Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 169(7): 1358-1358.e1, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28622516

ABSTRACT

During mitosis, a cell divides its duplicated genome into two identical daughter cells. This process must occur without errors to prevent proliferative diseases (e.g., cancer). A key mechanism controlling mitosis is the precise timing of more than 32,000 phosphorylation and dephosphorylation events by a network of kinases and counterbalancing phosphatases. The identity, magnitude, and temporal regulation of these events have emerged recently, largely from advances in mass spectrometry. Here, we show phosphoevents currently believed to be key regulators of mitosis. For an animated version of this SnapShot, please see http://www.cell.com/cell/enhanced/odonoghue2.


Subject(s)
Mitosis , Protein Kinases/metabolism , Animals , Humans , Phosphorylation
2.
Nat Rev Genet ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169218

ABSTRACT

Mosaic variegated aneuploidy (MVA) is a rare condition in which abnormal chromosome counts (that is, aneuploidies), affecting different chromosomes in each cell (making it variegated) are found only in a certain number of cells (making it mosaic). MVA is characterized by various developmental defects and, despite its rarity, presents a unique clinical scenario to understand the consequences of chromosomal instability and copy number variation in humans. Research from patients with MVA, genetically engineered mouse models and functional cellular studies have found the genetic causes to be mutations in components of the spindle-assembly checkpoint as well as in related proteins involved in centrosome dynamics during mitosis. MVA is accompanied by tumour susceptibility (depending on the genetic basis) as well as cellular and systemic stress, including chronic immune response and the associated clinical implications.

3.
Nat Rev Mol Cell Biol ; 17(5): 280-92, 2016 05.
Article in English | MEDLINE | ID: mdl-27033256

ABSTRACT

The roles of cyclins and their catalytic partners, the cyclin-dependent kinases (CDKs), as core components of the machinery that drives cell cycle progression are well established. Increasing evidence indicates that mammalian cyclins and CDKs also carry out important functions in other cellular processes, such as transcription, DNA damage repair, control of cell death, differentiation, the immune response and metabolism. Some of these non-canonical functions are performed by cyclins or CDKs, independently of their respective cell cycle partners, suggesting that there was a substantial divergence in the functions of these proteins during evolution.


Subject(s)
Cyclin-Dependent Kinases/physiology , Cyclins/physiology , Animals , Cell Cycle , Cell Differentiation , DNA Breaks, Double-Stranded , DNA Repair , Humans , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction , Transcriptional Activation
4.
EMBO J ; 42(2): e110833, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36354735

ABSTRACT

The AKT-mTOR pathway is a central regulator of cell growth and metabolism. Upon sustained mTOR activity, AKT activity is attenuated by a feedback loop that restrains upstream signaling. However, how cells control the signals that limit AKT activity is not fully understood. Here, we show that MASTL/Greatwall, a cell cycle kinase that supports mitosis by phosphorylating the PP2A/B55 inhibitors ENSA/ARPP19, inhibits PI3K-AKT activity by sustaining mTORC1- and S6K1-dependent phosphorylation of IRS1 and GRB10. Genetic depletion of MASTL results in an inefficient feedback loop and AKT hyperactivity. These defects are rescued by the expression of phosphomimetic ENSA/ARPP19 or inhibition of PP2A/B55 phosphatases. MASTL is directly phosphorylated by mTORC1, thereby limiting the PP2A/B55-dependent dephosphorylation of IRS1 and GRB10 downstream of mTORC1. Downregulation of MASTL results in increased glucose uptake in vitro and increased glucose tolerance in adult mice, suggesting the relevance of the MASTL-PP2A/B55 kinase-phosphatase module in controlling AKT and maintaining metabolic homeostasis.


