Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Cell Rep ; 43(4): 114044, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38568812

We identify a senescence restriction point (SeRP) as a critical event for cells to commit to senescence. The SeRP integrates the intensity and duration of oncogenic stress, keeps a memory of previous stresses, and combines oncogenic signals acting on different pathways by modulating chromatin accessibility. Chromatin regions opened upon commitment to senescence are enriched in nucleolar-associated domains, which are gene-poor regions enriched in repeated sequences. Once committed to senescence, cells no longer depend on the initial stress signal and exhibit a characteristic transcriptome regulated by a transcription factor network that includes ETV4, RUNX1, OCT1, and MAFB. Consistent with a tumor suppressor role for this network, the levels of ETV4 and RUNX1 are very high in benign lesions of the pancreas but decrease dramatically in pancreatic ductal adenocarcinomas. The discovery of senescence commitment and its chromatin-linked regulation suggests potential strategies for reinstating tumor suppression in human cancers.


Cellular Senescence , Chromatin , Humans , Chromatin/metabolism , Cellular Senescence/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Signal Transduction , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Transcription Factors/metabolism , Mice , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinogenesis/metabolism , Oncogenes
2.
Mol Cell ; 81(18): 3848-3865.e19, 2021 09 16.
Article En | MEDLINE | ID: mdl-34547241

Metabolic rewiring and redox balance play pivotal roles in cancer. Cellular senescence is a barrier for tumorigenesis circumvented in cancer cells by poorly understood mechanisms. We report a multi-enzymatic complex that reprograms NAD metabolism by transferring reducing equivalents from NADH to NADP+. This hydride transfer complex (HTC) is assembled by malate dehydrogenase 1, malic enzyme 1, and cytosolic pyruvate carboxylase. HTC is found in phase-separated bodies in the cytosol of cancer or hypoxic cells and can be assembled in vitro with recombinant proteins. HTC is repressed in senescent cells but induced by p53 inactivation. HTC enzymes are highly expressed in mouse and human prostate cancer models, and their inactivation triggers senescence. Exogenous expression of HTC is sufficient to bypass senescence, rescue cells from complex I inhibitors, and cooperate with oncogenic RAS to transform primary cells. Altogether, we provide evidence for a new multi-enzymatic complex that reprograms metabolism and overcomes cellular senescence.


Cellular Senescence/physiology , NAD/metabolism , Aging/metabolism , Aging/physiology , Animals , Cell Line, Tumor , Cellular Senescence/genetics , Cytosol , Glucose/metabolism , Humans , Hydrogen/chemistry , Hydrogen/metabolism , Malate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , NAD/physiology , Oxidation-Reduction , Pyruvate Carboxylase/metabolism , Pyruvic Acid/metabolism
3.
Cell Cycle ; 18(6-7): 759-770, 2019.
Article En | MEDLINE | ID: mdl-30874462

Senescence is a tumor suppressor program characterized by a stable growth arrest while maintaining cell viability. Senescence-associated ribogenesis defects (SARD) have been shown to regulate senescence through the ability of the ribosomal protein S14 (RPS14 or uS11) to bind and inhibit the cyclin-dependent kinase 4 (CDK4). Here we report another ribosomal protein that binds and inhibits CDK4 in senescent cells: L22 (RPL22 or eL22). Enforcing the expression of RPL22/eL22 is sufficient to induce an RB and p53-dependent cellular senescent phenotype in human fibroblasts. Mechanistically, RPL22/eL22 can interact with and inhibit CDK4-Cyclin D1 to decrease RB phosphorylation both in vitro and in cells. Briefly, we show that ribosome-free RPL22/eL22 causes a cell cycle arrest which could be relevant during situations of nucleolar stress such as cellular senescence or the response to cancer chemotherapy.


Cell Cycle/physiology , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Cell Cycle Checkpoints/physiology , Cell Line , Cellular Senescence/physiology , HEK293 Cells , Humans , Phosphorylation/physiology , Retinoblastoma Protein/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism
4.
Cytokine ; 117: 15-22, 2019 05.
Article En | MEDLINE | ID: mdl-30776684

The senescence-associated secretory phenotype (SASP) defines the ability of senescent cells to express and secrete a variety of extracellular modulators that includes cytokines, chemokines, proteases, growth factors and bioactive lipids. The role of the SASP depends on the context. The SASP reinforces the senescent cell cycle arrest, stimulates the immune-mediated clearance of potentially tumorigenic cells, limits fibrosis and promotes wound healing and tissue regeneration. On the other hand, the SASP can mediate chronic inflammation and stimulate the growth and survival of tumor cells. The regulation of the SASP occurs at multiple levels including chromatin remodelling, activation of specific transcription factors such as C/EBP and NF-κB, control of mRNA translation and intracellular trafficking. Several SASP modulators have already been identified setting the stage for future research on their clinical applications.


Cellular Senescence , Cellular Reprogramming , Cellular Senescence/genetics , Epigenesis, Genetic , Humans , Lipids/analysis , NF-kappa B/metabolism , Neoplasms/pathology
5.
Bioorg Med Chem ; 26(20): 5547-5554, 2018 11 01.
Article En | MEDLINE | ID: mdl-30309670

Lamin A contributes to the structure of nuclei in all mammalian cells and plays an important role in cell division and migration. Mature lamin A is derived from a farnesylated precursor protein, known as prelamin A, which undergoes post-translational cleavage catalyzed by the zinc metalloprotease STE24 (ZPMSTE24). Accumulation of farnesylated prelamin A in the nuclear envelope compromises cell division, impairs mitosis and induces an increased expression of inflammatory gene products. ZMPSTE24 has been proposed as a potential therapeutic target in oncology. A library of peptidomimetic compounds were synthesized and screened for their ability to induce accumulation of prelamin A in cancer cells and block cell migration in pancreatic ductal adenocarcinoma cells. The results of this study suggest that inhibitors of lamin A maturation may interfere with cell migration, the biological process required for cancer metastasis.


Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Lamin Type A/metabolism , Peptidomimetics/chemistry , Peptidomimetics/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antineoplastic Agents/chemical synthesis , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Humans , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Peptidomimetics/chemical synthesis , Phosphinic Acids/chemical synthesis , Phosphinic Acids/chemistry , Phosphinic Acids/pharmacology
6.
Nat Cell Biol ; 20(7): 789-799, 2018 07.
Article En | MEDLINE | ID: mdl-29941930

Cellular senescence is a tumour suppressor programme characterized by a stable cell cycle arrest. Here we report that cellular senescence triggered by a variety of stimuli leads to diminished ribosome biogenesis and the accumulation of both rRNA precursors and ribosomal proteins. These defects were associated with reduced expression of several ribosome biogenesis factors, the knockdown of which was also sufficient to induce senescence. Genetic analysis revealed that Rb but not p53 was required for the senescence response to altered ribosome biogenesis. Mechanistically, the ribosomal protein S14 (RPS14 or uS11) accumulates in the soluble non-ribosomal fraction of senescent cells, where it binds and inhibits CDK4 (cyclin-dependent kinase 4). Overexpression of RPS14 is sufficient to inhibit Rb phosphorylation, inducing cell cycle arrest and senescence. Here we describe a mechanism for maintaining the senescent cell cycle arrest that may be relevant for cancer therapy, as well as biomarkers to identify senescent cells.


Cell Cycle Checkpoints , Cellular Senescence , Neoplasms/metabolism , Retinoblastoma Protein/metabolism , Ribosomes/metabolism , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , HEK293 Cells , Humans , Neoplasms/genetics , Neoplasms/pathology , PC-3 Cells , Phosphorylation , Protein Binding , RNA Precursors/biosynthesis , RNA Precursors/genetics , RNA, Ribosomal/biosynthesis , RNA, Ribosomal/genetics , RNA-Binding Proteins , Retinoblastoma Protein/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/genetics , Signal Transduction , Time Factors
7.
Sci Rep ; 8(1): 7754, 2018 05 17.
Article En | MEDLINE | ID: mdl-29773808

Several regulators of SUMOylation have been previously linked to senescence but most targets of this modification in senescent cells remain unidentified. Using a two-step purification of a modified SUMO3, we profiled the SUMO proteome of senescent cells in a site-specific manner. We identified 25 SUMO sites on 23 proteins that were significantly regulated during senescence. Of note, most of these proteins were PML nuclear body (PML-NB) associated, which correlates with the increased number and size of PML-NBs observed in senescent cells. Interestingly, the sole SUMO E2 enzyme, UBC9, was more SUMOylated during senescence on its Lys-49. Functional studies of a UBC9 mutant at Lys-49 showed a decreased association to PML-NBs and the loss of UBC9's ability to delay senescence. We thus propose both pro- and anti-senescence functions of protein SUMOylation.


Cell Nucleus/metabolism , Cellular Senescence , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/metabolism , Proteome/analysis , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Humans , Protein Conformation , Sumoylation , Tumor Cells, Cultured , Ubiquitin-Conjugating Enzymes/chemistry
8.
FASEB J ; 31(11): 5012-5018, 2017 11.
Article En | MEDLINE | ID: mdl-28754713

The ubiquitin-associated protein 2-like (UBAP2L) gene remains poorly studied in human and mouse development. UBAP2L interacts with the Polycomb group protein B lymphoma Mo-MLV insertion region 1 homolog (BMI1) and determines the activity of mouse hematopoietic stem cells in vivo Here we show that loss of Ubap2l leads to disorganized respiratory epithelium of mutant neonates, which die of respiratory failure. We also show that UBAP2L overexpression leads to epithelial-mesenchymal transition-like phenotype in a non-small cell lung carcinoma (NSCLC) cell line. UBAP2L is amplified in 15% of human primary lung adenocarcinoma specimens. Such patients express higher levels of UBAP2L and show a reduction in survival when compared with those who do not have this gene amplification. Supporting a possible role for UBAP2L in lung tumor progression, NSCLC cells engineered to express low levels of this gene produce much smaller tumors in vivo than wild-type control cells. Together, these results suggest that UBAP2L contributes to epithelial lung cell identity in mice and that it plays an important role in human lung adenocarcinoma.-Aucagne, R., Girard, S., Mayotte, N., Lehnertz, B., Lopes-Paciencia, S., Gendron, P., Boucher, G., Chagraoui, J., Sauvageau, G. UBAP2L is amplified in a large subset of human lung adenocarcinoma and is critical for epithelial lung cell identity and tumor metastasis.


Adenocarcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carrier Proteins/biosynthesis , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Respiratory Mucosa/metabolism , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carrier Proteins/genetics , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Neoplasm Metastasis , Neoplasm Proteins/genetics , Respiratory Mucosa/pathology
9.
Aging (Albany NY) ; 8(2): 366-81, 2016 Feb.
Article En | MEDLINE | ID: mdl-26922519

Mutants of lamin A cause diseases including the Hutchinson-Gilford progeria syndrome (HGPS) characterized by premature aging. Lamin A undergoes a series of processing reactions, including farnesylation and proteolytic cleavage of the farnesylated C-terminal domain. The role of cleavage is unknown but mutations that affect this reaction lead to progeria. Here we show that interphase serine 22 phosphorylation of endogenous mutant lamin A (progerin) is defective in cells from HGPS patients. This defect can be mimicked by expressing progerin in human cells and prevented by inhibition of farnesylation. Furthermore, serine 22 phosphorylation of non-farnesylated progerin was enhanced by a mutation that disrupts lamin A head to tail interactions. The phosphorylation of lamin A or non-farnesylated progerin was associated to the formation of spherical intranuclear lamin A droplets that accumulate protein kinases of the CDK family capable of phosphorylating lamin A at serine 22. CDK inhibitors compromised the turnover of progerin, accelerated senescence of HGPS cells and reversed the effects of FTI on progerin levels. We discuss a model of progeria where faulty serine 22 phosphorylation compromises phase separation of lamin A polymers, leading to accumulation of functionally impaired lamin A structures.


Interphase , Lamin Type A/genetics , Lamin Type A/metabolism , Progeria/genetics , Progeria/metabolism , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Protein Prenylation , Serine/metabolism
...