caspase-2 axis in the cell cycle and DNA damage response. Exp Mol Med 2021; 53:517-527. [PMID: 33854186 PMCID: PMC8102494 DOI: 10.1038/s12276-021-00590-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 02/08/2023] Open
Abstract
Caspase-2 was discovered almost three decades ago. It was one of the first two mammalian homologs of CED-3, the other being interleukin 1β-converting enzyme (ICE/caspase-1). Despite high similarity with CED-3 and its fly and mammalian counterparts (DRONC and caspase-9, respectively), the function of caspase-2 in apoptosis has remained enigmatic. A number of recent studies suggest that caspase-2 plays an important role in the regulation of p53 in response to cellular stress and DNA damage to prevent the proliferation and accumulation of damaged or aberrant cells. Here, we review these recent observations and their implications in caspase-2-mediated cellular death, senescence, and tumor suppression.
Collapse
|
26
|
Tsabar M, Lovitch SB, Jambhekar A, Lahav G. Connecting Timescales in Biology: Can Early Dynamical Measurements Predict Long-Term Outcomes? Trends Cancer 2021; 7:301-308. [PMID: 33451930 PMCID: PMC8796003 DOI: 10.1016/j.trecan.2020.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022]
Abstract
Prediction of long-term outcomes from short-term measurements remains a fundamental challenge. Quantitative assessment of signaling dynamics, and the resulting transcriptomic and proteomic responses, has yielded fundamental insights into cellular outcomes. However, the utility of these measurements is limited by their short timescale (hours to days), while the consequences of these events frequently unfold over longer timescales. Here, we discuss the predictive power of static and dynamic measurements, drawing examples from fields that have harnessed the predictive capabilities of such measurements. We then explore potential approaches to close this timescale gap using complementary measurements and computational approaches, focusing on the example of dynamic measurements of signaling factors and their impacts on cellular outcomes.
Collapse
Affiliation(s)
- Michael Tsabar
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA; Laboratory of Systems Pharmacology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA; Broad institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Scott B Lovitch
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Galit Lahav
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Ramu D, Shan TW, Hirpara JL, Pervaiz S. Cellular senescence: Silent operator and therapeutic target in cancer. IUBMB Life 2021; 73:530-542. [PMID: 33675120 DOI: 10.1002/iub.2460] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 12/30/2022]
Abstract
The process of carcinogenesis and its progression involves an intricate interplay between a number of signaling networks, metabolic pathways and the microenvironment. These include an alteration in the cellular redox metabolism and deregulation of cell cycle checkpoints. Similar to the dichotomy of redox signaling in cancer cell fate and state determination, a diverging effect of an irreversible cell cycle arrest or senescence on carcinogenesis has been demonstrated. In this regard, while overwhelming oxidative stress has a damaging effect on tissue architecture and organ function and promotes death execution, a mild "pro-oxidant" environment is conducive for cell proliferation, growth and survival. Similarly, cellular senescence has been shown to elicit both a tumor suppressor and an oncogenic effect in a context-dependent manner. Notably, there appears to be a crosstalk between these two critical regulators of cell fate and state, particularly from the standpoint of the divergent effects on processes that promote or abate carcinogenesis. This review aims to provide an overview of these overarching themes and attempts to highlight critical intersection nodes, which are emerging as potential diagnostic and/or therapeutic targets for novel anticancer strategies.
Collapse
Affiliation(s)
- Deepika Ramu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Teoh Wei Shan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jayshree L Hirpara
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Medicine Healthy Longevity Program, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore, Singapore.,Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore, Singapore.,Faculté de Medicine, University of Paris, Paris, France
| |
Collapse
|
28
|
Krenning L, Raaijmakers JA, Medema RH. Centrosomes: Please keep your social distance! EMBO J 2021; 40:e107525. [PMID: 33491191 DOI: 10.15252/embj.2020107525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/24/2020] [Indexed: 01/08/2023] Open
Abstract
Accurate control of centrosome number is essential for proper chromosome segregation, and it is well established that centrosome abnormalities can trigger a p53-dependent cell cycle arrest. Two new studies published in The EMBO Journal demonstrate how PIDD1 is recruited to centrosomes and that the localization of PIDD1 to distal appendages of centrosomes is required for PIDDosome activation at clustered supernumerary centrosomes.