Subject(s)
Mechanistic Target of Rapamycin Complex 1 , Protein Phosphatase 2 , Protein Serine-Threonine Kinases , Animals , Mice , Cell Cycle/genetics , Glucose/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitosis , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
5.
EMBO J ; 42(1): e111251, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36326833

ABSTRACT

Maintenance of stemness is tightly linked to cell cycle regulation through protein phosphorylation by cyclin-dependent kinases (CDKs). However, how this process is reversed during differentiation is unknown. We report here that exit from stemness and differentiation of pluripotent cells along the neural lineage are controlled by CDC14, a CDK-counteracting phosphatase whose function in mammals remains obscure. Lack of the two CDC14 family members, CDC14A and CDC14B, results in deficient development of the neural system in the mouse and impairs neural differentiation from embryonic stem cells (ESCs). Mechanistically, CDC14 directly dephosphorylates specific proline-directed Ser/Thr residues of undifferentiated embryonic transcription Factor 1 (UTF1) during the exit from stemness, triggering its proteasome-dependent degradation. Multiomic single-cell analysis of transcription and chromatin accessibility in differentiating ESCs suggests that increased UTF1 levels in the absence of CDC14 prevent the proper firing of bivalent promoters required for differentiation. CDC14 phosphatases are dispensable for mitotic exit, suggesting that CDC14 phosphatases have evolved to control stemness rather than cell cycle exit and establish the CDK-CDC14 axis as a critical molecular switch for linking cell cycle regulation and self-renewal.


Subject(s)
Cell Cycle Proteins , Saccharomyces cerevisiae Proteins , Animals , Mice , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Cyclin-Dependent Kinases/metabolism , Cell Cycle , Phosphorylation/physiology , Mitosis , Saccharomyces cerevisiae Proteins/metabolism , Mammals
6.
Blood ; 144(2): 156-170, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38684032

ABSTRACT

ABSTRACT: Hematopoietic stem cells (HSCs) are characterized by the ability to self-renew and to replenish the hematopoietic system. The cell-cycle kinase cyclin-dependent kinase 6 (CDK6) regulates transcription, whereby it has both kinase-dependent and kinase-independent functions. Herein, we describe the complex role of CDK6, balancing quiescence, proliferation, self-renewal, and differentiation in activated HSCs. Mouse HSCs expressing kinase-inactivated CDK6 show enhanced long-term repopulation and homing, whereas HSCs lacking CDK6 have impaired functionality. The transcriptomes of basal and serially transplanted HSCs expressing kinase-inactivated CDK6 exhibit an expression pattern dominated by HSC quiescence and self-renewal, supporting a concept, in which myc-associated zinc finger protein (MAZ) and nuclear transcription factor Y subunit alpha (NFY-A) are critical CDK6 interactors. Pharmacologic kinase inhibition with a clinically used CDK4/6 inhibitor in murine and human HSCs validated our findings and resulted in increased repopulation capability and enhanced stemness. Our findings highlight a kinase-independent role of CDK6 in long-term HSC functionality. CDK6 kinase inhibition represents a possible strategy to improve HSC fitness.


Subject(s)
Cyclin-Dependent Kinase 6 , Hematopoietic Stem Cells , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase 6/genetics , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Mice , Humans , Adult Stem Cells/metabolism , Adult Stem Cells/cytology , Cell Proliferation , Cell Differentiation , Mice, Inbred C57BL , Hematopoietic Stem Cell Transplantation , Cell Self Renewal/drug effects
7.
Cell ; 144(4): 465-6, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21335229

ABSTRACT

Tumor cells frequently display an abnormal number of chromosomes, a phenomenon known as aneuploidy. Tang et al. (2011) now show that aneuploid cells are particularly sensitive to compounds that induce proteotoxic and energy stress. Could this vulnerability lead to new cancer therapies?

8.
Cell ; 144(2): 187-99, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21241890

ABSTRACT

PTEN is a frequently mutated tumor suppressor gene that opposes the PI3K/AKT pathway through dephosphorylation of phosphoinositide-3,4,5-triphosphate. Recently, nuclear compartmentalization of PTEN was found as a key component of its tumor-suppressive activity; however its nuclear function remains poorly defined. Here we show that nuclear PTEN interacts with APC/C, promotes APC/C association with CDH1, and thereby enhances the tumor-suppressive activity of the APC-CDH1 complex. We find that nuclear exclusion but not phosphatase inactivation of PTEN impairs APC-CDH1. This nuclear function of PTEN provides a straightforward mechanistic explanation for the fail-safe cellular senescence response elicited by acute PTEN loss and the tumor-suppressive activity of catalytically inactive PTEN. Importantly, we demonstrate that PTEN mutant and PTEN null states are not synonymous as they are differentially sensitive to pharmacological inhibition of APC-CDH1 targets such as PLK1 and Aurora kinases. This finding identifies a strategy for cancer patient stratification and, thus, optimization of targeted therapies. PAPERCLIP:


Subject(s)
Cadherins/metabolism , Cellular Senescence , PTEN Phosphohydrolase/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Antigens, CD , Aurora Kinases , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Male , Mice , Neoplasm Transplantation , PTEN Phosphohydrolase/genetics , Phosphoric Monoester Hydrolases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Transplantation, Heterologous
9.
EMBO J ; 40(11): e99692, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33856059

ABSTRACT

Chemical inhibitors of the deubiquitinase USP7 are currently being developed as anticancer agents based on their capacity to stabilize P53. Regardless of this activity, USP7 inhibitors also generate DNA damage in a p53-independent manner. However, the mechanism of this genotoxicity and its contribution to the anticancer effects of USP7 inhibitors are still under debate. Here we show that, surprisingly, even if USP7 inhibitors stop DNA replication, they also induce a widespread activation of CDK1 throughout the cell cycle, which leads to DNA damage and is toxic for mammalian cells. In addition, USP7 interacts with the phosphatase PP2A and supports its active localization in the cytoplasm. Accordingly, inhibition of USP7 or PP2A triggers very similar changes of the phosphoproteome, including a widespread increase in the phosphorylation of CDK1 targets. Importantly, the toxicity of USP7 inhibitors is alleviated by lowering CDK1 activity or by chemical activation of PP2A. Our work reveals that USP7 limits CDK1 activity at all cell cycle stages, providing a novel mechanism that explains the toxicity of USP7 inhibitors through untimely activation of CDK1.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle , Ubiquitin-Specific Peptidase 7/metabolism , Animals , Cells, Cultured , DNA Damage , HCT116 Cells , Humans , Mice , NIH 3T3 Cells , Protease Inhibitors/toxicity , Protein Phosphatase 2/metabolism , Protein Transport , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors
10.
Nature ; 568(7753): 557-560, 2019 04.
Article in English | MEDLINE | ID: mdl-30971822

ABSTRACT

The cell cycle is a tightly regulated process that is controlled by the conserved cyclin-dependent kinase (CDK)-cyclin protein complex1. However, control of the G0-to-G1 transition is not completely understood. Here we demonstrate that p38 MAPK gamma (p38γ) acts as a CDK-like kinase and thus cooperates with CDKs, regulating entry into the cell cycle. p38γ shares high sequence homology, inhibition sensitivity and substrate specificity with CDK family members. In mouse hepatocytes, p38γ induces proliferation after partial hepatectomy by promoting the phosphorylation of retinoblastoma tumour suppressor protein at known CDK target residues. Lack of p38γ or treatment with the p38γ inhibitor pirfenidone protects against the chemically induced formation of liver tumours. Furthermore, biopsies of human hepatocellular carcinoma show high expression of p38γ, suggesting that p38γ could be a therapeutic target in the treatment of this disease.


Subject(s)
Carcinogenesis/pathology , Cell Cycle , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Liver/enzymology , Liver/pathology , Mitogen-Activated Protein Kinase 12/metabolism , Aged , Animals , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Female , Hepatocytes/cytology , Hepatocytes/pathology , Humans , Liver/surgery , Liver Neoplasms/chemically induced , Male , Mice , Middle Aged , Mitogen-Activated Protein Kinase 12/antagonists & inhibitors , Phosphorylation , Pyridones/pharmacology , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Sequence Homology , Substrate Specificity
11.
EMBO J ; 39(16): e104324, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32614092

ABSTRACT

Full differentiation potential along with self-renewal capacity is a major property of pluripotent stem cells (PSCs). However, the differentiation capacity frequently decreases during expansion of PSCs in vitro. We show here that transient exposure to a single microRNA, expressed at early stages during normal development, improves the differentiation capacity of already-established murine and human PSCs. Short exposure to miR-203 in PSCs (miPSCs) induces a transient expression of 2C markers that later results in expanded differentiation potency to multiple lineages, as well as improved efficiency in tetraploid complementation and human-mouse interspecies chimerism assays. Mechanistically, these effects are at least partially mediated by direct repression of de novo DNA methyltransferases Dnmt3a and Dnmt3b, leading to transient and reversible erasure of DNA methylation. These data support the use of transient exposure to miR-203 as a versatile method to reset the epigenetic memory in PSCs, and improve their effectiveness in regenerative medicine.


Subject(s)
Cell Differentiation , DNA Methylation , Epigenesis, Genetic , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/metabolism , Animals , Cell Line , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Humans , Induced Pluripotent Stem Cells/cytology , Mice , MicroRNAs/genetics , DNA Methyltransferase 3B
12.
J Cell Sci ; 135(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35694956

ABSTRACT

Chromosome segregation requires that centromeres properly attach to spindle microtubules. This essential step regulates the accuracy of cell division and must therefore be precisely regulated. One of the main centromeric regulatory signaling pathways is the haspin-H3T3ph-chromosomal passenger complex (CPC) cascade, which is responsible for the recruitment of the CPC to the centromeres. During mitosis, the haspin kinase phosphorylates histone H3 at threonine 3 (H3T3ph), an essential epigenetic mark that recruits the CPC, in which the catalytic component is Aurora B kinase (AURKB). However, the centromeric haspin-H3T3ph-CPC pathway remains largely uncharacterized in mammalian male meiosis. We have analyzed haspin functions by either its chemical inhibition with LDN-192960 in cultured spermatocytes, or the ablation of the Haspin gene in Haspin-/- mice. Our studies suggest that haspin kinase activity is required for proper chromosome congression both during meiotic divisions and for the recruitment of Aurora B and kinesin MCAK (also known as KIF2C) to meiotic centromeres. However, the absence of H3T3ph histone mark does not alter borealin (or CDCA8) and SGO2 centromeric localization. These results add new and relevant information regarding the regulation of the haspin-H3T3ph-CPC pathway and centromere function during meiosis.


Subject(s)
Aurora Kinase B , Chromosome Segregation , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Animals , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Cell Cycle Proteins/metabolism , Centromere/metabolism , Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kinesins/genetics , Male , Mammals/metabolism , Meiosis/genetics , Mice , Mitosis , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Threonine/metabolism
13.
Breast Cancer Res ; 25(1): 91, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37542268

ABSTRACT

A hallmark of many malignant tumors is dedifferentiated (immature) cells bearing slight or no resemblance to the normal cells from which the cancer originated. Tumor dedifferentiated cells exhibit a higher capacity to survive to chemo and radiotherapies and have the ability to incite tumor relapse. Inducing cancer cell differentiation would abolish their self-renewal and invasive capacity and could be combined with the current standard of care, especially in poorly differentiated and aggressive tumors (with worst prognosis). However, differentiation therapy is still in its early stages and the intrinsic complexity of solid tumor heterogeneity demands innovative approaches in order to be efficiently translated into the clinic. We demonstrate here that microRNA 203, a potent driver of differentiation in pluripotent stem cells (ESCs and iPSCs), promotes the differentiation of mammary gland tumor cells. Combining mouse in vivo approaches and both mouse and human-derived tridimensional organoid cultures, we report that miR-203 influences the self-renewal capacity, plasticity and differentiation potential of breast cancer cells and prevents tumor cell growth in vivo. Our work sheds light on differentiation-based antitumor therapies and offers miR-203 as a promising tool for directly confronting the tumor-maintaining and regeneration capability of cancer cells.


Subject(s)
Breast Neoplasms , MicroRNAs , Humans , Mice , Animals , Female , MicroRNAs/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Neoplasm Recurrence, Local/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Neoplastic Stem Cells/pathology
14.
EMBO Rep ; 22(4): e51030, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33615693

ABSTRACT

Cell division requires the regulation of karyokinesis and cytokinesis, which includes an essential role of the achromatic spindle. Although the functions of centrosomes are well characterised in somatic cells, their role during vertebrate spermatogenesis remains elusive. We have studied the dynamics of the meiotic centrosomes in male mouse during both meiotic divisions. Results show that meiotic centrosomes duplicate twice: first duplication occurs in the leptotene/zygotene transition, while the second occurs in interkinesis. The maturation of duplicated centrosomes during the early stages of prophase I and II are followed by their separation and migration to opposite poles to form bipolar spindles I and II. The study of the genetic mouse model Plk1(Δ/Δ) indicates a central role of Polo-like kinase 1 in pericentriolar matrix assembly, in centrosome maturation and migration, and in the formation of the bipolar spindles during spermatogenesis. In addition, in vitro inhibition of Polo-like kinase 1 and Aurora A in organotypic cultures of seminiferous tubules points out to a prominent role of both kinases in the regulation of the formation of meiotic bipolar spindles.


Subject(s)
Cell Cycle Proteins , Centrosome , Animals , Cell Cycle Proteins/genetics , Male , Meiosis , Mice , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/genetics , Spindle Apparatus , Polo-Like Kinase 1
15.
Semin Cell Dev Biol ; 107: 28-35, 2020 11.
Article in English | MEDLINE | ID: mdl-32334991

ABSTRACT

Proper progression throughout the cell division cycle depends on the expression level of a family of proteins known as cyclins, and the subsequent activation of cyclin-dependent kinases (Cdks). Among the numerous members of the mammalian cyclin family, only a few of them, cyclins A, B, C, D and E, are known to display critical roles in the cell cycle. These functions will be reviewed here with a special focus on their relevance in different cell types in vivo and their implications in human disease.


Subject(s)
Cell Cycle , Cyclins/metabolism , Mammals/metabolism , Animals , Cell Cycle/genetics , Chromosome Segregation/genetics , DNA Replication/genetics , Humans , Transcription, Genetic
16.
Mol Cell Proteomics ; 19(2): 326-343, 2020 02.
Article in English | MEDLINE | ID: mdl-31852836

ABSTRACT

The human MASTL (Microtubule-associated serine/threonine kinase-like) gene encodes an essential protein in the cell cycle. MASTL is a key factor preventing early dephosphorylation of M-phase targets of Cdk1/CycB. Little is known about the mechanism of MASTL activation and regulation. MASTL contains a non-conserved insertion of 550 residues within its activation loop, splitting the kinase domain, and making it unique. Here, we show that this non-conserved middle region (NCMR) of the protein is crucial for target specificity and activity. We performed a phosphoproteomic assay with different MASTL constructs identifying key phosphorylation sites for its activation and determining whether they arise from autophosphorylation or exogenous kinases, thus generating an activation model. Hydrogen/deuterium exchange data complements this analysis revealing that the C-lobe in full-length MASTL forms a stable structure, whereas the N-lobe is dynamic and the NCMR and C-tail contain few localized regions with higher-order structure. Our results indicate that truncated versions of MASTL conserving a cryptic C-Lobe in the NCMR, display catalytic activity and different targets, thus establishing a possible link with truncated mutations observed in cancer-related databases.


Subject(s)
Microtubule-Associated Proteins , Protein Serine-Threonine Kinases , Catalysis , Cell Line, Tumor , HEK293 Cells , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
17.
EMBO J ; 36(12): 1688-1706, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28465321

ABSTRACT

Retinal ganglion cells (RGCs) are the sole projecting neurons of the retina and their axons form the optic nerve. Here, we show that embryogenesis-associated mouse RGC differentiation depends on mitophagy, the programmed autophagic clearance of mitochondria. The elimination of mitochondria during RGC differentiation was coupled to a metabolic shift with increased lactate production and elevated expression of glycolytic enzymes at the mRNA level. Pharmacological and genetic inhibition of either mitophagy or glycolysis consistently inhibited RGC differentiation. Local hypoxia triggered expression of the mitophagy regulator BCL2/adenovirus E1B 19-kDa-interacting protein 3-like (BNIP3L, best known as NIX) at peak RGC differentiation. Retinas from NIX-deficient mice displayed increased mitochondrial mass, reduced expression of glycolytic enzymes and decreased neuronal differentiation. Similarly, we provide evidence that NIX-dependent mitophagy contributes to mitochondrial elimination during macrophage polarization towards the proinflammatory and more glycolytic M1 phenotype, but not to M2 macrophage differentiation, which primarily relies on oxidative phosphorylation. In summary, developmentally controlled mitophagy promotes a metabolic switch towards glycolysis, which in turn contributes to cellular differentiation in several distinct developmental contexts.


Subject(s)
Cell Differentiation , Glycolysis , Mitophagy , Retina/embryology , Retinal Ganglion Cells/physiology , Animals , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/metabolism
18.
Mol Cell ; 51(5): 647-61, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24034696

ABSTRACT

Reversible cellular quiescence is critical for developmental processes in metazoan organisms and is characterized by a reduction in cell size and transcriptional activity. We show that the Aurora B kinase and the polycomb protein Ring1B have essential roles in regulating transcriptionally active genes in quiescent lymphocytes. Ring1B and Aurora B bind to a wide range of active promoters in resting B and T cells. Conditional knockout of either protein results in reduced transcription and binding of RNA Pol II to promoter regions and decreased cell viability. Aurora B phosphorylates histone H3S28 at active promoters in resting B cells as well as inhibiting Ring1B-mediated ubiquitination of histone H2A and enhancing binding and activity of the USP16 deubiquitinase at transcribed genes. Our results identify a mechanism for regulating transcription in quiescent cells that has implications for epigenetic regulation of the choice between proliferation and quiescence.


Subject(s)
Aurora Kinase B/metabolism , B-Lymphocytes/physiology , Polycomb Repressive Complex 1/metabolism , Promoter Regions, Genetic , T-Lymphocytes/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Aurora Kinase B/genetics , Cell Survival , Cells, Cultured , Gene Expression Regulation , Gene Knockout Techniques , Histones/metabolism , Mice , Polycomb Repressive Complex 1/genetics , RNA Polymerase II/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
19.
Mol Cell ; 48(5): 681-91, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23103253

ABSTRACT

The mammalian target of rapamycin (mTOR) pathway, which is essential for cell proliferation, is repressed in certain cell types in hypoxia. However, hypoxia-inducible factor 2α (HIF2α) can act as a proliferation-promoting factor in some biological settings. This paradoxical situation led us to study whether HIF2α has a specific effect on mTORC1 regulation. Here we show that activation of the HIF2α pathway increases mTORC1 activity by upregulating expression of the amino acid carrier SLC7A5. At the molecular level we also show that HIF2α binds to the Slc7a5 proximal promoter. Our findings identify a link between the oxygen-sensing HIF2α pathway and mTORC1 regulation, revealing the molecular basis of the tumor-promoting properties of HIF2α in von Hippel-Lindau-deficient cells. We also describe relevant physiological scenarios, including those that occur in liver and lung tissue, wherein HIF2α or low-oxygen tension drive mTORC1 activity and SLC7A5 expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Large Neutral Amino Acid-Transporter 1/genetics , Liver/metabolism , Lung/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, SCID , Multiprotein Complexes , Neoplasm Transplantation , Promoter Regions, Genetic , Proteins/genetics , RNA Interference , Signal Transduction , TOR Serine-Threonine Kinases , Time Factors , Transfection , Tumor Burden , Up-Regulation , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
20.
Nucleic Acids Res ; 46(9): 4546-4559, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29590434

ABSTRACT

The cellular response to DNA damage is essential for maintaining the integrity of the genome. Recent evidence has identified E2F7 as a key player in DNA damage-dependent transcriptional regulation of cell-cycle genes. However, the contribution of E2F7 to cellular responses upon genotoxic damage is still poorly defined. Here we show that E2F7 represses the expression of genes involved in the maintenance of genomic stability, both throughout the cell cycle and upon induction of DNA lesions that interfere with replication fork progression. Knockdown of E2F7 leads to a reduction in 53BP1 and FANCD2 foci and to fewer chromosomal aberrations following treatment with agents that cause interstrand crosslink (ICL) lesions but not upon ionizing radiation. Accordingly, E2F7-depleted cells exhibit enhanced cell-cycle re-entry and clonogenic survival after exposure to ICL-inducing agents. We further report that expression and functional activity of E2F7 are p53-independent in this context. Using a cell-based assay, we show that E2F7 restricts homologous recombination through the transcriptional repression of RAD51. Finally, we present evidence that downregulation of E2F7 confers an increased resistance to chemotherapy in recombination-deficient cells. Taken together, our results reveal an E2F7-dependent transcriptional program that contributes to the regulation of DNA repair and genomic integrity.


Subject(s)
DNA Repair , E2F7 Transcription Factor/physiology , Gene Expression Regulation , Genomic Instability , Cell Cycle/genetics , Cell Line , Chromosome Breakage , DNA Damage , E2F7 Transcription Factor/metabolism , Humans , Promoter Regions, Genetic , Recombinational DNA Repair , Transcription, Genetic , Transcriptome , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL