Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.629
Filter
Add more filters

Publication year range
1.
Cell ; 186(3): 528-542.e14, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36681079

ABSTRACT

Whole-genome duplication (WGD) is a frequent event in cancer evolution and an important driver of aneuploidy. The role of the p53 tumor suppressor in WGD has been enigmatic: p53 can block the proliferation of tetraploid cells, acting as a barrier to WGD, but can also promote mitotic bypass, a key step in WGD via endoreduplication. In wild-type (WT) p53 tumors, WGD is frequently associated with activation of the E2F pathway, especially amplification of CCNE1, encoding cyclin E1. Here, we show that elevated cyclin E1 expression causes replicative stress, which activates ATR- and Chk1-dependent G2 phase arrest. p53, via its downstream target p21, together with Wee1, then inhibits mitotic cyclin-dependent kinase activity sufficiently to activate APC/CCdh1 and promote mitotic bypass. Cyclin E expression suppresses p53-dependent senescence after mitotic bypass, allowing cells to complete endoreduplication. Our results indicate that p53 can contribute to cancer evolution through the promotion of WGD.


Subject(s)
Cyclin E , Gene Duplication , Neoplasms , Tumor Suppressor Protein p53 , Humans , Cell Line, Tumor , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Mitosis , Neoplasms/genetics , Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism
2.
Cell ; 178(2): 267-269, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31299197

ABSTRACT

Time-lapse imaging reveals a nuanced role for p21 in cancer cells challenged with chemotherapeutic drugs: cells with either high or low p21 are biased toward senescence, whereas intermediate p21 allows cells to re-enter the cell cycle after drug treatment.


Subject(s)
Cellular Senescence , Cell Cycle , Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21
3.
Cell ; 178(2): 361-373.e12, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31204100

ABSTRACT

Chemotherapy is designed to induce cell death. However, at non-lethal doses, cancer cells can choose to remain proliferative or become senescent. The slow development of senescence makes studying this decision challenging. Here, by analyzing single-cell p21 dynamics before, during, and days after drug treatment, we link three distinct patterns of early p21 dynamics to final cell fate. Surprisingly, while high p21 expression is classically associated with senescence, we find the opposite at early times during drug treatment: most senescence-fated cells express much lower p21 levels than proliferation-fated cells. We demonstrate that these dynamics lead to a p21 "Goldilocks zone" for proliferation, in which modest increases of p21 expression can lead to an undesirable increase of cancer cell proliferation. Our study identifies a counter-intuitive role for early p21 dynamics in the cell-fate decision and pinpoints a source of proliferative cancer cells that can emerge after exposure to non-lethal doses of chemotherapy.


Subject(s)
Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Doxorubicin/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/drug effects , Humans , Models, Biological , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism
4.
Mol Cell ; 84(7): 1224-1242.e13, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38458201

ABSTRACT

Although mismatch repair (MMR) is essential for correcting DNA replication errors, it can also recognize other lesions, such as oxidized bases. In G0 and G1, MMR is kept in check through unknown mechanisms as it is error-prone during these cell cycle phases. We show that in mammalian cells, D-type cyclins are recruited to sites of oxidative DNA damage in a PCNA- and p21-dependent manner. D-type cyclins inhibit the proteasomal degradation of p21, which competes with MMR proteins for binding to PCNA, thereby inhibiting MMR. The ability of D-type cyclins to limit MMR is CDK4- and CDK6-independent and is conserved in G0 and G1. At the G1/S transition, the timely, cullin-RING ubiquitin ligase (CRL)-dependent degradation of D-type cyclins and p21 enables MMR activity to efficiently repair DNA replication errors. Persistent expression of D-type cyclins during S-phase inhibits the binding of MMR proteins to PCNA, increases the mutational burden, and promotes microsatellite instability.