Collapse
Affiliation(s)
- Lenno Krenning
- Division of Cell Biology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jonne A Raaijmakers
- Division of Cell Biology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - René H Medema
- Division of Cell Biology, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
29
|
Burigotto M, Mattivi A, Migliorati D, Magnani G, Valentini C, Roccuzzo M, Offterdinger M, Pizzato M, Schmidt A, Villunger A, Maffini S, Fava LL. Centriolar distal appendages activate the centrosome-PIDDosome-p53 signalling axis via ANKRD26. EMBO J 2021; 40:e104844. [PMID: 33350486 PMCID: PMC7883297 DOI: 10.15252/embj.2020104844] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Centrosome amplification results into genetic instability and predisposes cells to neoplastic transformation. Supernumerary centrosomes trigger p53 stabilization dependent on the PIDDosome (a multiprotein complex composed by PIDD1, RAIDD and Caspase-2), whose activation results in cleavage of p53's key inhibitor, MDM2. Here, we demonstrate that PIDD1 is recruited to mature centrosomes by the centriolar distal appendage protein ANKRD26. PIDDosome-dependent Caspase-2 activation requires not only PIDD1 centrosomal localization, but also its autoproteolysis. Following cytokinesis failure, supernumerary centrosomes form clusters, which appear to be necessary for PIDDosome activation. In addition, in the context of DNA damage, activation of the complex results from a p53-dependent elevation of PIDD1 levels independently of centrosome amplification. We propose that PIDDosome activation can in both cases be promoted by an ANKRD26-dependent local increase in PIDD1 concentration close to the centrosome. Collectively, these findings provide a paradigm for how centrosomes can contribute to cell fate determination by igniting a signalling cascade.
Collapse
Affiliation(s)
- Matteo Burigotto
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Alessia Mattivi
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Daniele Migliorati
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Giovanni Magnani
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Chiara Valentini
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Michela Roccuzzo
- Advanced Imaging Core FacilityDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Martin Offterdinger
- Division of NeurobiochemistryBioopticsBiocenterMedical University of InnsbruckInnsbruckAustria
| | - Massimo Pizzato
- Laboratory of Virus‐Cell InteractionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| | - Alexander Schmidt
- Proteomics Core FacilityBiozentrumUniversity of BaselBaselSwitzerland
| | - Andreas Villunger
- Division of Developmental ImmunologyBiocenterMedical University of InnsbruckInnsbruckAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Stefano Maffini
- Department of Mechanistic Cell BiologyMax Planck Institute of Molecular PhysiologyDortmundGermany
| | - Luca L Fava
- Armenise‐Harvard Laboratory of Cell DivisionDepartment of Cellular, Computational and Integrative Biology—CIBIOUniversity of TrentoTrentoItaly
| |
Collapse
|
30
|
Activation of DNA damage response signaling in mammalian cells by ionizing radiation. Free Radic Res 2021; 55:581-594. [PMID: 33455476 DOI: 10.1080/10715762.2021.1876853] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cellular responses to DNA damage are fundamental to preserve genomic integrity during various endogenous and exogenous stresses. Following radiation therapy and chemotherapy, this DNA damage response (DDR) also determines development of carcinogenesis and therapeutic outcome. In humans, DNA damage activates a robust network of signal transduction cascades, driven primarily through phosphorylation events. These responses primarily involve two key non-redundant signal transducing proteins of phosphatidylinositol 3-kinase-like (PIKK) family - ATR and ATM, and their downstream kinases (hChk1 and hChk2). They further phosphorylate effectors proteins such as p53, Cdc25A and Cdc25C which function either to activate the DNA damage checkpoints and cell death mechanisms, or DNA repair pathways. Identification of molecular pathways that determine signaling after DNA damage and trigger DNA repair in response to differing types of DNA lesions allows for a far better understanding of the consequences of radiation and chemotherapy on normal and tumor cells. Here we highlight the network of DNA damage response pathways that are activated after treatment with different types of radiation. Further, we discuss regulation of cell cycle checkpoint and DNA repair processes in the context of DDR in response to radiation.
Collapse
|
31
|
Tang Y, Qian W, Zhang B, Liu W, Sun X, Ji W, Ma L, Zhu D. None-Loss Target Release of Biomimetic CaCO 3 Nanocomposites for Screening Bioactive Components and Target Proteins. ACS APPLIED BIO MATERIALS 2021. [DOI: 10.1021/acsabm.0c01197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yingying Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Wenhui Qian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
- Department of Pharmacy, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, Jiangsu, China
| | - Bei Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Wenya Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
- Department of Pharmacy, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, Jiangsu, China
| | - Xuetong Sun
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Wenwen Ji
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Lijuan Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| | - Dong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210002, Jiangsu, China
| |
Collapse
|
32
|
Petroni G, Vitale I, Galluzzi L. Caspase 2 and p53 Reunited in Tumor Control. Trends Cell Biol 2020; 30:917-918. [PMID: 32921524 DOI: 10.1016/j.tcb.2020.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 11/18/2022]
Abstract
Recent findings (Tsabar et al.) demonstrate that human cancer cells that evade the cell cycle blockage normally imposed by DNA damage experience sustained p53 signaling upon MDM2 degradation by caspase 2. Such a response may be connected to the delivery of immunostimulatory signals to ensure the elimination of genetically instable cancer cells.
Collapse
Affiliation(s)
- Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA; Université de Paris, Paris, France.
| |
Collapse
|