1
|
Lu D, Faizi M, Drown B, Simerzin A, François J, Bradshaw G, Kelleher N, Jambhekar A, Gunawardena J, Lahav G. Temporal regulation of gene expression through integration of p53 dynamics and modifications. SCIENCE ADVANCES 2024; 10:eadp2229. [PMID: 39454005 PMCID: PMC11506164 DOI: 10.1126/sciadv.adp2229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/20/2024] [Indexed: 10/27/2024]
Abstract
The master regulator of the DNA damage response, the transcription factor p53, orchestrates multiple downstream responses and coordinates repair processes. In response to double-strand DNA breaks, p53 exhibits pulses of expression, but how it achieves temporal coordination of downstream responses remains unclear. Here, we show that p53's posttranslational modification state is altered between its first and second pulses of expression. We show that acetylations at two sites, K373 and K382, were reduced in the second pulse, and these acetylations differentially affected p53 target genes, resulting in changes in gene expression programs over time. This interplay between dynamics and modification may offer a strategy for cellular hubs like p53 to temporally organize multiple processes in individual cells.
Collapse
Affiliation(s)
- Dan Lu
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Marjan Faizi
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Bryon Drown
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Alina Simerzin
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Joshua François
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Gary Bradshaw
- Laboratory of Systems Pharmacology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Neil Kelleher
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
- Ludwig Center at Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy Gunawardena
- 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
- Ludwig Center at Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
2
|
Jiménez A, Lucchetti A, Heltberg MS, Moretto L, Sanchez C, Jambhekar A, Jensen MH, Lahav G. Entrainment and multi-stability of the p53 oscillator in human cells. Cell Syst 2024; 15:956-968.e3. [PMID: 39368467 DOI: 10.1016/j.cels.2024.09.001] [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: 01/09/2024] [Revised: 06/25/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
The tumor suppressor p53 responds to cellular stress and activates transcription programs critical for regulating cell fate. DNA damage triggers oscillations in p53 levels with a robust period. Guided by the theory of synchronization and entrainment, we developed a mathematical model and experimental system to test the ability of the p53 oscillator to entrain to external drug pulses of various periods and strengths. We found that the p53 oscillator can be locked and entrained to a wide range of entrainment modes. External periods far from p53's natural oscillations increased the heterogeneity between individual cells whereas stronger inputs reduced it. Single-cell measurements allowed deriving the phase response curves (PRCs) and multiple Arnold tongues of p53. In addition, multi-stability and non-linear behaviors were mathematically predicted and experimentally detected, including mode hopping, period doubling, and chaos. Our work revealed critical dynamical properties of the p53 oscillator and provided insights into understanding and controlling it. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Alba Jiménez
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Alessandra Lucchetti
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA; Niels Bohr Institute, University of Copenhagen, Copenhagen 2100, Denmark
| | - Mathias S Heltberg
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA; Niels Bohr Institute, University of Copenhagen, Copenhagen 2100, Denmark
| | - Liv Moretto
- Niels Bohr Institute, University of Copenhagen, Copenhagen 2100, Denmark
| | - Carlos Sanchez
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA
| | - Mogens H Jensen
- Niels Bohr Institute, University of Copenhagen, Copenhagen 2100, Denmark.
| | - Galit Lahav
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Fischer M, Sammons MA. Determinants of p53 DNA binding, gene regulation, and cell fate decisions. Cell Death Differ 2024; 31:836-843. [PMID: 38951700 PMCID: PMC11239874 DOI: 10.1038/s41418-024-01326-1] [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: 03/09/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 07/03/2024] Open
Abstract
The extent to which transcription factors read and respond to specific information content within short DNA sequences remains an important question that the tumor suppressor p53 is helping us answer. We discuss recent insights into how local information content at p53 binding sites might control modes of p53 target gene activation and cell fate decisions. Significant prior work has yielded data supporting two potential models of how p53 determines cell fate through its target genes: a selective target gene binding and activation model and a p53 level threshold model. Both of these models largely revolve around an analogy of whether p53 is acting in a "smart" or "dumb" manner. Here, we synthesize recent and past studies on p53 decoding of DNA sequence, chromatin context, and cellular signaling cascades to elicit variable cell fates critical in human development, homeostasis, and disease.
Collapse
Affiliation(s)
- Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745, Jena, Germany.
| | - Morgan A Sammons
- Department of Biological Sciences and The RNA Institute, The State University of New York at Albany, 1400 Washington Avenue, Albany, NY, 12222, USA.
| |
Collapse
|
4
|
Fischer M. Gene regulation by the tumor suppressor p53 - The omics era. Biochim Biophys Acta Rev Cancer 2024; 1879:189111. [PMID: 38740351 DOI: 10.1016/j.bbcan.2024.189111] [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: 11/27/2023] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
The transcription factor p53 is activated in response to a variety of cellular stresses and serves as a prominent and potent tumor suppressor. Since its discovery, we have sought to understand how p53 functions as both a transcription factor and a tumor suppressor. Two decades ago, the field of gene regulation entered the omics era and began to study the regulation of entire genomes. The omics perspective has greatly expanded our understanding of p53 functions and has begun to reveal its gene regulatory network. In this mini-review, I discuss recent insights into the p53 transcriptional program from high-throughput analyses.
Collapse
Affiliation(s)
- Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany.
| |
Collapse
|
5
|
Serra F, Nieto-Aliseda A, Fanlo-Escudero L, Rovirosa L, Cabrera-Pasadas M, Lazarenkov A, Urmeneta B, Alcalde-Merino A, Nola EM, Okorokov AL, Fraser P, Graupera M, Castillo SD, Sardina JL, Valencia A, Javierre BM. p53 rapidly restructures 3D chromatin organization to trigger a transcriptional response. Nat Commun 2024; 15:2821. [PMID: 38561401 PMCID: PMC10984980 DOI: 10.1038/s41467-024-46666-1] [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: 06/15/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Activation of the p53 tumor suppressor triggers a transcriptional program to control cellular response to stress. However, the molecular mechanisms by which p53 controls gene transcription are not completely understood. Here, we uncover the critical role of spatio-temporal genome architecture in this process. We demonstrate that p53 drives direct and indirect changes in genome compartments, topologically associating domains, and DNA loops prior to one hour of its activation, which escort the p53 transcriptional program. Focusing on p53-bound enhancers, we report 340 genes directly regulated by p53 over a median distance of 116 kb, with 74% of these genes not previously identified. Finally, we showcase that p53 controls transcription of distal genes through newly formed and pre-existing enhancer-promoter loops in a cohesin dependent manner. Collectively, our findings demonstrate a previously unappreciated architectural role of p53 as regulator at distinct topological layers and provide a reliable set of new p53 direct target genes that may help designs of cancer therapies.
Collapse
Affiliation(s)
- François Serra
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | | | | | | | - Mónica Cabrera-Pasadas
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Barcelona Supercomputing Center, Barcelona, Spain
| | | | - Blanca Urmeneta
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | | | - Emanuele M Nola
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Andrei L Okorokov
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Peter Fraser
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Mariona Graupera
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jose L Sardina
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Alfonso Valencia
- Barcelona Supercomputing Center, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Biola M Javierre
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain.
- Institute for Health Science Research Germans Trias i Pujol, Barcelona, Spain.
| |
Collapse
|
6
|
Song B, Yang P, Zhang S. Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy. Cancer Commun (Lond) 2024; 44:297-360. [PMID: 38311377 PMCID: PMC10958678 DOI: 10.1002/cac2.12520] [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: 07/26/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Bin Song
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ping Yang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Shuyu Zhang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanP. R. China
- Laboratory of Radiation MedicineNHC Key Laboratory of Nuclear Technology Medical TransformationWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
7
|
Ding Y, Tous C, Choi J, Chen J, Wong WW. Orthogonal inducible control of Cas13 circuits enables programmable RNA regulation in mammalian cells. Nat Commun 2024; 15:1572. [PMID: 38383558 PMCID: PMC10881482 DOI: 10.1038/s41467-024-45795-x] [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: 04/26/2023] [Accepted: 02/06/2024] [Indexed: 02/23/2024] Open
Abstract
RNA plays an indispensable role in mammalian cell functions. Cas13, a class of RNA-guided ribonuclease, is a flexible tool for modifying and regulating coding and non-coding RNAs, with enormous potential for creating new cell functions. However, the lack of control over Cas13 activity has limited its cell engineering capability. Here, we present the CRISTAL (Control of RNA with Inducible SpliT CAs13 Orthologs and Exogenous Ligands) platform. CRISTAL is powered by a collection (10 total) of orthogonal split inducible Cas13 effectors that can be turned ON or OFF via small molecules in multiple cell types, providing precise temporal control. Also, we engineer Cas13 logic circuits that can respond to endogenous signaling and exogenous small molecule inputs. Furthermore, the orthogonality, low leakiness, and high dynamic range of our inducible Cas13d and Cas13b enable the design and construction of a robust incoherent feedforward loop, leading to near-perfect and tunable adaptation response. Finally, using our inducible Cas13 effectors, we achieve simultaneous multiplexed control of multiple genes in vitro and in mice. Together, our CRISTAL design represents a powerful platform for precisely regulating RNA dynamics to advance cell engineering and elucidate RNA biology.
Collapse
Affiliation(s)
- Yage Ding
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 2215, USA
| | - Cristina Tous
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 2215, USA
| | - Jaehoon Choi
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 2215, USA
| | - Jingyao Chen
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 2215, USA
| | - Wilson W Wong
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 2215, USA.
| |
Collapse
|
8
|
Park SY, Kim D, Jung JW, An HJ, Lee J, Park Y, Lee D, Lee S, Kim JM. Targeting class A GPCRs for hard tissue regeneration. Biomaterials 2024; 304:122425. [PMID: 38100905 DOI: 10.1016/j.biomaterials.2023.122425] [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: 04/04/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
G protein-coupled receptors (GPCRs) play important roles in various pathogeneses and physiological regulations. Owing to their functional diversity, GPCRs are considered one of the primary pharmaceutical targets. However, drugs targeting GPCRs have not been developed yet to regenerate hard tissues such as teeth and bones. Mesenchymal stromal cells (MSCs) have high proliferation and multi-lineage differentiation potential, which are essential for hard tissue regeneration. Here, we present a strategy for targeting class A GPCRs for hard tissue regeneration by promoting the differentiation of endogenous MSCs into osteogenic and odontogenic progenitor cells. Through in vitro screening targeted at class A GPCRs, we identified six target receptors (LPAR1, F2R, F2RL1, F2RL2, S1PR1, and ADORA2A) and candidate drugs with potent biomineralization effects. Through a combination of profiling whole transcriptome and accessible chromatin regions, we identified that p53 acts as a key transcriptional activator of genes that modulate the biomineralization process. Moreover, the therapeutic potential of class A GPCR-targeting drugs was demonstrated in tooth pulpotomy and calvarial defect models. The selected drugs revealed potent regenerative effects in both tooth and bone defects, represented by newly formed highly mineralized regions. Consequently, this study provides translational evidence for a new regenerative strategy for damaged hard tissue.
Collapse
Affiliation(s)
- So Young Park
- Department of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dohyun Kim
- Department of Conservative Dentistry and Oral Science Research Center, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Ju Won Jung
- Department of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyun-Ju An
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Jaemin Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Bundang-gu, Seongnam-si, 13496, Republic of Korea
| | - Yeji Park
- Department of Conservative Dentistry and Oral Science Research Center, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Dasun Lee
- Department of Conservative Dentistry and Oral Science Research Center, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, Bundang-gu, Seongnam-si, 13496, Republic of Korea.
| | - Jin Man Kim
- Department of Oral Microbiology and Immunology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
9
|
Qiao S, Bernasek S, Gallagher KD, O'Connell J, Yamada S, Bagheri N, Amaral LAN, Carthew RW. Energy metabolism modulates the regulatory impact of activators on gene expression. Development 2024; 151:dev201986. [PMID: 38063847 PMCID: PMC10820824 DOI: 10.1242/dev.201986] [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: 05/15/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023]
Abstract
Gene expression is a regulated process fueled by ATP consumption. Therefore, regulation must be coupled to constraints imposed by the level of energy metabolism. Here, we explore this relationship both theoretically and experimentally. A stylized mathematical model predicts that activators of gene expression have variable impact depending on metabolic rate. Activators become less essential when metabolic rate is reduced and more essential when metabolic rate is enhanced. We find that, in the Drosophila eye, expression dynamics of the yan gene are less affected by loss of EGFR-mediated activation when metabolism is reduced, and the opposite effect is seen when metabolism is enhanced. The effects are also seen at the level of pattern regularity in the adult eye, where loss of EGFR-mediated activation is mitigated by lower metabolism. We propose that gene activation is tuned by energy metabolism to allow for faithful expression dynamics in the face of variable metabolic conditions.
Collapse
Affiliation(s)
- Sha Qiao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Sebastian Bernasek
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Kevin D. Gallagher
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| | - Jessica O'Connell
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Shigehiro Yamada
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Neda Bagheri
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL 60208, USA
| | - Luis A. N. Amaral
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL 60208, USA
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208, USA
| | - Richard W. Carthew
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
10
|
Ram A, Murphy D, DeCuzzi N, Patankar M, Hu J, Pargett M, Albeck JG. A guide to ERK dynamics, part 2: downstream decoding. Biochem J 2023; 480:1909-1928. [PMID: 38038975 PMCID: PMC10754290 DOI: 10.1042/bcj20230277] [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: 07/09/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 12/02/2023]
Abstract
Signaling by the extracellular signal-regulated kinase (ERK) pathway controls many cellular processes, including cell division, death, and differentiation. In this second installment of a two-part review, we address the question of how the ERK pathway exerts distinct and context-specific effects on multiple processes. We discuss how the dynamics of ERK activity induce selective changes in gene expression programs, with insights from both experiments and computational models. With a focus on single-cell biosensor-based studies, we summarize four major functional modes for ERK signaling in tissues: adjusting the size of cell populations, gradient-based patterning, wave propagation of morphological changes, and diversification of cellular gene expression states. These modes of operation are disrupted in cancer and other related diseases and represent potential targets for therapeutic intervention. By understanding the dynamic mechanisms involved in ERK signaling, there is potential for pharmacological strategies that not only simply inhibit ERK, but also restore functional activity patterns and improve disease outcomes.
Collapse
Affiliation(s)
- Abhineet Ram
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Devan Murphy
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Nicholaus DeCuzzi
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Madhura Patankar
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Jason Hu
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - Michael Pargett
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| | - John G. Albeck
- Department of Molecular and Cellular Biology, University of California, Davis, CA, U.S.A
| |
Collapse
|
11
|
Safieh J, Chazan A, Saleem H, Vyas P, Danin-Poleg Y, Ron D, Haran TE. A molecular mechanism for the "digital" response of p53 to stress. Proc Natl Acad Sci U S A 2023; 120:e2305713120. [PMID: 38015851 PMCID: PMC10710088 DOI: 10.1073/pnas.2305713120] [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: 04/09/2023] [Accepted: 10/25/2023] [Indexed: 11/30/2023] Open
Abstract
The tumor suppressor protein p53 accumulates in response to cellular stress and consequently orchestrates the expression of multiple genes in a p53-level and time-dependent manner to overcome stress consequences, for which a molecular mechanism is currently unknown. Previously, we reported that DNA torsional flexibility distinguishes among p53 response elements (REs) and that transactivation at basal p53 levels is correlated with p53 REs flexibility. Here, we calculated the flexibility of ~200 p53 REs. By connecting functional outcomes of p53-target genes' activation to the calculated flexibility of their REs, we show that genes known to belong to pathways that are activated rapidly upon stress contain REs that are significantly more flexible relative to REs of genes known to be involved in pathways that are activated later in the response to stress. The global structural properties of several p53 REs belonging to different pathways were experimentally validated. Additionally, reporter-gene expression driven by flexible p53 REs occurred at lower p53 levels and with faster rates than expression from rigid REs. Furthermore, analysis of published endogenous mRNA levels of p53-target genes as a function of REs' flexibility showed that early versus late genes differ significantly in their flexibility properties of their REs and that highly flexible p53 REs enable high-activation level exclusively to early-response genes. Overall, we demonstrate that DNA flexibility of p53 REs contributes significantly to functional selectivity in the p53 system by facilitating the initial steps of p53-dependent target-genes expression, thereby contributing to survival versus death decisions in the p53 system.
Collapse
Affiliation(s)
- Jessy Safieh
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Ariel Chazan
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Hanna Saleem
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Pratik Vyas
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Yael Danin-Poleg
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Dina Ron
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| | - Tali E. Haran
- Department of Biology, Technion, Technion City, Haifa2300003, Israel
| |
Collapse
|
12
|
Yue Z, Lin J, Lu X, Gao Q, Pan M, Zhang Y, Shen S, Zhu WG, Paus R. Keratin 17 Impacts Global Gene Expression and Controls G2/M Cell Cycle Transition in Ionizing Radiation-Induced Skin Damage. J Invest Dermatol 2023; 143:2436-2446.e13. [PMID: 37414246 DOI: 10.1016/j.jid.2023.02.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 07/08/2023]
Abstract
Keratin 17 (K17) is a cytoskeletal protein that is part of the intermediate filaments in epidermal keratinocytes. In K17-/- mice, ionizing radiation induced more severe hair follicle damage, whereas the epidermal inflammatory response was attenuated compared with that in wild-type mice. Both p53 and K17 have a major impact on global gene expression because over 70% of the differentially expressed genes in the skin of wild-type mice showed no expression change in p53-/- or K17-/- skin after ionizing radiation. K17 does not interfere with the dynamics of p53 activation; rather, global p53 binding in the genome is altered in K17-/- mice. The absence of K17 leads to aberrant cell cycle progression and mitotic catastrophe in epidermal keratinocytes, which is due to nuclear retention, thus reducing the degradation of B-Myb, a key regulator of the G2/M cell cycle transition. These results expand our understanding of the role of K17 in regulating global gene expression and ionizing radiation-induced skin damage.
Collapse
Affiliation(s)
- ZhiCao Yue
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China.
| | - JianQiong Lin
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - XiaoPeng Lu
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China; Department of Biochemistry & Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - QingXiang Gao
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - MeiPing Pan
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - YaFei Zhang
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - SiTing Shen
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - Wei-Guo Zhu
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China; Department of Biochemistry & Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Ralf Paus
- Dr. Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA; Center for Dermatology Research, School of Biological Sciences, The University of Manchester and NIHR Biomedical Research Center, Manchester, United Kingdom
| |
Collapse
|
13
|
Wang NB, Lende-Dorn BA, Adewumi HO, Beitz AM, Han P, O'Shea TM, Galloway KE. Proliferation history and transcription factor levels drive direct conversion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.26.568736. [PMID: 38077004 PMCID: PMC10705288 DOI: 10.1101/2023.11.26.568736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The sparse and stochastic nature of reprogramming has obscured our understanding of how transcription factors drive cells to new identities. To overcome this limit, we developed a compact, portable reprogramming system that increases direct conversion of fibroblasts to motor neurons by two orders of magnitude. We show that subpopulations with different reprogramming potentials are distinguishable by proliferation history. By controlling for proliferation history and titrating each transcription factor, we find that conversion correlates with levels of the pioneer transcription factor Ngn2, whereas conversion shows a biphasic response to Lhx3. Increasing the proliferation rate of adult human fibroblasts generates morphologically mature, induced motor neurons at high rates. Using compact, optimized, polycistronic cassettes, we generate motor neurons that graft with the murine central nervous system, demonstrating the potential for in vivo therapies.
Collapse
Affiliation(s)
- Nathan B Wang
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | | | - Honour O Adewumi
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Adam M Beitz
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Patrick Han
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| | - Timothy M O'Shea
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kate E Galloway
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA
| |
Collapse
|
14
|
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
|
15
|
Qiao S, Bernasek S, Gallagher KD, Yamada S, Bagheri N, Amaral LA, Carthew RW. Energy metabolism modulates the regulatory impact of activators on gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563842. [PMID: 37961620 PMCID: PMC10634812 DOI: 10.1101/2023.10.24.563842] [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
Gene expression is a regulated process fueled by ATP consumption. Therefore, regulation must be coupled to constraints imposed by the level of energy metabolism. Here, we explore this relationship both theoretically and experimentally. A stylized mathematical model predicts that activators of gene expression have variable impact depending on metabolic rate. Activators become less essential when metabolic rate is reduced and more essential when metabolic rate is enhanced. We find that in the Drosophila eye, expression dynamics of the yan gene are less affected by loss of EGFR-mediated activation when metabolism is reduced, and the opposite effect is seen when metabolism is enhanced. The effects are also seen at the level of pattern regularity in the adult eye, where loss of EGFR-mediated activation is mitigated by lower metabolism. We propose that gene activation is tuned by energy metabolism to allow for faithful expression dynamics in the face of variable metabolic conditions.
Collapse
Affiliation(s)
- Sha Qiao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL
| | - Sebastian Bernasek
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL
| | - Kevin D. Gallagher
- Department of Molecular Biosciences, Northwestern University, Evanston, IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL
| | - Shigehiro Yamada
- Department of Molecular Biosciences, Northwestern University, Evanston, IL
| | - Neda Bagheri
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL
| | - Luis A.N. Amaral
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL
- Department of Physics and Astronomy, Northwestern University, Evanston, IL
| | - Richard W. Carthew
- Department of Molecular Biosciences, Northwestern University, Evanston, IL
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL
- Lead Contact
| |
Collapse
|
16
|
Zhang Z, Zhang M, Zhou J, Wang D. Genome-wide CRISPR screening reveals ADCK3 as a key regulator in sensitizing endometrial carcinoma cells to MPA therapy. Br J Cancer 2023; 129:601-611. [PMID: 37402867 PMCID: PMC10421920 DOI: 10.1038/s41416-023-02347-2] [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: 01/10/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND The effectiveness of conservative treatment of endometrial carcinoma (EC) with oral progesterone therapy, such as medroxyprogesterone acetate (MPA), can be blunted due to primary or acquired resistance, but the underlying mechanisms remain incompletely defined. METHODS Genome-wide CRISPR screening was performed to identify potential regulators in response to MPA in Ishikawa cells. Crystal violet staining, RT-qPCR, western blotting, ChIP-qPCR and luciferase assays were employed to elucidate the p53-AarF domain-containing kinase 3 (ADCK3) regulatory axis and its roles in sensitizing EC cells to MPA treatment. RESULTS ADCK3 is identified as a previously unrecognized regulator in response to MPA in EC cells. Loss of ADCK3 in EC cells markedly alleviated MPA-induced cell death. Mechanistically, loss of ADCK3 primarily suppresses MPA-mediated ferroptosis by abrogating arachidonate 15-lipoxygenase (ALOX15) transcriptional activation. Moreover, we validated ADCK3 as a direct downstream target of the tumor suppressor p53 in EC cells. By stimulating the p53-ADCK3 axis, the small-molecule compound Nutlin3A synergized with MPA to efficiently inhibit EC cell growth. CONCLUSIONS Our findings reveal ADCK3 as a key regulator of EC cells in response to MPA and shed light on a potential strategy for conservative EC treatment by activating the p53-ADCK3 axis to sensitize MPA-mediated cell death.
Collapse
Affiliation(s)
- Zijing Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 100005, Beijing, China
| | - Meng Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 100005, Beijing, China
| | - Jingyi Zhou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, 100044, Beijing, China.
| | - Donglai Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases & Department of Medical Genetics, Institute of Basic Medical Sciences & School of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 100005, Beijing, China.
| |
Collapse
|
17
|
Rodriguez-Pastrana I, Birli E, Coutts AS. p53-dependent DNA repair during the DNA damage response requires actin nucleation by JMY. Cell Death Differ 2023:10.1038/s41418-023-01170-9. [PMID: 37142657 DOI: 10.1038/s41418-023-01170-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 05/06/2023] Open
Abstract
The tumour suppressor p53 is a nuclear transcription factor with key roles during DNA damage to enable a variety of cellular responses including cell cycle arrest, apoptosis and DNA repair. JMY is an actin nucleator and DNA damage-responsive protein whose sub-cellular localisation is responsive to stress and during DNA damage JMY undergoes nuclear accumulation. To gain an understanding of the wider role for nuclear JMY in transcriptional regulation, we performed transcriptomics to identify JMY-mediated changes in gene expression during the DNA damage response. We show that JMY is required for effective regulation of key p53 target genes involved in DNA repair, including XPC, XRCC5 (Ku80) and TP53I3 (PIG3). Moreover, JMY depletion or knockout leads to increased DNA damage and nuclear JMY requires its Arp2/3-dependent actin nucleation function to promote the clearance of DNA lesions. In human patient samples a lack of JMY is associated with increased tumour mutation count and in cells results in reduced cell survival and increased sensitivity to DNA damage response kinase inhibition. Collectively, we demonstrate that JMY enables p53-dependent DNA repair under genotoxic stress and suggest a role for actin in JMY nuclear activity during the DNA damage response.
Collapse
Affiliation(s)
- Ignacio Rodriguez-Pastrana
- School of Science and Technology, Department of Biosciences, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK
| | - Eleni Birli
- School of Science and Technology, Department of Biosciences, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK
- John van Geest Cancer Research Centre, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK
| | - Amanda S Coutts
- School of Science and Technology, Department of Biosciences, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK.
| |
Collapse
|
18
|
Ding Y, Tous C, Choi J, Chen J, Wong WW. Orthogonal inducible control of Cas13 circuits enables programmable RNA regulation in mammalian cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.20.533499. [PMID: 36993327 PMCID: PMC10055290 DOI: 10.1101/2023.03.20.533499] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
RNA plays an indispensable role in mammalian cell functions. Cas13, a class of RNA-guided ribonuclease, is a flexible tool for modifying and regulating coding and non-coding RNAs, with enormous potential for creating new cell functions. However, the lack of control over Cas13 activity has limited its cell engineering capability. Here, we present the CRISTAL ( C ontrol of R NA with Inducible S pli T C A s13 Orthologs and Exogenous L igands) platform. CRISTAL is powered by a collection (10 total) of orthogonal split inducible Cas13s that can be turned ON or OFF via small molecules in multiple cell types, providing precise temporal control. Also, we engineered Cas13 logic circuits that can respond to endogenous signaling and exogenous small molecule inputs. Furthermore, the orthogonality, low leakiness, and high dynamic range of our inducible Cas13d and Cas13b enable the design and construction of a robust incoherent feedforward loop, leading to near-perfect and tunable adaptation response. Finally, using our inducible Cas13s, we achieve simultaneous multiplexed control of multiple genes in vitro and in mice. Together, our CRISTAL design represents a powerful platform for precisely regulating RNA dynamics to advance cell engineering and elucidate RNA biology.
Collapse
|
19
|
Wang P, Wang HY, Gao XJ, Zhu HX, Zhang XP, Liu F, Wang W. Encoding and Decoding of p53 Dynamics in Cellular Response to Stresses. Cells 2023; 12:cells12030490. [PMID: 36766831 PMCID: PMC9914463 DOI: 10.3390/cells12030490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
In the cellular response to stresses, the tumor suppressor p53 is activated to maintain genomic integrity and fidelity. As a transcription factor, p53 exhibits rich dynamics to allow for discrimination of the type and intensity of stresses and to direct the selective activation of target genes involved in different processes including cell cycle arrest and apoptosis. In this review, we focused on how stresses are encoded into p53 dynamics and how the dynamics are decoded into cellular outcomes. Theoretical modeling may provide a global view of signaling in the p53 network by coupling the encoding and decoding processes. We discussed the significance of modeling in revealing the mechanisms of the transition between p53 dynamic modes. Moreover, we shed light on the crosstalk between the p53 network and other signaling networks. This review may advance the understanding of operating principles of the p53 signaling network comprehensively and provide insights into p53 dynamics-based cancer therapy.
Collapse
Affiliation(s)
- Ping Wang
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Key Laboratory of High Performance Scientific Computation, School of Science, Xihua University, Chengdu 610039, China
| | - Hang-Yu Wang
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
| | - Xing-Jie Gao
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
| | - Hua-Xia Zhu
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
| | - Xiao-Peng Zhang
- Kuang Yaming Honors School, Nanjing University, Nanjing 210023, China
- Institute of Brain Sciences, Nanjing University, Nanjing 210093, China
- Correspondence: (X.-P.Z.); (W.W.)
| | - Feng Liu
- Institute of Brain Sciences, Nanjing University, Nanjing 210093, China
- National Laboratory of Solid State Microstructure, Nanjing University, Nanjing 210093, China
- Department of Physics, Nanjing University, Nanjing 210093, China
| | - Wei Wang
- Institute of Brain Sciences, Nanjing University, Nanjing 210093, China
- National Laboratory of Solid State Microstructure, Nanjing University, Nanjing 210093, China
- Department of Physics, Nanjing University, Nanjing 210093, China
- Correspondence: (X.-P.Z.); (W.W.)
| |
Collapse
|
20
|
Tsujino T, Takai T, Hinohara K, Gui F, Tsutsumi T, Bai X, Miao C, Feng C, Gui B, Sztupinszki Z, Simoneau A, Xie N, Fazli L, Dong X, Azuma H, Choudhury AD, Mouw KW, Szallasi Z, Zou L, Kibel AS, Jia L. CRISPR screens reveal genetic determinants of PARP inhibitor sensitivity and resistance in prostate cancer. Nat Commun 2023; 14:252. [PMID: 36650183 PMCID: PMC9845315 DOI: 10.1038/s41467-023-35880-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023] Open
Abstract
Prostate cancer harboring BRCA1/2 mutations are often exceptionally sensitive to PARP inhibitors. However, genomic alterations in other DNA damage response genes have not been consistently predictive of clinical response to PARP inhibition. Here, we perform genome-wide CRISPR-Cas9 knockout screens in BRCA1/2-proficient prostate cancer cells and identify previously unknown genes whose loss has a profound impact on PARP inhibitor response. Specifically, MMS22L deletion, frequently observed (up to 14%) in prostate cancer, renders cells hypersensitive to PARP inhibitors by disrupting RAD51 loading required for homologous recombination repair, although this response is TP53-dependent. Unexpectedly, loss of CHEK2 confers resistance rather than sensitivity to PARP inhibition through increased expression of BRCA2, a target of CHEK2-TP53-E2F7-mediated transcriptional repression. Combined PARP and ATR inhibition overcomes PARP inhibitor resistance caused by CHEK2 loss. Our findings may inform the use of PARP inhibitors beyond BRCA1/2-deficient tumors and support reevaluation of current biomarkers for PARP inhibition in prostate cancer.
Collapse
Affiliation(s)
- Takuya Tsujino
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Tomoaki Takai
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kunihiko Hinohara
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fu Gui
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Takeshi Tsutsumi
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Xiao Bai
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Chenkui Miao
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Chao Feng
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Bin Gui
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Zsofia Sztupinszki
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Antoine Simoneau
- Department of Pathology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA
| | - Ning Xie
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Xuesen Dong
- Vancouver Prostate Centre, Vancouver General Hospital, Vancouver, British Columbia, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Atish D Choudhury
- Department of Medical Oncology, Dana-Farber Cancer Institute & Harvard Medical School, Boston, MA, USA
| | - Kent W Mouw
- Department of Radiation Oncology, Dana-Farber Cancer Institute & Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Zoltan Szallasi
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Lee Zou
- Department of Pathology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA
| | - Adam S Kibel
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Li Jia
- Division of Urology, Department of Surgery, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Hanson RL, Batchelor E. Coordination of MAPK and p53 dynamics in the cellular responses to DNA damage and oxidative stress. Mol Syst Biol 2022; 18:e11401. [PMID: 36472304 PMCID: PMC9724178 DOI: 10.15252/msb.202211401] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022] Open
Abstract
In response to different cellular stresses, the transcription factor p53 undergoes different dynamics. p53 dynamics, in turn, control cell fate. However, distinct stresses can generate the same p53 dynamics but different cell fate outcomes, suggesting integration of dynamic information from other pathways is important for cell fate regulation. To determine how MAPK activities affect p53-mediated responses to DNA breaks and oxidative stress, we simultaneously tracked p53 and either ERK, JNK, or p38 activities in single cells. While p53 dynamics were comparable between the stresses, cell fate outcomes were distinct. Combining MAPK dynamics with p53 dynamics was important for distinguishing between the stresses and for generating temporal ordering of cell fate pathways. Furthermore, cross-talk between MAPKs and p53 controlled the balance between proliferation and cell death. These findings provide insight into how cells integrate signaling pathways with distinct temporal patterns of activity to encode stress specificity and drive different cell fate decisions.
Collapse
Affiliation(s)
- Ryan L Hanson
- Department of Integrative Biology and PhysiologyUniversity of MinnesotaMinneapolisMNUSA
| | - Eric Batchelor
- Department of Integrative Biology and PhysiologyUniversity of MinnesotaMinneapolisMNUSA
- Masonic Cancer CenterUniversity of MinnesotaMinneapolisMNUSA
| |
Collapse
|
22
|
Sampadi B, Vermeulen S, Mišovic B, Boei JJ, Batth TS, Chang JG, Paulsen MT, Magnuson B, Schimmel J, Kool H, Olie CS, Everts B, Vertegaal ACO, Olsen JV, Ljungman M, Jeggo PA, Mullenders LHF, Vrieling H. Divergent Molecular and Cellular Responses to Low and High-Dose Ionizing Radiation. Cells 2022; 11:cells11233794. [PMID: 36497055 PMCID: PMC9739411 DOI: 10.3390/cells11233794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer risk after ionizing radiation (IR) is assumed to be linear with the dose; however, for low doses, definite evidence is lacking. Here, using temporal multi-omic systems analyses after a low (LD; 0.1 Gy) or a high (HD; 1 Gy) dose of X-rays, we show that, although the DNA damage response (DDR) displayed dose proportionality, many other molecular and cellular responses did not. Phosphoproteomics uncovered a novel mode of phospho-signaling via S12-PPP1R7, and large-scale dephosphorylation events that regulate mitotic exit control in undamaged cells and the G2/M checkpoint upon IR in a dose-dependent manner. The phosphoproteomics of irradiated DNA double-strand breaks (DSBs) repair-deficient cells unveiled extended phospho-signaling duration in either a dose-dependent (DDR signaling) or independent (mTOR-ERK-MAPK signaling) manner without affecting signal magnitude. Nascent transcriptomics revealed the transcriptional activation of genes involved in NRF2-regulated antioxidant defense, redox-sensitive ERK-MAPK signaling, glycolysis and mitochondrial function after LD, suggesting a prominent role for reactive oxygen species (ROS) in molecular and cellular responses to LD exposure, whereas DDR genes were prominently activated after HD. However, how and to what extent the observed dose-dependent differences in molecular and cellular responses may impact cancer development remain unclear, as the induction of chromosomal damage was found to be dose-proportional (10-200 mGy).
Collapse
Affiliation(s)
- Bharath Sampadi
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
- Correspondence: (B.S.); (H.V.)
| | - Sylvia Vermeulen
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Branislav Mišovic
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Jan J. Boei
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Tanveer S. Batth
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Science, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Jer-Gung Chang
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Michelle T. Paulsen
- Department of Radiation Oncology, Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brian Magnuson
- Department of Radiation Oncology, Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Joost Schimmel
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Hanneke Kool
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Cyriel S. Olie
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands
| | - Alfred C. O. Vertegaal
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Jesper V. Olsen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Science, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Mats Ljungman
- Department of Radiation Oncology, Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Penny A. Jeggo
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN1 9RQ, UK
| | - Leon H. F. Mullenders
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya 464-8601, Japan
| | - Harry Vrieling
- Department of Human Genetics, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
- Correspondence: (B.S.); (H.V.)
| |
Collapse
|
23
|
Chen Z, Cui L, Xu L, Liu Z, Liang Y, Li X, Zhang Y, Li Y, Liu S, Li H. Characterization of chicken p53 transcriptional function via parallel genome-wide chromatin occupancy and gene expression analysis. Poult Sci 2022; 101:102164. [PMID: 36167023 PMCID: PMC9513273 DOI: 10.1016/j.psj.2022.102164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/14/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022] Open
Abstract
The tumor suppressor p53, which acts primarily as a transcription factor, can regulate infections from various viruses in chickens. However, the underlying mechanisms of the antiviral functions of chicken p53 (chp53) remain unclear due to the lack of detailed information on its transcriptional regulation. Here, to gain comprehensive insights into chp53 transcriptional regulatory function in a global and unbiased manner, we determined the genome-wide chromatin occupancy of chp53 by chromatin immunoprecipitation, which was followed by sequencing and chp53-mediated gene expression profile by RNA sequencing using chemically immortalized leghorn male hepatoma (LMH) cells with ectopic expression of chp53 as the model. The integrated parallel genome-wide chromatin occupancy and gene expression analysis characterized chp53 chromatin occupancy and identified 754 direct target genes of chp53. Furthermore, functional annotation and cross-species comparative biological analyses revealed the conserved key biological functions and DNA binding motifs of p53 between chickens and humans, which may be due to the consensus amino acid sequence and structure of p53 DNA-binding domains. The present study, to our knowledge, provides the first comprehensive characterization of the chp53 transcriptional regulatory network, and can possibly help to improve our understanding of p53 transcriptional regulatory mechanisms and their antiviral functions in chickens.
Collapse
|
24
|
Maltz E, Wollman R. Quantifying the phenotypic information in mRNA abundance. Mol Syst Biol 2022; 18:e11001. [PMID: 35965452 PMCID: PMC9376724 DOI: 10.15252/msb.202211001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 11/09/2022] Open
Abstract
Quantifying the dependency between mRNA abundance and downstream cellular phenotypes is a fundamental open problem in biology. Advances in multimodal single-cell measurement technologies provide an opportunity to apply new computational frameworks to dissect the contribution of individual genes and gene combinations to a given phenotype. Using an information theory approach, we analyzed multimodal data of the expression of 83 genes in the Ca2+ signaling network and the dynamic Ca2+ response in the same cell. We found that the overall expression levels of these 83 genes explain approximately 60% of Ca2+ signal entropy. The average contribution of each single gene was 17%, revealing a large degree of redundancy between genes. Using different heuristics, we estimated the dependency between the size of a gene set and its information content, revealing that on average, a set of 53 genes contains 54% of the information about Ca2+ signaling. Our results provide the first direct quantification of information content about complex cellular phenotype that exists in mRNA abundance measurements.
Collapse
Affiliation(s)
- Evan Maltz
- Department of Chemistry and BiochemistryUCLALos AngelesCAUSA
- Institute of Quantitative and Computational BioscienceUCLALos AngelesCAUSA
| | - Roy Wollman
- Department of Chemistry and BiochemistryUCLALos AngelesCAUSA
- Institute of Quantitative and Computational BioscienceUCLALos AngelesCAUSA
- Department of Integrative Biology and PhysiologyUCLALos AngelesCAUSA
| |
Collapse
|
25
|
Venkatachalam V, Jambhekar A, Lahav G. Reading oscillatory instructions: How cells achieve time-dependent responses to oscillating transcription factors. Curr Opin Cell Biol 2022; 77:102099. [PMID: 35690043 DOI: 10.1016/j.ceb.2022.102099] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/16/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022]
Affiliation(s)
- Veena Venkatachalam
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, 210 Longwood Avenue, Boston, MA 02115, USA; Department of Radiation Oncology, Dana-Farber Brigham Cancer Center, 75 Francis St, Boston, MA 02115, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, 210 Longwood Avenue, Boston, MA 02115, USA; Ludwig Center at Harvard, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02215, USA
| | - Galit Lahav
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, 210 Longwood Avenue, Boston, MA 02115, USA; Ludwig Center at Harvard, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02215, USA.
| |
Collapse
|
26
|
Jiménez A, Lu D, Kalocsay M, Berberich MJ, Balbi P, Jambhekar A, Lahav G. Time‐series transcriptomics and proteomics reveal alternative modes to decode p53 oscillations. Mol Syst Biol 2022; 18:e10588. [PMID: 35285572 PMCID: PMC8919251 DOI: 10.15252/msb.202110588] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 12/21/2022] Open
Affiliation(s)
- Alba Jiménez
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
| | - Dan Lu
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
| | - Marian Kalocsay
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
- Laboratory of Systems Pharmacology Blavatnik Institute at Harvard Medical School Boston MA USA
| | - Matthew J Berberich
- Laboratory of Systems Pharmacology Blavatnik Institute at Harvard Medical School Boston MA USA
- Center for Protein Degradation Dana‐Farber Cancer Institute Boston MA USA
| | - Petra Balbi
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
| | - Ashwini Jambhekar
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
- Ludwig Center at Harvard Medical School Boston MA USA
| | - Galit Lahav
- Department of Systems Biology Blavatnik Institute at Harvard Medical School Boston MA USA
- Ludwig Center at Harvard Medical School Boston MA USA
| |
Collapse
|
27
|
Wu Y, Xue L, Huang W, Deng M, Lin Y. Profiling transcription factor activity dynamics using intronic reads in time-series transcriptome data. PLoS Comput Biol 2022; 18:e1009762. [PMID: 35007289 PMCID: PMC8782462 DOI: 10.1371/journal.pcbi.1009762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 01/21/2022] [Accepted: 12/15/2021] [Indexed: 11/19/2022] Open
Abstract
Activities of transcription factors (TFs) are temporally modulated to regulate dynamic cellular processes, including development, homeostasis, and disease. Recent developments of bioinformatic tools have enabled the analysis of TF activities using transcriptome data. However, because these methods typically use exon-based target expression levels, the estimated TF activities have limited temporal accuracy. To address this, we proposed a TF activity measure based on intron-level information in time-series RNA-seq data, and implemented it to decode the temporal control of TF activities during dynamic processes. We showed that TF activities inferred from intronic reads can better recapitulate instantaneous TF activities compared to the exon-based measure. By analyzing public and our own time-series transcriptome data, we found that intron-based TF activities improve the characterization of temporal phasing of cycling TFs during circadian rhythm, and facilitate the discovery of two temporally opposing TF modules during T cell activation. Collectively, we anticipate that the proposed approach would be broadly applicable for decoding global transcriptional architecture during dynamic processes.
Collapse
Affiliation(s)
- Yan Wu
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- School of Mathematical Sciences and Center for Statistical Science, Peking University, Beijing, China
| | - Lingfeng Xue
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wen Huang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Minghua Deng
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- School of Mathematical Sciences and Center for Statistical Science, Peking University, Beijing, China
| | - Yihan Lin
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- * E-mail:
| |
Collapse
|
28
|
Lahmann I, Zhang Y, Baum K, Wolf J, Birchmeier C. An oscillatory network controlling self-renewal of skeletal muscle stem cells. Exp Cell Res 2021; 409:112933. [PMID: 34793773 DOI: 10.1016/j.yexcr.2021.112933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/04/2021] [Accepted: 11/14/2021] [Indexed: 12/11/2022]
Abstract
The balance between proliferation and differentiation of muscle stem cells is tightly controlled, ensuring the maintenance of a cellular pool needed for muscle growth and repair. Muscle stem cells can proliferate, they can generate differentiating cells, or they self-renew to produce new stem cells. Notch signaling plays a crucial role in this process. Recent studies revealed that expression of the Notch effector HES1 oscillates in activated muscle stem cells. The oscillatory expression of HES1 periodically represses transcription from the genes encoding the myogenic transcription factor MYOD and the Notch ligand DLL1, thereby driving MYOD and DLL1 oscillations. This oscillatory network allows muscle progenitor cells and activated muscle stem cells to remain in a proliferative and 'undecided' state, in which they can either differentiate or self-renew. When HES1 is downregulated, MYOD oscillations become unstable and are replaced by sustained expression, which drives the cells into terminal differentiation. During development and regeneration, proliferating stem cells contact each other and the stability of the oscillatory expression depends on regular DLL1 inputs provided by neighboring cells. In such communities of cells that receive and provide Notch signals, the appropriate timing of DLL1 inputs is important, as sustained DLL1 cannot replace oscillatory DLL1. Thus, in cell communities, DLL1 oscillations ensure the appropriate balance between self-renewal and differentiation. In summary, oscillations in myogenic cells are an important example of dynamic gene expression determining cell fate.
Collapse
Affiliation(s)
- Ines Lahmann
- Neurowissenschaftliches Forschungszentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany
| | - Yao Zhang
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany
| | - Katharina Baum
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Mathematical Modelling of Cellular Processes, 13125, Berlin, Germany; New address: Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, 14482, Potsdam, Germany
| | - Jana Wolf
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Mathematical Modelling of Cellular Processes, 13125, Berlin, Germany; Free University Berlin, Department of Mathematics and Computer Science, Arnimallee 14, 14195, Berlin, Germany
| | - Carmen Birchmeier
- Neurowissenschaftliches Forschungszentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, 13125, Berlin, Germany.
| |
Collapse
|
29
|
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
|
30
|
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: 23] [Impact Index Per Article: 7.7] [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
|
31
|
Chung H, Parkhurst CN, Magee EM, Phillips D, Habibi E, Chen F, Yeung BZ, Waldman J, Artis D, Regev A. Joint single-cell measurements of nuclear proteins and RNA in vivo. Nat Methods 2021; 18:1204-1212. [PMID: 34608310 PMCID: PMC8532076 DOI: 10.1038/s41592-021-01278-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/19/2021] [Indexed: 02/08/2023]
Abstract
Identifying gene-regulatory targets of nuclear proteins in tissues is a challenge. Here we describe intranuclear cellular indexing of transcriptomes and epitopes (inCITE-seq), a scalable method that measures multiplexed intranuclear protein levels and the transcriptome in parallel across thousands of nuclei, enabling joint analysis of transcription factor (TF) levels and gene expression in vivo. We apply inCITE-seq to characterize cell state-related changes upon pharmacological induction of neuronal activity in the mouse brain. Modeling gene expression as a linear combination of quantitative protein levels revealed genome-wide associations of each TF and recovered known gene targets. TF-associated genes were coexpressed as distinct modules that each reflected positive or negative TF levels, showing that our approach can disentangle relative putative contributions of TFs to gene expression and add interpretability to inferred gene networks. inCITE-seq can illuminate how combinations of nuclear proteins shape gene expression in native tissue contexts, with direct applications to solid or frozen tissues and clinical specimens.
Collapse
Affiliation(s)
- Hattie Chung
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Christopher N Parkhurst
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Emma M Magee
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Devan Phillips
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Ehsan Habibi
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Fei Chen
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Julia Waldman
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Genentech, South San Francisco, CA, USA.
| |
Collapse
|
32
|
p53-Dependent Repression: DREAM or Reality? Cancers (Basel) 2021; 13:cancers13194850. [PMID: 34638334 PMCID: PMC8508069 DOI: 10.3390/cancers13194850] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary The tumor suppressor p53 is a complex cell signaling hub encompassing multiple transcription programs and governs a vast repertoire of biological responses. However, despite several decades of research, how p53 selects one program over another is still elusive. Recent attempts have used meta-analyses of p53 ChIP-seq data to determine the core p53 transcriptional program, conserved across different models and stimuli. This review highlights the complexity of the multiple layers of p53 regulation and the context specificity of p53 target genes. More specifically, we discuss the controversy over the mechanisms of p53-dependent transcriptional repression and its potential role in the flexibility of p53 response. Abstract p53 is a major tumor suppressor that integrates diverse types of signaling in mammalian cells. In response to a broad range of intra- or extra-cellular stimuli, p53 controls the expression of multiple target genes and elicits a vast repertoire of biological responses. The exact code by which p53 integrates the various stresses and translates them into an appropriate transcriptional response is still obscure. p53 is tightly regulated at multiple levels, leading to a wide diversity in p53 complexes on its target promoters and providing adaptability to its transcriptional program. As p53-targeted therapies are making their way into clinics, we need to understand how to direct p53 towards the desired outcome (i.e., cell death, senescence or other) selectively in cancer cells without affecting normal tissues or the immune system. While the core p53 transcriptional program has been proposed, the mechanisms conferring a cell type- and stimuli-dependent transcriptional outcome by p53 require further investigations. The mechanism by which p53 localizes to repressed promoters and manages its co-repressor interactions is controversial and remains an important gap in our understanding of the p53 cistrome. We hope that our review of the recent literature will help to stimulate the appreciation and investigation of largely unexplored p53-mediated repression.
Collapse
|
33
|
Leiz J, Rutkiewicz M, Birchmeier C, Heinemann U, Schmidt-Ott KM. Technologies for profiling the impact of genomic variants on transcription factor binding. MED GENET-BERLIN 2021; 33:147-155. [PMID: 38836027 PMCID: PMC11006259 DOI: 10.1515/medgen-2021-2073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/24/2021] [Indexed: 06/06/2024]
Abstract
Transcription factors (TFs) bind DNA in a sequence-specific manner and thereby regulate target gene expression. TF binding and its regulatory activity is highly context dependent, and is not only determined by specific cell types or differentiation stages but also relies on other regulatory mechanisms, such as DNA and chromatin modifications. Interactions between TFs and their DNA binding sites are critical mediators of phenotypic variation and play important roles in the onset of disease. A continuously growing number of studies therefore attempts to elucidate TF:DNA interactions to gain knowledge about regulatory mechanisms and disease-causing variants. Here we summarize how TF-binding characteristics and the impact of variants can be investigated, how bioinformatic tools can be used to analyze and predict TF:DNA binding, and what additional information can be obtained from the TF protein structure.
Collapse
Affiliation(s)
- Janna Leiz
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Intensive Care Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Molecular and Translational Kidney Research, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Maria Rutkiewicz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Macromolecular Structure and Interaction, Berlin, Germany
| | - Carmen Birchmeier
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology and Signal Transduction, Berlin, Germany
| | - Udo Heinemann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Macromolecular Structure and Interaction, Berlin, Germany
| | - Kai M Schmidt-Ott
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Nephrology and Intensive Care Medicine, Hindenburgdamm 30, 12203 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Molecular and Translational Kidney Research, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| |
Collapse
|
34
|
Shi X. A Hill type equation can predict target gene expression driven by p53 pulsing. FEBS Open Bio 2021; 11:1799-1808. [PMID: 33955710 PMCID: PMC8167869 DOI: 10.1002/2211-5463.13179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 11/27/2022] Open
Abstract
Many factors determine target gene expression dynamics under p53 pulsing. In this study, I sought to determine the mechanism by which duration, frequency, binding affinity and maximal transcription rate affect the expression dynamics of target genes. Using an analytical method to solve a simple model, I found that the fold change of target gene expression increases relative to the number of p53 pulses, and the optimal frequency, 0.18 h-1 , from two real p53 pulses drives the maximal fold change with a decay rate of 0.18 h-1 . Moreover, p53 pulses may also lead to a higher fold change than sustained p53. Finally, I discovered that a Hill-type equation, including these effect factors, can characterise target gene expression. The average error between the theoretical predictions and experiments was 23%. Collectively, this equation advances the understanding of transcription factor dynamics, where duration and frequency play a significant role in the fine regulation of target gene expression with higher binding affinity.
Collapse
Affiliation(s)
- Xiaomin Shi
- Department of Mathematics and International Center for Quantum and Molecular StructuresShanghai UniversityChina
| |
Collapse
|
35
|
Liebl MC, Hofmann TG. The Role of p53 Signaling in Colorectal Cancer. Cancers (Basel) 2021; 13:2125. [PMID: 33924934 PMCID: PMC8125348 DOI: 10.3390/cancers13092125] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/24/2022] Open
Abstract
The transcription factor p53 functions as a critical tumor suppressor by orchestrating a plethora of cellular responses such as DNA repair, cell cycle arrest, cellular senescence, cell death, cell differentiation, and metabolism. In unstressed cells, p53 levels are kept low due to its polyubiquitination by the E3 ubiquitin ligase MDM2. In response to various stress signals, including DNA damage and aberrant growth signals, the interaction between p53 and MDM2 is blocked and p53 becomes stabilized, allowing p53 to regulate a diverse set of cellular responses mainly through the transactivation of its target genes. The outcome of p53 activation is controlled by its dynamics, its interactions with other proteins, and post-translational modifications. Due to its involvement in several tumor-suppressing pathways, p53 function is frequently impaired in human cancers. In colorectal cancer (CRC), the TP53 gene is mutated in 43% of tumors, and the remaining tumors often have compromised p53 functioning because of alterations in the genes encoding proteins involved in p53 regulation, such as ATM (13%) or DNA-PKcs (11%). TP53 mutations in CRC are usually missense mutations that impair wild-type p53 function (loss-of-function) and that even might provide neo-morphic (gain-of-function) activities such as promoting cancer cell stemness, cell proliferation, invasion, and metastasis, thereby promoting cancer progression. Although the first compounds targeting p53 are in clinical trials, a better understanding of wild-type and mutant p53 functions will likely pave the way for novel CRC therapies.
Collapse
Affiliation(s)
- Magdalena C. Liebl
- Institute of Toxicology, University Medical Center Mainz, Johannes Gutenberg University, 55131 Mainz, Germany;
| | | |
Collapse
|
36
|
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: 1.0] [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
|
37
|
Freewoman JM, Snape R, Cui F. Temporal gene regulation by p53 is associated with the rotational setting of its binding sites in nucleosomes. Cell Cycle 2021; 20:792-807. [PMID: 33764853 PMCID: PMC8098069 DOI: 10.1080/15384101.2021.1904554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The tumor suppressor protein p53 is a DNA-binding transcription factor (TF) that, once activated, coordinates the expression of thousands of target genes. Increased p53 binding to gene promoters occurs shortly after p53 activation. Intriguingly, gene transcription exhibits differential kinetics with some genes being induced early (early genes) and others being induced late (late genes). To understand pre-binding factors contributing to the temporal gene regulation by p53, we performed time-course RNA sequencing experiments in human colon cancer cell line HCT116 treated with fluorouracil to identify early and late genes. Published p53 ChIP fragments co-localized with the early or late genes were used to uncover p53 binding sites (BS). We demonstrate that the BS associated with early genes are clustered around gene starts with decreased nucleosome occupancy. DNA analysis shows that these BS are likely exposed on nucleosomal surface if wrapped into nucleosomes, thereby facilitating stable interactions with and fast induction by p53. By contrast, p53 BS associated with late genes are distributed uniformly across the genes with increased nucleosome occupancy. Predicted rotational settings of these BS show limited accessibility. We therefore propose a hypothetical model in which the BS are fully, partially or not accessible to p53 in the nucleosomal context. The partial accessibility of the BS allows subunits of a p53 tetramer to bind, but the resulting p53-DNA complex may not be stable enough to recruit cofactors, which leads to delayed induction. Our work highlights the importance of DNA conformations of p53 BS in gene expression dynamics.
Collapse
Affiliation(s)
- Julia M Freewoman
- Thomas H. Gosnell School of Life Sciences, College of Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Rajiv Snape
- Thomas H. Gosnell School of Life Sciences, College of Science, Rochester Institute of Technology, Rochester, NY, USA
| | - Feng Cui
- Thomas H. Gosnell School of Life Sciences, College of Science, Rochester Institute of Technology, Rochester, NY, USA
| |
Collapse
|
38
|
Friedel L, Loewer A. The guardian's choice: how p53 enables context-specific decision-making in individual cells. FEBS J 2021; 289:40-52. [PMID: 33590949 DOI: 10.1111/febs.15767] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/03/2021] [Accepted: 02/15/2021] [Indexed: 01/20/2023]
Abstract
p53 plays a central role in defending the genomic integrity of our cells. In response to genotoxic stress, this tumour suppressor orchestrates the expression of hundreds of target genes, which induce a variety of cellular outcomes ranging from damage repair to induction of apoptosis. In this review, we examine how the p53 response is regulated on several levels in individual cells to allow precise and context-specific fate decisions. We discuss that the p53 response is not only controlled by its canonical regulators but also controlled by interconnected signalling pathways that influence the dynamics of p53 accumulation upon damage and modulate its transcriptional activity at target gene promoters. Additionally, we consider how the p53 response is diversified through a variety of mechanisms at the promoter level and beyond to induce context-specific outcomes in individual cells. These layers of regulation allow p53 to react in a stimulus-specific manner and fine-tune its signalling according to the individual needs of a given cell, enabling it to take the right decision on survival or death.
Collapse
Affiliation(s)
- Laura Friedel
- Systems Biology of the Stress Response, Department of Biology, Technical University of Darmstadt, Germany
| | - Alexander Loewer
- Systems Biology of the Stress Response, Department of Biology, Technical University of Darmstadt, Germany
| |
Collapse
|
39
|
Aharon-Hefetz N, Frumkin I, Mayshar Y, Dahan O, Pilpel Y, Rak R. Manipulation of the human tRNA pool reveals distinct tRNA sets that act in cellular proliferation or cell cycle arrest. eLife 2020; 9:e58461. [PMID: 33357381 PMCID: PMC7781600 DOI: 10.7554/elife.58461] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Different subsets of the tRNA pool in human cells are expressed in different cellular conditions. The 'proliferation-tRNAs' are induced upon normal and cancerous cell division, while the 'differentiation-tRNAs' are active in non-dividing, differentiated cells. Here we examine the essentiality of the various tRNAs upon cellular growth and arrest. We established a CRISPR-based editing procedure with sgRNAs that each target a tRNA family. We measured tRNA essentiality for cellular growth and found that most proliferation-tRNAs are essential compared to differentiation- tRNAs in rapidly growing cell lines. Yet in more slowly dividing lines, the differentiation-tRNAs were more essential. In addition, we measured the essentiality of each tRNA family upon response to cell cycle arresting signals. Here we detected a more complex behavior with both proliferation-tRNAs and differentiation tRNAs showing various levels of essentiality. These results provide the so-far most comprehensive functional characterization of human tRNAs with intricate roles in various cellular states.
Collapse
Affiliation(s)
- Noa Aharon-Hefetz
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Idan Frumkin
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Yoav Mayshar
- Department of Molecular Cell Biology, Weizmann Institute of ScienceRehovotIsrael
| | - Orna Dahan
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Yitzhak Pilpel
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Roni Rak
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
40
|
Liu Y, Leslie PL, Zhang Y. Life and Death Decision-Making by p53 and Implications for Cancer Immunotherapy. Trends Cancer 2020; 7:226-239. [PMID: 33199193 DOI: 10.1016/j.trecan.2020.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/11/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022]
Abstract
The tumor-suppressor protein p53 is mutated in approximately half of all cancers, whereas the p53 signaling network is perturbed in almost all cancers. In response to different stress stimuli, p53 selectively activates genes to elicit a cell survival or cell death response. How p53 makes the decision between life and death remains a fascinating question and an exciting field of research. Understanding how this decision is made has major implications for improving cancer treatments, particularly in recently evolved immune checkpoint inhibition therapy. We highlight progress and challenges in understanding the mechanisms governing the p53 life and death decision-making process, and discuss how this decision is relevant to immune system regulation. Finally, we discuss how knowledge of the p53 pro-survival and pro-death decision node can be applied to optimize immune checkpoint inhibitor therapy for cancer treatment.
Collapse
Affiliation(s)
- Yong Liu
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7461, USA; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Patrick L Leslie
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7461, USA
| | - Yanping Zhang
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7461, USA.
| |
Collapse
|
41
|
Pokrass MJ, Ryan KA, Xin T, Pielstick B, Timp W, Greco V, Regot S. Cell-Cycle-Dependent ERK Signaling Dynamics Direct Fate Specification in the Mammalian Preimplantation Embryo. Dev Cell 2020; 55:328-340.e5. [PMID: 33091369 PMCID: PMC7658051 DOI: 10.1016/j.devcel.2020.09.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/12/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
Despite the noisy nature of single cells, multicellular organisms robustly generate different cell types from one zygote. This process involves dynamic cross regulation between signaling and gene expression that is difficult to capture with fixed-cell approaches. To study signaling dynamics and fate specification during preimplantation development, we generated a transgenic mouse expressing the ERK kinase translocation reporter and measured ERK activity in single cells of live embryos. Our results show primarily active ERK in both the inner cell mass and trophectoderm cells due to fibroblast growth factor (FGF) signaling. Strikingly, a subset of mitotic events results in a short pulse of ERK inactivity in both daughter cells that correlates with elevated endpoint NANOG levels. Moreover, endogenous tagging of Nanog in embryonic stem cells reveals that ERK inhibition promotes enhanced stabilization of NANOG protein after mitosis. Our data show that cell cycle, signaling, and differentiation are coordinated during preimplantation development.
Collapse
Affiliation(s)
- Michael J Pokrass
- Department Molecular Biology and Genetics, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biochemistry, Cellular, and Molecular Biology Graduate Program, Baltimore, MD, USA
| | - Kathleen A Ryan
- Department Molecular Biology and Genetics, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tianchi Xin
- Genetics Department, Yale School of Medicine, New Haven, CT 06520, USA
| | - Brittany Pielstick
- Biochemistry, Cellular, and Molecular Biology Graduate Program, Baltimore, MD, USA; Biomedical Engineering Department, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Winston Timp
- Biomedical Engineering Department, the Johns Hopkins University, Baltimore, MD 21218, USA
| | - Valentina Greco
- Genetics Department, Yale School of Medicine, New Haven, CT 06520, USA
| | - Sergi Regot
- Department Molecular Biology and Genetics, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department Oncology, the Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
42
|
IFI16 promotes human embryonic stem cell trilineage specification through interaction with p53. NPJ Regen Med 2020; 5:18. [PMID: 33298947 PMCID: PMC7596047 DOI: 10.1038/s41536-020-00104-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/25/2020] [Indexed: 11/08/2022] Open
Abstract
Transcriptional regulation plays an essential role in the self-renewal and differentiation of human embryonic stem cells (hESCs). However, how external signals disrupt the self-renewal regulatory network and further drive hESC differentiation remains largely unknown. Here, we found the immune regulative protein, gamma-interferon-inducible protein 16 (IFI16) was involved in the regulation of both self-renewal and differentiation gene expression during hESC trilineage specification through interaction with p53. IFI16 expression levels were upregulated through JNK activation. IFI16 knockdown delayed the downregulation of self-renewal gene expression and suppressed the upregulation of differentiation gene expression, while IFI16 overexpression accelerated trilineage specification. Furthermore, IFI16 stabilized p53-binding in the genome through IFI16-p53 interaction and differentially regulated self-renewal and differentiation gene expression. Together, our results suggest a particular role of IFI16 in differential gene expression regulation during trilineage specification of hESCs in a manner that is dependent on the genome-wide profile of p53-binding directed by IFI16-p53 interaction.
Collapse
|
43
|
Xia X, Fu X, Du J, Wu B, Zhao X, Zhu J, Zhao Z. Regulation of circadian rhythm and sleep by miR-375-timeless interaction in Drosophila. FASEB J 2020; 34:16536-16551. [PMID: 33078445 DOI: 10.1096/fj.202001107r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 01/14/2023]
Abstract
MicroRNAs are important coordinators of circadian regulation that mediate the fine-tuning of gene expression. Although many studies have shown the effects of individual miRNAs on the circadian clock, the global functional miRNA-mRNA interaction network involved in the circadian system remains poorly understood. Here, we used CLEAR (Covalent Ligation of Endogenous Argonaute-bound RNAs)-CLIP (Cross-Linking and Immuno-Precipitation) to explore the regulatory functions of miRNAs in the circadian system by comparing the miRNA-mRNA interactions between Drosophila wild-type strain W1118 and a mutant of the key circadian transcriptional regulator Clock (Clkjrk ). This experimental approach unambiguously identified tens of thousands of miRNA-mRNA interactions in both the head and body. The miRNA-mRNA interactome showed dramatic changes in the Clkjrk flies. Particularly, among ~300 miRNA-mRNA circadian relevant interactions, multiple interactions involving core clock genes pdp1, tim, and vri displayed distinct changes as a result of the Clk mutation. Based on the CLEAR-CLIP analysis, we found a novel regulation of the circadian rhythm and sleep by the miR-375-timeless interaction. The results indicated that Clk disruption abolished normal rhythmic expression of miR-375 and the functional regulation occurred in the l-LNv neurons, where miR-375 modulated the circadian rhythm and sleep via targeting timeless. This work provides the first global view of miRNA regulation in the circadian rhythm.
Collapse
Affiliation(s)
- Xiju Xia
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Xiaonan Fu
- The Interdisciplinary Ph.D. Program in Genetics, Bioinformatics, and Computational Biology, Virginia Tech, Blacksburg, VA, USA
| | - Juan Du
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Binbin Wu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Xianguo Zhao
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jinsong Zhu
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, USA
| | - Zhangwu Zhao
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
44
|
Hanson RL, Batchelor E. Rucaparib Treatment Alters p53 Oscillations in Single Cells to Enhance DNA-Double-Strand-Break-Induced Cell Cycle Arrest. Cell Rep 2020; 33:108240. [PMID: 33053351 DOI: 10.1016/j.celrep.2020.108240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 08/12/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
DNA double strand breaks induce oscillatory expression of the transcription factor p53 that is dependent on ataxia telangiectasia mutated (ATM) activity and the rate of double strand break resolution. Although p53 dynamics are known to play a role in the regulation of cell fate determination, the consequences of the variability in dynamics associated with differences in repair rates and utilized repair pathways are unknown. Using single-cell time-lapse microscopy, we found that disruption of specific repair pathways has distinct impacts on p53 dynamics. The small-molecule rucaparib, an inhibitor of the alternative end-joining-associated protein poly (ADP-ribose) polymerase (PARP), increased p53 pulse duration, altering the temporal expression of multiple p53 target genes. As a result, combination treatments of the radiomimetic drug neocarzinostatin with rucaparib drove prolonged growth arrest beyond that of DNA damage alone. This study highlights how pharmacological manipulation of DNA repair pathways may be used to alter p53 dynamics to enhance therapeutic regimens.
Collapse
Affiliation(s)
- Ryan L Hanson
- Department of Integrative Biology and Physiology, University of Minnesota, Cancer and Cardiovascular Research Building 3-136, 2231 6th Street SE, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Eric Batchelor
- Department of Integrative Biology and Physiology, University of Minnesota, Cancer and Cardiovascular Research Building 3-136, 2231 6th Street SE, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
45
|
Sammons MA, Nguyen TAT, McDade SS, Fischer M. Tumor suppressor p53: from engaging DNA to target gene regulation. Nucleic Acids Res 2020; 48:8848-8869. [PMID: 32797160 PMCID: PMC7498329 DOI: 10.1093/nar/gkaa666] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 12/13/2022] Open
Abstract
The p53 transcription factor confers its potent tumor suppressor functions primarily through the regulation of a large network of target genes. The recent explosion of next generation sequencing protocols has enabled the study of the p53 gene regulatory network (GRN) and underlying mechanisms at an unprecedented depth and scale, helping us to understand precisely how p53 controls gene regulation. Here, we discuss our current understanding of where and how p53 binds to DNA and chromatin, its pioneer-like role, and how this affects gene regulation. We provide an overview of the p53 GRN and the direct and indirect mechanisms through which p53 affects gene regulation. In particular, we focus on delineating the ubiquitous and cell type-specific network of regulatory elements that p53 engages; reviewing our understanding of how, where, and when p53 binds to DNA and the mechanisms through which these events regulate transcription. Finally, we discuss the evolution of the p53 GRN and how recent work has revealed remarkable differences between vertebrates, which are of particular importance to cancer researchers using mouse models.
Collapse
Affiliation(s)
- Morgan A Sammons
- Department of Biological Sciences and The RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
| | - Thuy-Ai T Nguyen
- Genome Integrity & Structural Biology Laboratory and Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Simon S McDade
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK
| | - Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging – Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany
| |
Collapse
|
46
|
Zambrano S, Loffreda A, Carelli E, Stefanelli G, Colombo F, Bertrand E, Tacchetti C, Agresti A, Bianchi ME, Molina N, Mazza D. First Responders Shape a Prompt and Sharp NF-κB-Mediated Transcriptional Response to TNF-α. iScience 2020; 23:101529. [PMID: 33083759 PMCID: PMC7509218 DOI: 10.1016/j.isci.2020.101529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/05/2020] [Accepted: 09/01/2020] [Indexed: 12/01/2022] Open
Abstract
Nuclear factor (NF)-κB controls the transcriptional response to inflammatory signals by translocating into the nucleus, but we lack a single-cell characterization of the resulting transcription dynamics. Here we show that upon tumor necrosis factor (TNF)-α transcription of NF-κB target genes is heterogeneous in individual cells but results in an average nascent transcription profile that is prompt (i.e., occurs almost immediately) and sharp (i.e., increases and decreases rapidly) compared with NF-κB nuclear localization. Using an NF-κB-controlled MS2 reporter we show that the single-cell nascent transcription is more heterogeneous than NF-κB translocation dynamics, with a fraction of synchronized “first responders” that shape the average transcriptional profile and are more prone to respond to multiple TNF-α stimulations. A mathematical model combining NF-κB-mediated gene activation and a gene refractory state is able to reproduce these features. Our work shows how the expression of target genes induced by transcriptional activators can be heterogeneous across single cells and yet time resolved on average. Nascent transcription upon TNF-α is heterogeneous, with a subset of “first responders” The average nascent transcription is prompt and sharper than NF-κB response First responders do not depend on NF-κB dynamics and respond more to pulsed stimuli A model including NF-κB and a gene refractory state reproduces these observations
Collapse
Affiliation(s)
- Samuel Zambrano
- School of Medicine, Vita-Salute San Raffaele University, Milan 20132, Italy.,Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Alessia Loffreda
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Elena Carelli
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Giacomo Stefanelli
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Federica Colombo
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy.,Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan 20133, Italy
| | - Edouard Bertrand
- Institut de Génétique Moléculaire de Montpellier, CNRS, Montpellier 34293, France
| | - Carlo Tacchetti
- School of Medicine, Vita-Salute San Raffaele University, Milan 20132, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Alessandra Agresti
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Marco E Bianchi
- School of Medicine, Vita-Salute San Raffaele University, Milan 20132, Italy.,Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Nacho Molina
- Institut de Génétique et Biologie Moléculaire Cellulaire, Illkirch-Graffenstaden 67404, France
| | - Davide Mazza
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| |
Collapse
|
47
|
Zhang Y, Liu H, Li Z, Miao Z, Zhou J. Oscillatory Dynamics of p53-Mdm2 Circuit in Response to DNA Damage Caused by Ionizing Radiation. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2020; 17:1703-1713. [PMID: 30762566 DOI: 10.1109/tcbb.2019.2899574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Although the dynamical behavior of the p53-Mdm2 loop has been extensively studied, the understanding of the mechanism underlying the regulation of this pathway still remains limited. Herein, we developed an integrated model with five basic components and three ubiquitous time delays for the p53-Mdm2 interaction in response to DNA damage following ionizing radiation (IR). We showed that a sufficient amount of activated ATM level can initiate the p53 oscillations with nearly the same amplitude over a wide range of the ATM level; a proper range of p53 level is also required for generating the oscillations, for too high or too low levels it would fail to generate the oscillations; and increased Mdm2 level leads to decreased amplitude of the p53 oscillation and reduced expression of the p53 activity. Moreover, we found that the negative feedback loop formed between p53 and nuclear Mdm2 plays a dominant role in determining the p53 dynamics, whereas when interaction strength of the negative feedback loop becomes weaker, the positive feedback loop formed between p53 and cytoplasmatic Mdm2 can induce different types of dynamics. Furthermore, we demonstrated that the total time delay required for protein production and nuclear translocation of Mdm2 can induce p53 oscillations even when the p53 level is at a certain stable high steady state or at a certain stable low steady state. In addition, the two important features of the oscillatory dynamics-amplitude and period-can be controlled by such time delay. These results are in agreement with multiple experimental observations and may enrich our understanding of the dynamics of the p53 network.
Collapse
|
48
|
Suarez OJ, Vega CJ, Sanchez EN, González-Santiago AE, Rodríguez-Jorge O, Alanis AY, Chen G, Hernandez-Vargas EA. Pinning Control for the p53-Mdm2 Network Dynamics Regulated by p14ARF. Front Physiol 2020; 11:976. [PMID: 32982771 PMCID: PMC7485292 DOI: 10.3389/fphys.2020.00976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 07/17/2020] [Indexed: 01/26/2023] Open
Abstract
p53 regulates the cellular response to genotoxic damage and prevents carcinogenic events. Theoretical and experimental studies state that the p53-Mdm2 network constitutes the core module of regulatory interactions activated by cellular stress induced by a variety of signaling pathways. In this paper, a strategy to control the p53-Mdm2 network regulated by p14ARF is developed, based on the pinning control technique, which consists into applying local feedback controllers to a small number of nodes (pinned ones) in the network. Pinned nodes are selected on the basis of their importance level in a topological hierarchy, their degree of connectivity within the network, and the biological role they perform. In this paper, two cases are considered. For the first case, the oscillatory pattern under gamma-radiation is recovered; afterward, as the second case, increased expression of p53 level is taken into account. For both cases, the control law is applied to p14ARF (pinned node based on a virtual leader methodology), and overexpressed Mdm2-mediated p53 degradation condition is considered as carcinogenic initial behavior. The approach in this paper uses a computational algorithm, which opens an alternative path to understand the cellular responses to stress, doing it possible to model and control the gene regulatory network dynamics in two different biological contexts. As the main result of the proposed control technique, the two mentioned desired behaviors are obtained.
Collapse
Affiliation(s)
- Oscar J. Suarez
- Electrical Engineering Department, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Guadalajara, Mexico
| | - Carlos J. Vega
- Electrical Engineering Department, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Guadalajara, Mexico
| | - Edgar N. Sanchez
- Electrical Engineering Department, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Guadalajara, Mexico
| | - Ana E. González-Santiago
- Biomedical Sciences Department, Centro de Investigación Multidisciplinario en Salud, Universidad de Guadalajara, Tonalá, Mexico
| | - Otoniel Rodríguez-Jorge
- Biochemistry and Molecular Biology Department, Instituto de Investigaciones Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Alma Y. Alanis
- Computer Sciences Department, Universidad de Guadalajara, Guadalajara, Mexico
| | - Guanrong Chen
- Electrical Engineering Department, City University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
49
|
Tsabar M, Mock CS, Venkatachalam V, Reyes J, Karhohs KW, Oliver TG, Regev A, Jambhekar A, Lahav G. A Switch in p53 Dynamics Marks Cells That Escape from DSB-Induced Cell Cycle Arrest. Cell Rep 2020; 32:107995. [PMID: 32755587 PMCID: PMC7521664 DOI: 10.1016/j.celrep.2020.107995] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 05/21/2020] [Accepted: 07/14/2020] [Indexed: 01/01/2023] Open
Abstract
Cellular responses to stimuli can evolve over time, resulting in distinct early and late phases in response to a single signal. DNA damage induces a complex response that is largely orchestrated by the transcription factor p53, whose dynamics influence whether a damaged cell will arrest and repair the damage or will initiate cell death. How p53 responses and cellular outcomes evolve in the presence of continuous DNA damage remains unknown. Here, we have found that a subset of cells switches from oscillating to sustained p53 dynamics several days after undergoing damage. The switch results from cell cycle progression in the presence of damaged DNA, which activates the caspase-2-PIDDosome, a complex that stabilizes p53 by inactivating its negative regulator MDM2. This work defines a molecular pathway that is activated if the canonical checkpoints fail to halt mitosis in the presence of damaged DNA.
Collapse
Affiliation(s)
- Michael Tsabar
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Caroline S Mock
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Veena Venkatachalam
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Harvard Radiation Oncology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Jose Reyes
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kyle W Karhohs
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Trudy G Oliver
- Huntsman Cancer Institute at University of Utah, Salt Lake City, UT 84112, USA
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ashwini Jambhekar
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Galit Lahav
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Finkle JD, Bagheri N. Hybrid analysis of gene dynamics predicts context-specific expression and offers regulatory insights. Bioinformatics 2020; 35:4671-4678. [PMID: 30994899 PMCID: PMC6853664 DOI: 10.1093/bioinformatics/btz256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/07/2019] [Accepted: 04/11/2019] [Indexed: 12/02/2022] Open
Abstract
Motivation To understand the regulatory pathways underlying diseases, studies often investigate the differential gene expression between genetically or chemically differing cell populations. Differential expression analysis identifies global changes in transcription and enables the inference of functional roles of applied perturbations. This approach has transformed the discovery of genetic drivers of disease and possible therapies. However, differential expression analysis does not provide quantitative predictions of gene expression in untested conditions. We present a hybrid approach, termed Differential Expression in Python (DiffExPy), that uniquely combines discrete, differential expression analysis with in silico differential equation simulations to yield accurate, quantitative predictions of gene expression from time-series data. Results To demonstrate the distinct insight provided by DiffExpy, we applied it to published, in vitro, time-series RNA-seq data from several genetic PI3K/PTEN variants of MCF10a cells stimulated with epidermal growth factor. DiffExPy proposed ensembles of several minimal differential equation systems for each differentially expressed gene. These systems provide quantitative models of expression for several previously uncharacterized genes and uncover new regulation by the PI3K/PTEN pathways. We validated model predictions on expression data from conditions that were not used for model training. Our discrete, differential expression analysis also identified SUZ12 and FOXA1 as possible regulators of specific groups of genes that exhibit late changes in expression. Our work reveals how DiffExPy generates quantitatively predictive models with testable, biological hypotheses from time-series expression data. Availability and implementation DiffExPy is available on GitHub (https://github.com/bagherilab/diffexpy). Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Justin D Finkle
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Neda Bagheri
- Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA.,Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, USA.,Center for Synthetic Biology, Northwestern University, Evanston, IL 60208, USA.,Chemistry of Life Processes, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|