Subject(s)
Cyclins , DNA Mismatch Repair , Animals , Cyclins/genetics , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Interphase , Mammals/metabolism
5.
Mol Cell ; 83(22): 4062-4077.e5, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977118

ABSTRACT

Abnormal increases in cell size are associated with senescence and cell cycle exit. The mechanisms by which overgrowth primes cells to withdraw from the cell cycle remain unknown. We address this question using CDK4/6 inhibitors, which arrest cells in G0/G1 and are licensed to treat advanced HR+/HER2- breast cancer. We demonstrate that CDK4/6-inhibited cells overgrow during G0/G1, causing p38/p53/p21-dependent cell cycle withdrawal. Cell cycle withdrawal is triggered by biphasic p21 induction. The first p21 wave is caused by osmotic stress, leading to p38- and size-dependent accumulation of p21. CDK4/6 inhibitor washout results in some cells entering S-phase. Overgrown cells experience replication stress, resulting in a second p21 wave that promotes cell cycle withdrawal from G2 or the subsequent G1. We propose that the levels of p21 integrate signals from overgrowth-triggered stresses to determine cell fate. This model explains how hypertrophy can drive senescence and why CDK4/6 inhibitors have long-lasting effects in patients.


Subject(s)
Tumor Suppressor Protein p53 , Humans , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cell Cycle , Cell Division , Tumor Suppressor Protein p53/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism
6.
Mol Cell ; 83(22): 4047-4061.e6, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977117

ABSTRACT

CDK4/6 inhibitors are remarkable anti-cancer drugs that can arrest tumor cells in G1 and induce their senescence while causing only relatively mild toxicities in healthy tissues. How they achieve this mechanistically is unclear. We show here that tumor cells are specifically vulnerable to CDK4/6 inhibition because during the G1 arrest, oncogenic signals drive toxic cell overgrowth. This overgrowth causes permanent cell cycle withdrawal by either preventing progression from G1 or inducing genotoxic damage during the subsequent S-phase and mitosis. Inhibiting or reverting oncogenic signals that converge onto mTOR can rescue this excessive growth, DNA damage, and cell cycle exit in cancer cells. Conversely, inducing oncogenic signals in non-transformed cells can drive these toxic phenotypes and sensitize the cells to CDK4/6 inhibition. Together, this demonstrates that cell cycle arrest and oncogenic cell growth is a synthetic lethal combination that is exploited by CDK4/6 inhibitors to induce tumor-specific toxicity.


Subject(s)
Antineoplastic Agents , Neoplasms , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G1 Phase Cell Cycle Checkpoints , Tumor Suppressor Protein p53/genetics , Cell Cycle , Cell Cycle Proteins/metabolism , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics
7.
Nature ; 619(7968): 167-175, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37344586

ABSTRACT

Healthy skin is a mosaic of wild-type and mutant clones1,2. Although injury can cooperate with mutated Ras family proteins to promote tumorigenesis3-12, the consequences in genetically mosaic skin are unknown. Here we show that after injury, wild-type cells suppress aberrant growth induced by oncogenic Ras. HrasG12V/+ and KrasG12D/+ cells outcompete wild-type cells in uninjured, mosaic tissue but their expansion is prevented after injury owing to an increase in the fraction of proliferating wild-type cells. Mechanistically, we show that, unlike HrasG12V/+ cells, wild-type cells respond to autocrine and paracrine secretion of EGFR ligands, and this differential activation of the EGFR pathway explains the competitive switch during injury repair. Inhibition of EGFR signalling via drug or genetic approaches diminishes the proportion of dividing wild-type cells after injury, leading to the expansion of HrasG12V/+ cells. Increased proliferation of wild-type cells via constitutive loss of the cell cycle inhibitor p21 counteracts the expansion of HrasG12V/+ cells even in the absence of injury. Thus, injury has a role in switching the competitive balance between oncogenic and wild-type cells in genetically mosaic skin.


Subject(s)
Cell Proliferation , Genes, ras , Mosaicism , Mutation , Skin , ras Proteins , Cell Cycle , Cell Proliferation/genetics , ErbB Receptors/metabolism , ras Proteins/genetics , ras Proteins/metabolism , Skin/cytology , Skin/injuries , Skin/metabolism , Skin/pathology , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism
8.
Genes Dev ; 35(5-6): 379-391, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33602872

ABSTRACT

Senescence is a key barrier to neoplastic transformation. To identify senescence regulators relevant to cancer, we screened a genome-wide shRNA library. Here, we describe exportin 7 (XPO7) as a novel regulator of senescence and validate its function in telomere-induced, replicative, and oncogene-induced senescence (OIS). XPO7 is a bidirectional transporter that regulates the nuclear-cytoplasmic shuttling of a broad range of substrates. Depletion of XPO7 results in reduced levels of TCF3 and an impaired induction of the cyclin-dependent kinase inhibitor p21CIP1 during OIS. Deletion of XPO7 correlates with poorer overall survival in several cancer types. Moreover, depletion of XPO7 alleviated OIS and increased tumor formation in a mouse model of liver cancer. Our results suggest that XPO7 is a novel tumor suppressor that regulates p21CIP1 expression to control senescence and tumorigenesis.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Karyopherins/genetics , Karyopherins/metabolism , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Humans , Mice , Neoplasms/physiopathology , Telomeric Repeat Binding Protein 2/genetics
9.
Cell ; 155(2): 369-83, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24075009

ABSTRACT

Tissue homeostasis in metazoans is regulated by transitions of cells between quiescence and proliferation. The hallmark of proliferating populations is progression through the cell cycle, which is driven by cyclin-dependent kinase (CDK) activity. Here, we introduce a live-cell sensor for CDK2 activity and unexpectedly found that proliferating cells bifurcate into two populations as they exit mitosis. Many cells immediately commit to the next cell cycle by building up CDK2 activity from an intermediate level, while other cells lack CDK2 activity and enter a transient state of quiescence. This bifurcation is directly controlled by the CDK inhibitor p21 and is regulated by mitogens during a restriction window at the end of the previous cell cycle. Thus, cells decide at the end of mitosis to either start the next cell cycle by immediately building up CDK2 activity or to enter a transient G0-like state by suppressing CDK2 activity.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Mitosis , 3T3 Cells , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Mice , Retinoblastoma Protein/metabolism
10.
Cell ; 155(5): 1104-18, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24238962

ABSTRACT

Cellular senescence disables proliferation in damaged cells, and it is relevant for cancer and aging. Here, we show that senescence occurs during mammalian embryonic development at multiple locations, including the mesonephros and the endolymphatic sac of the inner ear, which we have analyzed in detail. Mechanistically, senescence in both structures is strictly dependent on p21, but independent of DNA damage, p53, or other cell-cycle inhibitors, and it is regulated by the TGF-ß/SMAD and PI3K/FOXO pathways. Developmentally programmed senescence is followed by macrophage infiltration, clearance of senescent cells, and tissue remodeling. Loss of senescence due to the absence of p21 is partially compensated by apoptosis but still results in detectable developmental abnormalities. Importantly, the mesonephros and endolymphatic sac of human embryos also show evidence of senescence. We conclude that the role of developmentally programmed senescence is to promote tissue remodeling and propose that this is the evolutionary origin of damage-induced senescence.


Subject(s)
Cellular Senescence , Embryonic Development , Endolymphatic Sac/embryology , Mesonephros/embryology , Animals , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endolymphatic Sac/cytology , Female , Humans , Kidney/embryology , Male , Mesonephros/cytology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
11.
Cell ; 155(5): 1119-30, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24238961

ABSTRACT

Senescence is a form of cell-cycle arrest linked to tumor suppression and aging. However, it remains controversial and has not been documented in nonpathologic states. Here we describe senescence as a normal developmental mechanism found throughout the embryo, including the apical ectodermal ridge (AER) and the neural roof plate, two signaling centers in embryonic patterning. Embryonic senescent cells are nonproliferative and share features with oncogene-induced senescence (OIS), including expression of p21, p15, and mediators of the senescence-associated secretory phenotype (SASP). Interestingly, mice deficient in p21 have defects in embryonic senescence, AER maintenance, and patterning. Surprisingly, the underlying mesenchyme was identified as a source for senescence instruction in the AER, whereas the ultimate fate of these senescent cells is apoptosis and macrophage-mediated clearance. We propose that senescence is a normal programmed mechanism that plays instructive roles in development, and that OIS is an evolutionarily adapted reactivation of a developmental process.


Subject(s)
Cellular Senescence , Embryonic Development , Animals , Apoptosis , Chick Embryo , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/immunology , Embryo, Mammalian/metabolism , Extremities/embryology , Fibroblasts/cytology , Humans , Mice , Paracrine Communication
12.
Cell ; 154(2): 297-310, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23870121

ABSTRACT

The H3K4me3 mark in chromatin is closely correlated with actively transcribed genes, although the mechanisms involved in its generation and function are not fully understood. In vitro studies with recombinant chromatin and purified human factors demonstrate a robust SET1 complex (SET1C)-mediated H3K4 trimethylation that is dependent upon p53- and p300-mediated H3 acetylation, a corresponding SET1C-mediated enhancement of p53- and p300-dependent transcription that reflects a primary effect of SET1C through H3K4 trimethylation, and direct SET1C-p53 and SET1C-p300 interactions indicative of a targeted recruitment mechanism. Complementary cell-based assays demonstrate a DNA-damage-induced p53-SET1C interaction, a corresponding enrichment of SET1C and H3K4me3 on a p53 target gene (p21/WAF1), and a corresponding codependency of H3K4 trimethylation and transcription upon p300 and SET1C. These results establish a mechanism in which SET1C and p300 act cooperatively, through direct interactions and coupled histone modifications, to facilitate the function of p53.


Subject(s)
E1A-Associated p300 Protein/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Acetylation , Amino Acid Sequence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , HCT116 Cells , Histone Code , Histones/metabolism , Humans , Methylation , Molecular Sequence Data , Multiprotein Complexes/metabolism , Transcription, Genetic
13.
Genes Dev ; 34(7-8): 489-494, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32139422

ABSTRACT

Young mammals possess a limited regenerative capacity in some tissues, which is lost upon maturation. We investigated whether cellular senescence might play a role in such loss during liver regeneration. We found that following partial hepatectomy, the senescence-associated genes p21, p16Ink4a, and p19Arf become dynamically expressed in different cell types when regenerative capacity decreases, but without a full senescent response. However, we show that treatment with a senescence-inhibiting drug improves regeneration, by disrupting aberrantly prolonged p21 expression. This work suggests that senescence may initially develop from heterogeneous cellular responses, and that senotherapeutic drugs might be useful in promoting organ regeneration.


Subject(s)
Biphenyl Compounds/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation/drug effects , Liver/physiology , Nitrophenols/pharmacology , Regeneration/drug effects , Sulfonamides/pharmacology , Animals , Cells, Cultured , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Female , Male , Mice , Mice, Inbred C57BL , Models, Animal , Piperazines/pharmacology
14.
Nat Immunol ; 16(10): 1060-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26343536

ABSTRACT

Treatment with ionizing radiation (IR) can lead to the accumulation of tumor-infiltrating regulatory T cells (Treg cells) and subsequent resistance of tumors to radiotherapy. Here we focused on the contribution of the epidermal mononuclear phagocytes Langerhans cells (LCs) to this phenomenon because of their ability to resist depletion by high-dose IR. We found that LCs resisted apoptosis and rapidly repaired DNA damage after exposure to IR. In particular, we found that the cyclin-dependent kinase inhibitor CDKN1A (p21) was overexpressed in LCs and that Cdkn1a(-/-) LCs underwent apoptosis and accumulated DNA damage following IR treatment. Wild-type LCs upregulated major histocompatibility complex class II molecules, migrated to the draining lymph nodes and induced an increase in Treg cell numbers upon exposure to IR, but Cdkn1a(-/-) LCs did not. Our findings suggest a means for manipulating the resistance of LCs to IR to enhance the response of cutaneous tumors to radiotherapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Langerhans Cells/radiation effects , Radiation, Ionizing , T-Lymphocytes, Regulatory/radiation effects , Animals , Cell Survival/genetics , Cell Survival/radiation effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Flow Cytometry , Mice , Microarray Analysis , Polymerase Chain Reaction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Up-Regulation
15.
Nat Immunol ; 15(6): 571-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24777532

ABSTRACT

Intestinal regulatory T cells (Treg cells) are necessary for the suppression of excessive immune responses to commensal bacteria. However, the molecular machinery that controls the homeostasis of intestinal Treg cells has remained largely unknown. Here we report that colonization of germ-free mice with gut microbiota upregulated expression of the DNA-methylation adaptor Uhrf1 in Treg cells. Mice with T cell-specific deficiency in Uhrf1 (Uhrf1(fl/fl)Cd4-Cre mice) showed defective proliferation and functional maturation of colonic Treg cells. Uhrf1 deficiency resulted in derepression of the gene (Cdkn1a) that encodes the cyclin-dependent kinase inhibitor p21 due to hypomethylation of its promoter region, which resulted in cell-cycle arrest of Treg cells. As a consequence, Uhrf1(fl/fl)Cd4-Cre mice spontaneously developed severe colitis. Thus, Uhrf1-dependent epigenetic silencing of Cdkn1a was required for the maintenance of gut immunological homeostasis. This mechanism enforces symbiotic host-microbe interactions without an inflammatory response.


Subject(s)
Colitis/immunology , Colon/immunology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Epigenesis, Genetic , Nuclear Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CCAAT-Enhancer-Binding Proteins , Cell Cycle Checkpoints , Cell Proliferation , Cells, Cultured , Clostridium/immunology , Colitis/genetics , Colon/microbiology , DNA Methylation , Gene Expression Profiling , Interleukin-2 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microbiota/immunology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering , Symbiosis/immunology , Ubiquitin-Protein Ligases , Up-Regulation
16.
EMBO Rep ; 25(5): 2418-2440, 2024 May.
Article in English | MEDLINE | ID: mdl-38605277

ABSTRACT

Microcephaly is a common feature in inherited bone marrow failure syndromes, prompting investigations into shared pathways between neurogenesis and hematopoiesis. To understand this association, we studied the role of the microcephaly gene Mcph1 in hematological development. Our research revealed that Mcph1-knockout mice exhibited congenital macrocytic anemia due to impaired terminal erythroid differentiation during fetal development. Anemia's cause is a failure to complete cell division, evident from tetraploid erythroid progenitors with DNA content exceeding 4n. Gene expression profiling demonstrated activation of the p53 pathway in Mcph1-deficient erythroid precursors, leading to overexpression of Cdkn1a/p21, a major mediator of p53-dependent cell cycle arrest. Surprisingly, fetal brain analysis revealed hypertrophied binucleated neuroprogenitors overexpressing p21 in Mcph1-knockout mice, indicating a shared pathophysiological mechanism underlying both erythroid and neurological defects. However, inactivating p53 in Mcph1-/- mice failed to reverse anemia and microcephaly, suggesting that p53 activation in Mcph1-deficient cells resulted from their proliferation defect rather than causing it. These findings shed new light on Mcph1's function in fetal hematopoietic development, emphasizing the impact of disrupted cell division on neurogenesis and erythropoiesis - a common limiting pathway.


Subject(s)
Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p21 , Erythropoiesis , Mice, Knockout , Microcephaly , Tumor Suppressor Protein p53 , Animals , Mice , Anemia, Macrocytic/genetics , Anemia, Macrocytic/pathology , Anemia, Macrocytic/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Microcephaly/genetics , Microcephaly/pathology , Mutation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Mol Cell ; 71(4): 581-591.e5, 2018 08 16.
Article in English | MEDLINE | ID: mdl-30057196

ABSTRACT

Biological signals need to be robust and filter small fluctuations yet maintain sensitivity to signals across a wide range of magnitudes. Here, we studied how fluctuations in DNA damage signaling relate to maintenance of long-term cell-cycle arrest. Using live-cell imaging, we quantified division profiles of individual human cells in the course of 1 week after irradiation. We found a subset of cells that initially establish cell-cycle arrest and then sporadically escape and divide. Using fluorescent reporters and mathematical modeling, we determined that fluctuations in the oscillatory pattern of the tumor suppressor p53 trigger a sharp switch between p21 and CDK2, leading to escape from arrest. Transient perturbation of p53 stability mimicked the noise in individual cells and was sufficient to trigger escape from arrest. Our results show that the self-reinforcing circuitry that mediates cell-cycle transitions can translate small fluctuations in p53 signaling into large phenotypic changes.


Subject(s)
Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/metabolism , Models, Statistical , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/radiation effects , Cell Division/radiation effects , Cell Line, Transformed , Cell Proliferation/radiation effects , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage , Epithelial Cells/cytology , Epithelial Cells/radiation effects , Gamma Rays , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Protein Stability , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/radiation effects , Time-Lapse Imaging , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Red Fluorescent Protein
18.
Nucleic Acids Res ; 52(12): 6945-6963, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38783095

ABSTRACT

Cellular senescence, a major driver of aging, can be stimulated by DNA damage, and is counteracted by the DNA repair machinery. Here we show that in p16INK4a-deficient cells, senescence induction by the environmental genotoxin B[a]P or ionizing radiation (IR) completely depends on p21CIP1. Immunoprecipitation-based mass spectrometry interactomics data revealed that during senescence induction and maintenance, p21CIP1 specifically inhibits CDK4 and thereby activates the DREAM complex. Genome-wide transcriptomics revealed striking similarities in the response induced by B[a]P and IR. Among the top 100 repressed genes 78 were identical between B[a]P and IR and 76 were DREAM targets. The DREAM complex transcriptionally silences the main proliferation-associated transcription factors E2F1, FOXM1 and B-Myb as well as multiple DNA repair factors. Knockdown of p21CIP1, E2F4 or E2F5 diminished both, repression of these factors and senescence. The transcriptional profiles evoked by B[a]P and IR largely overlapped with the profile induced by pharmacological CDK4 inhibition, further illustrating the role of CDK4 inhibition in genotoxic stress-induced senescence. Moreover, data obtained by live-cell time-lapse microscopy suggest the inhibition of CDK4 by p21CIP1 is especially important for arresting cells which slip through mitosis. Overall, we identified the p21CIP1/CDK4/DREAM axis as a master regulator of genotoxic stress-induced senescence.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , DNA Damage , Kv Channel-Interacting Proteins , Cellular Senescence/radiation effects , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 4/genetics , Humans , Kv Channel-Interacting Proteins/metabolism , Kv Channel-Interacting Proteins/genetics , Radiation, Ionizing , DNA Repair , Gene Expression Regulation/radiation effects , Repressor Proteins
19.
Nucleic Acids Res ; 52(7): 3778-3793, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38348929

ABSTRACT

DNA replication stress, caused by various endogenous and exogenous agents, halt or stall DNA replication progression. Cells have developed diverse mechanisms to tolerate and overcome replication stress, enabling them to continue replication. One effective strategy to overcome stalled replication involves skipping the DNA lesion using a specialized polymerase known as PrimPol, which reinitiates DNA synthesis downstream of the damage. However, the mechanism regulating PrimPol repriming is largely unclear. In this study, we observe that knockdown of STN1 or CTC1, components of the CTC1/STN1/TEN1 complex, leads to enhanced replication progression following UV exposure. We find that such increased replication is dependent on PrimPol, and PrimPol recruitment to stalled forks increases upon CST depletion. Moreover, we find that p21 is upregulated in STN1-depleted cells in a p53-independent manner, and p21 depletion restores normal replication rates caused by STN1 deficiency. We identify that p21 interacts with PrimPol, and STN1 depletion stimulates p21-PrimPol interaction and facilitates PrimPol recruitment to stalled forks. Our findings reveal a previously undescribed interplay between CST, PrimPol and p21 in promoting repriming in response to stalled replication, and shed light on the regulation of PrimPol repriming at stalled forks.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21 , DNA Primase , DNA Replication , DNA-Directed DNA Polymerase , Multifunctional Enzymes , Telomere-Binding Proteins , Ultraviolet Rays , Humans , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Primase/metabolism , DNA Primase/genetics , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Telomere-Binding Proteins/metabolism , Telomere-Binding Proteins/genetics , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , DNA Damage
20.
J Biol Chem ; 300(4): 107136, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38447798

ABSTRACT

Polyploid giant cancer cells (PGCC) are frequently detected in tumors and are increasingly recognized for their roles in chromosomal instability and associated genome evolution that leads to cancer recurrence. We previously reported that therapy stress promotes polyploidy, and that acid ceramidase plays a role in depolyploidization. In this study, we used an RNA-seq approach to gain a better understanding of the underlying transcriptomic changes that occur as cancer cells progress through polyploidization and depolyploidization. Our results revealed gene signatures that are associated with disease-free and/or overall survival in several cancers and identified the cell cycle inhibitor CDKN1A/p21 as the major hub in PGCC and early progeny. Increased expression of p21 in PGCC was limited to the cytoplasm. We previously demonstrated that the sphingolipid enzyme acid ceramidase is dispensable for polyploidization upon therapy stress but plays a crucial role in depolyploidization. The current study demonstrates that treatment of cells with ceramide is not sufficient for p53-independent induction of p21 and that knockdown of acid ceramidase, which hydrolyzes ceramide, does not interfere with upregulation of p21. In contrast, blocking the expression of p21 with UC2288 prevented the induction of acid ceramidase and inhibited both the formation of PGCC from parental cells as well as the generation of progeny from PGCC. Taken together, our data suggest that p21 functions upstream of acid ceramidase and plays an important role in polyploidization and depolyploidization.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21 , Giant Cells , Neoplasms , Polyploidy , Humans , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Giant Cells/metabolism , Giant Cells/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL