Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Nat Rev Mol Cell Biol ; 23(1): 74-88, 2022 01.
Article in English | MEDLINE | ID: mdl-34508254

ABSTRACT

Cancer is a group of diseases in which cells divide continuously and excessively. Cell division is tightly regulated by multiple evolutionarily conserved cell cycle control mechanisms, to ensure the production of two genetically identical cells. Cell cycle checkpoints operate as DNA surveillance mechanisms that prevent the accumulation and propagation of genetic errors during cell division. Checkpoints can delay cell cycle progression or, in response to irreparable DNA damage, induce cell cycle exit or cell death. Cancer-associated mutations that perturb cell cycle control allow continuous cell division chiefly by compromising the ability of cells to exit the cell cycle. Continuous rounds of division, however, create increased reliance on other cell cycle control mechanisms to prevent catastrophic levels of damage and maintain cell viability. New detailed insights into cell cycle control mechanisms and their role in cancer reveal how these dependencies can be best exploited in cancer treatment.


Subject(s)
Cell Cycle Checkpoints , Neoplasms/pathology , Animals , DNA Damage/genetics , DNA Replication/genetics , Humans , Neoplasms/genetics , Neoplasms/therapy , Oncogenes , Spindle Apparatus/metabolism
2.
Mol Cell ; 83(22): 4078-4092.e6, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977119

ABSTRACT

Tumor growth is driven by continued cellular growth and proliferation. Cyclin-dependent kinase 7's (CDK7) role in activating mitotic CDKs and global gene expression makes it therefore an attractive target for cancer therapies. However, what makes cancer cells particularly sensitive to CDK7 inhibition (CDK7i) remains unclear. Here, we address this question. We show that CDK7i, by samuraciclib, induces a permanent cell-cycle exit, known as senescence, without promoting DNA damage signaling or cell death. A chemogenetic genome-wide CRISPR knockout screen identified that active mTOR (mammalian target of rapamycin) signaling promotes samuraciclib-induced senescence. mTOR inhibition decreases samuraciclib sensitivity, and increased mTOR-dependent growth signaling correlates with sensitivity in cancer cell lines. Reverting a growth-promoting mutation in PIK3CA to wild type decreases sensitivity to CDK7i. Our work establishes that enhanced growth alone promotes CDK7i sensitivity, providing an explanation for why some cancers are more sensitive to CDK inhibition than normally growing cells.


Subject(s)
Cyclin-Dependent Kinases , Neoplasms , Humans , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinase-Activating Kinase , Signal Transduction , Cell Cycle , Enzyme Inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Cell Line, Tumor
3.
Nat Rev Mol Cell Biol ; 14(8): 518-28, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23877564

ABSTRACT

The accurate transition from G1 phase of the cell cycle to S phase is crucial for the control of eukaryotic cell proliferation, and its misregulation promotes oncogenesis. During G1 phase, growth-dependent cyclin-dependent kinase (CDK) activity promotes DNA replication and initiates G1-to-S phase transition. CDK activation initiates a positive feedback loop that further increases CDK activity, and this commits the cell to division by inducing genome-wide transcriptional changes. G1-S transcripts encode proteins that regulate downstream cell cycle events. Recent work is beginning to reveal the complex molecular mechanisms that control the temporal order of transcriptional activation and inactivation, determine distinct functional subgroups of genes and link cell cycle-dependent transcription to DNA replication stress in yeast and mammals.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle/genetics , G1 Phase/genetics , S Phase/genetics , Transcription, Genetic/physiology , Animals , Cell Cycle/physiology , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/physiology , G1 Phase/physiology , Humans , Mammals/genetics , Mammals/physiology , Models, Biological , S Phase/physiology , Yeasts/genetics , Yeasts/physiology
4.
EMBO J ; 36(5): 604-616, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28122869

ABSTRACT

An unresolved question is how HIV-1 achieves efficient replication in terminally differentiated macrophages despite the restriction factor SAMHD1. We reveal inducible changes in expression of cell cycle-associated proteins including MCM2 and cyclins A, E, D1/D3 in macrophages, without evidence for DNA synthesis or mitosis. These changes are induced by activation of the Raf/MEK/ERK kinase cascade, culminating in upregulation of CDK1 with subsequent SAMHD1 T592 phosphorylation and deactivation of its antiviral activity. HIV infection is limited to these G1-like phase macrophages at the single-cell level. Depletion of SAMHD1 in macrophages decouples the association between infection and expression of cell cycle-associated proteins, with terminally differentiated macrophages becoming highly susceptible to HIV-1. We observe both embryo-derived and monocyte-derived tissue-resident macrophages in a G1-like phase at frequencies approaching 20%, suggesting how macrophages sustain HIV-1 replication in vivo Finally, we reveal a SAMHD1-dependent antiretroviral activity of histone deacetylase inhibitors acting via p53 activation. These data provide a basis for host-directed therapeutic approaches aimed at limiting HIV-1 burden in macrophages that may contribute to curative interventions.


Subject(s)
G1 Phase , HIV-1/physiology , Immune Evasion , Macrophages/immunology , Macrophages/virology , Monomeric GTP-Binding Proteins/metabolism , Protein Processing, Post-Translational , Cells, Cultured , HIV-1/immunology , Humans , Immunity, Innate , Phosphorylation , SAM Domain and HD Domain-Containing Protein 1
5.
Nat Commun ; 15(1): 1579, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383676

ABSTRACT

Oncogene-induced replication stress is a crucial driver of genomic instability and one of the key events contributing to the onset and evolution of cancer. Despite its critical role in cancer, the mechanisms that generate oncogene-induced replication stress remain not fully understood. Here, we report that an oncogenic c-Myc-dependent increase in cohesins on DNA contributes to the induction of replication stress. Accumulation of cohesins on chromatin is not sufficient to cause replication stress, but also requires cohesins to accumulate at specific sites in a CTCF-dependent manner. We propose that the increased accumulation of cohesins at CTCF site interferes with the progression of replication forks, contributing to oncogene-induced replication stress. This is different from, and independent of, previously suggested mechanisms of oncogene-induced replication stress. This, together with the reported protective role of cohesins in preventing replication stress-induced DNA damage, supports a double-edge involvement of cohesins in causing and tolerating oncogene-induced replication stress.


Subject(s)
Cohesins , Neoplasms , Humans , Chromatin , Cell Cycle Proteins/metabolism , DNA Replication , DNA
6.
Viruses ; 14(2)2022 02 19.
Article in English | MEDLINE | ID: mdl-35216024

ABSTRACT

Modulation of the host cell cycle is a common strategy used by viruses to create a pro-replicative environment. To facilitate viral genome replication, vaccinia virus (VACV) has been reported to alter cell cycle regulation and trigger the host cell DNA damage response. However, the cellular factors and viral effectors that mediate these changes remain unknown. Here, we set out to investigate the effect of VACV infection on cell proliferation and host cell cycle progression. Using a subset of VACV mutants, we characterise the stage of infection required for inhibition of cell proliferation and define the viral effectors required to dysregulate the host cell cycle. Consistent with previous studies, we show that VACV inhibits and subsequently shifts the host cell cycle. We demonstrate that these two phenomena are independent of one another, with viral early genes being responsible for cell cycle inhibition, and post-replicative viral gene(s) responsible for the cell cycle shift. Extending previous findings, we show that the viral kinase F10 is required to activate the DNA damage checkpoint and that the viral B1 kinase and/or B12 pseudokinase mediate degradation of checkpoint effectors p53 and p21 during infection. We conclude that VACV modulates host cell proliferation and host cell cycle progression through temporal expression of multiple VACV effector proteins. (209/200.).


Subject(s)
Cell Cycle/physiology , DNA Damage , Host-Pathogen Interactions/genetics , Vaccinia virus/genetics , Viral Proteins/genetics , Cell Proliferation , HCT116 Cells , HeLa Cells , Humans , Mutation , Tumor Suppressor Protein p53 , Vaccinia virus/physiology , Virus Replication
7.
Metallomics ; 14(7)2022 07 25.
Article in English | MEDLINE | ID: mdl-35689667

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a disease that remains refractory to existing treatments including the nucleoside analogue gemcitabine. In the current study we demonstrate that an organometallic nucleoside analogue, the ferronucleoside 1-(S,Rp), is cytotoxic in a panel of PDAC cell lines including gemcitabine-resistant MIAPaCa2, with IC50 values comparable to cisplatin. Biochemical studies show that the mechanism of action is inhibition of DNA replication, S-phase cell cycle arrest and stalling of DNA-replication forks, which were directly observed at single molecule resolution by DNA-fibre fluorography. In agreement with this, transcriptional changes following treatment with 1-(S,Rp) include activation of three of the four genes (HUS1, RAD1, RAD17) of the 9-1-1 check point complex clamp and two of the three genes (MRE11, NBN) that form the MRN complex as well as activation of multiple downstream targets. Furthermore, there was evidence of phosphorylation of checkpoint kinases 1 and 2 as well as RPA1 and gamma H2AX, all of which are considered biochemical markers of replication stress. Studies in p53-deficient cell lines showed activation of CDKN1A (p21) and GADD45A by 1-(S,Rp) was at least partially independent of p53. In conclusion, because of its potency and activity in gemcitabine-resistant cells, 1-(S,Rp) is a promising candidate molecule for development of new treatments for PDAC.


Subject(s)
DNA Replication , Nucleosides , Pancreatic Neoplasms , Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Metallocenes , Nucleosides/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , S Phase , Tumor Suppressor Protein p53/metabolism , Pancreatic Neoplasms
8.
J Cell Biol ; 175(4): 595-605, 2006 Nov 20.
Article in English | MEDLINE | ID: mdl-17101693

ABSTRACT

Ubiquitously expressed micro- and millicalpain, which both require the calpain small 1 (CAPNS1) regulatory subunit for function, play important roles in numerous biological and pathological phenomena. We have previously shown that the product of GAS2, a gene specifically induced at growth arrest, is an inhibitor of millicalpain and that its overexpression sensitizes cells to apoptosis in a p53-dependent manner (Benetti, R., G. Del Sal, M. Monte, G. Paroni, C. Brancolini, and C. Schneider. 2001. EMBO J. 20:2702-2714). More recently, we have shown that calpain is also involved in nuclear factor kappaB activation and its relative prosurvival function in response to ceramide, in which calpain deficiency strengthens the proapoptotic effect of ceramide (Demarchi, F., C. Bertoli, P.A. Greer, and C. Schneider. 2005. Cell Death Differ. 12:512-522). Here, we further explore the involvement of calpain in the apoptotic switch and find that in calpain-deficient cells, autophagy is impaired with a resulting dramatic increase in apoptotic cell death. Immunostaining of the endogenous autophagosome marker LC3 and electron microscopy experiments demonstrate that autophagy is impaired in CAPNS1-deficient cells. Accordingly, the enhancement of lysosomal activity and long-lived protein degradation, which normally occur upon starvation, is also reduced. In CAPNS1-depleted cells, ectopic LC3 accumulates in early endosome-like vesicles that may represent a salvage pathway for protein degradation when autophagy is defective.


Subject(s)
Autophagy/physiology , Calpain/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Calpain/deficiency , Ceramides/pharmacology , Endosomes/drug effects , Endosomes/ultrastructure , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/ultrastructure , Gene Expression/drug effects , Gene Silencing , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Phagosomes/drug effects , Phagosomes/ultrastructure , Protein Processing, Post-Translational/drug effects , Sirolimus/pharmacology
9.
Mol Cell Oncol ; 8(2): 1839294, 2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33855165

ABSTRACT

DNA replication capacity, the maximal amount of DNA a cell can synthesize at any given time during S phase, is controlled by E2F-dependent transcription. Controlling replication capacity limits the replication rate and provides a robust mechanism to keep replication fork speed within an optimal range whilst ensuring timely completion of genome duplication.

10.
Cancer Discov ; 11(10): 2456-2473, 2021 10.
Article in English | MEDLINE | ID: mdl-33947663

ABSTRACT

APOBEC3 enzymes are cytosine deaminases implicated in cancer. Precisely when APOBEC3 expression is induced during cancer development remains to be defined. Here we show that specific APOBEC3 genes are upregulated in breast ductal carcinoma in situ, and in preinvasive lung cancer lesions coincident with cellular proliferation. We observe evidence of APOBEC3-mediated subclonal mutagenesis propagated from TRACERx preinvasive to invasive non-small cell lung cancer (NSCLC) lesions. We find that APOBEC3B exacerbates DNA replication stress and chromosomal instability through incomplete replication of genomic DNA, manifested by accumulation of mitotic ultrafine bridges and 53BP1 nuclear bodies in the G1 phase of the cell cycle. Analysis of TRACERx NSCLC clinical samples and mouse lung cancer models revealed APOBEC3B expression driving replication stress and chromosome missegregation. We propose that APOBEC3 is functionally implicated in the onset of chromosomal instability and somatic mutational heterogeneity in preinvasive disease, providing fuel for selection early in cancer evolution. SIGNIFICANCE: This study reveals the dynamics and drivers of APOBEC3 gene expression in preinvasive disease and the exacerbation of cellular diversity by APOBEC3B through DNA replication stress to promote chromosomal instability early in cancer evolution.This article is highlighted in the In This Issue feature, p. 2355.


Subject(s)
APOBEC Deaminases/genetics , Breast Neoplasms/genetics , Carcinoma, Ductal/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Animals , Cell Line, Tumor , Chromosomal Instability , DNA Replication , Female , Humans , Mice
11.
Nat Commun ; 11(1): 3503, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665547

ABSTRACT

DNA replication timing is tightly regulated during S-phase. S-phase length is determined by DNA synthesis rate, which depends on the number of active replication forks and their velocity. Here, we show that E2F-dependent transcription, through E2F6, determines the replication capacity of a cell, defined as the maximal amount of DNA a cell can synthesise per unit time during S-phase. Increasing or decreasing E2F-dependent transcription during S-phase increases or decreases replication capacity, and thereby replication rates, thus shortening or lengthening S-phase, respectively. The changes in replication rate occur mainly through changes in fork speed without affecting the number of active forks. An increase in fork speed does not induce replication stress directly, but increases DNA damage over time causing cell cycle arrest. Thus, E2F-dependent transcription determines the DNA replication capacity of a cell, which affects the replication rate, controlling the time it takes to duplicate the genome and complete S-phase.


Subject(s)
Chromatin/metabolism , DNA Replication/physiology , Blotting, Western , Chromatin/genetics , DNA Damage/genetics , DNA Damage/physiology , DNA Replication/genetics , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , S Phase/genetics , S Phase/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Cancer Discov ; 10(7): 998-1017, 2020 07.
Article in English | MEDLINE | ID: mdl-32349972

ABSTRACT

Loss-of-function mutations of EZH2, the enzymatic component of PRC2, have been associated with poor outcome and chemotherapy resistance in T-cell acute lymphoblastic leukemia (T-ALL). Using isogenic T-ALL cells, with and without CRISPR/Cas9-induced EZH2-inactivating mutations, we performed a cell-based synthetic lethal drug screen. EZH2-deficient cells exhibited increased sensitivity to structurally diverse inhibitors of CHK1, an interaction that could be validated genetically. Furthermore, small-molecule inhibition of CHK1 had efficacy in delaying tumor progression in isogenic EZH2-deficient, but not EZH2 wild-type, T-ALL cells in vivo, as well as in a primary cell model of PRC2-mutant ALL. Mechanistically, EZH2 deficiency resulted in a gene-expression signature of immature T-ALL cells, marked transcriptional upregulation of MYCN, increased replication stress, and enhanced dependency on CHK1 for cell survival. Finally, we demonstrate this phenotype is mediated through derepression of a distal PRC2-regulated MYCN enhancer. In conclusion, we highlight a novel and clinically exploitable pathway in high-risk EZH2-mutated T-ALL. SIGNIFICANCE: Loss-of-function mutations of PRC2 genes are associated with chemotherapy resistance in T-ALL, yet no specific therapy for this aggressive subtype is currently clinically available. Our work demonstrates that loss of EZH2 activity leads to MYCN-driven replication stress, resulting in increased sensitivity to CHK1 inhibition, a finding with immediate clinical relevance.This article is highlighted in the In This Issue feature, p. 890.


Subject(s)
Checkpoint Kinase 1/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Cell Proliferation , Humans , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics
13.
Cell Rep ; 15(7): 1412-1422, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27160911

ABSTRACT

Recent work established DNA replication stress as a crucial driver of genomic instability and a key event at the onset of cancer. Post-translational modifications play an important role in the cellular response to replication stress by regulating the activity of key components to prevent replication-stress-induced DNA damage. Here, we establish a far greater role for transcriptional control in determining the outcome of replication-stress-induced events than previously suspected. Sustained E2F-dependent transcription is both required and sufficient for many crucial checkpoint functions, including fork stalling, stabilization, and resolution. Importantly, we also find that, in the context of oncogene-induced replication stress, where increased E2F activity is thought to cause replication stress, E2F activity is required to limit levels of DNA damage. These data suggest a model in which cells experiencing oncogene-induced replication stress through deregulation of E2F-dependent transcription become addicted to E2F activity to cope with high levels of replication stress.


Subject(s)
DNA Damage/genetics , DNA Replication/genetics , E2F Transcription Factors/metabolism , Transcription, Genetic , Cell Cycle Checkpoints/genetics , Cell Line , Humans , Oncogenes , Protein Biosynthesis/genetics
14.
Elife ; 3: e03475, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24986860

ABSTRACT

New data on the relationship between two proteins, cyclin D and Rb, suggest that we need to re-evaluate our understanding of how cells enter into the cell cycle.


Subject(s)
Cyclin D1/metabolism , Retinoblastoma Protein/metabolism , Animals , Humans
15.
Curr Biol ; 23(17): 1629-37, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-23954429

ABSTRACT

BACKGROUND: In eukaryotic cells, detection of replication stress results in the activation of the DNA replication checkpoint, a signaling cascade whose central players are the kinases ATR and Chk1. The checkpoint response prevents the accumulation of DNA damage and ensures cell viability by delaying progression into mitosis. However, the role and mechanism of the replication checkpoint transcriptional response in human cells, which is p53 independent, is largely unknown. RESULTS: We show that, in response to DNA replication stress, the regular E2F-dependent cell-cycle transcriptional program is maintained at high levels, and we establish the mechanisms governing such transcriptional upregulation. E2F6, a repressor of E2F-dependent G1/S transcription, replaces the activating E2Fs at promoters to repress transcription in cells progressing into S phase in unperturbed conditions. After replication stress, the checkpoint kinase Chk1 phosphorylates E2F6, leading to its dissociation from promoters. This promotes E2F-dependent transcription, which mediates cell survival by preventing DNA damage and cell death. CONCLUSIONS: This work reveals, for the first time, that the regular cell-cycle transcriptional program is part of the DNA replication checkpoint response in human cells and establishes the molecular mechanism involved. We show that maintaining high levels of G1/S cell-cycle transcription in response to replication stress contributes to two key functions of the DNA replication checkpoint response, namely, preventing genomic instability and cell death. Given the critical role of replication stress in oncogene transformation, a detailed understanding of the molecular mechanisms involved in the checkpoint response will contribute to a better insight into cancer development.


Subject(s)
Cell Cycle/genetics , DNA Replication , E2F6 Transcription Factor/physiology , Protein Kinases/physiology , Transcription, Genetic/physiology , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Damage , Humans , Promoter Regions, Genetic
17.
Cell Cycle ; 9(4): 755-60, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20107320

ABSTRACT

Senescence represents an important barrier against cellular transformation. Here we show that CAPNS1 depletion impairs senescence induction both in BJ-ET H-Ras(v12) inducible human fibroblasts upon Ras induction and in HT1080 targeted to senescence by treatment with low doses of doxorubicin. We further show that CAPNS1 depletion is coupled to reduced levels of H2AX phosphorylation, not only in Ras(v12) induced BJ-ET fibroblasts, but also in a number of cellular systems upon genotoxic stress. In particular CAPNS1 depletion affects gamma-H2AX appearance or persistence in U2OS osteosarcoma cells 24 hours after MMC addition or UV light exposure; in HT1080 upon camptothecin treatment for 4 hours and 48 hours after addition of MMC; in MDA-MB-231, 24 hours after UV light exposure and 2 hours after bleomycin addition. Overall this study unveils a novel link between calpain, cellular senescence and DNA damage response.


Subject(s)
Calpain/metabolism , Cellular Senescence , DNA Damage , Alkylating Agents/pharmacology , Calpain/genetics , Cell Line, Tumor , DNA Repair , Histones/metabolism , Humans , Mitomycin/pharmacology , Phosphorylation , RNA Interference , Ultraviolet Rays
18.
Mol Cell Biol ; 29(10): 2594-608, 2009 May.
Article in English | MEDLINE | ID: mdl-19289499

ABSTRACT

Recently, autophagy has emerged as a critical process in the control of T-cell homeostasis. Given the pivotal role of NF-kappaB in the signaling events of T cells, we have analyzed and unveiled a conserved NF-kappaB binding site in the promoter of the murine and human BECN1 autophagic gene (Atg6). Accordingly, we demonstrate that the NF-kappaB family member p65/RelA upregulates BECN1 mRNA and protein levels in different cellular systems. Moreover, p65-mediated upregulation of BECN1 is coupled to increased autophagy. The newly identified kappaB site in the BECN1 promoter specifically interacts with p65 both in vitro and in living Jurkat cells upon phorbol myristate acetate (PMA)-ionomycin stimulation, where p65 induction is coupled to BECN1 upregulation and autophagy induction. Finally, anti-CD3- and PMA-ionomycin-mediated activation of T-cell receptor signaling in peripheral T cells from lymph nodes of healthy mice results in an upregulation of BECN1 expression that can be blocked by the NF-kappaB inhibitor BAY 11-7082. Altogether, these data suggest that autophagy could represent a novel route modulated by p65 to regulate cell survival and control T-cell homeostasis.


Subject(s)
Apoptosis Regulatory Proteins , Autophagy/physiology , Membrane Proteins , NF-kappa B/metabolism , Transcription Factor RelA/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Beclin-1 , Cell Line , Homeostasis , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Jurkat Cells/physiology , Lymphocyte Activation/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Promoter Regions, Genetic , Sequence Alignment , Signal Transduction/physiology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA/genetics
19.
Autophagy ; 3(3): 235-7, 2007.
Article in English | MEDLINE | ID: mdl-17224628

ABSTRACT

Ubiquitously expressed micro- and m-calpain proteases consist of 80-kDa catalytic subunits encoded by the Capn1 and Capn2 genes, respectively, and a common 28-kDa regulatory subunit encoded by the calpain small 1 (Capns1) gene. The micro- and m-calpain proteases have been implicated in both pro- or anti-apoptotic functions. We have found that Capns1 depletion is coupled to increased sensitivity to apoptosis triggered by a number of autophagy-inducing stimuli in mammalian cells. Therefore we investigated the involvement of calpains in autophagy using MEFs derived from Capns1 knockout mice and Capns1 depleted human cells as model systems. We found that autophagy is impaired in Capns1-deficient cells by immunostaining of the endogenous autophagosome marker LC3 and electron microscopy experiments. Accordingly, the enhancement of lysosomal activity and long-lived proteins degradation, normally occurring upon starvation, are also reduced. In Capns1-depleted cells ectopic LC3 accumulates in early endosome-like vesicles that might represent a salvage pathway for protein degradation when autophagy is defective.


Subject(s)
Autophagy , Calpain/metabolism , Phagosomes/metabolism , Animals , Cell Line, Tumor , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice
20.
J Biol Chem ; 278(41): 39583-90, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-12871932

ABSTRACT

A number of different kinases have been implicated in NF-kappa B regulation and survival function. Here we investigated the molecular cross-talk between glycogen synthase kinase-3 beta (GSK-3 beta) and the p105 precursor of the NF-kappa B p50 subunit. GSK-3 beta forms an in vivo complex with and specifically phosphorylates NF-kappa B1/p105 at Ser-903 and Ser-907 in vitro. In addition, the p105 phosphorylation level is reduced in fibroblasts lacking GSK-3 beta as compared with wild-type cells. GSK-3 beta has a dual effect on p105: it stabilizes p105 under resting conditions and primes p105 for degradation upon tumor necrosis factor (TNF)-alpha treatment. Indeed, constitutive processing of p105 to p50 occurs at a higher rate in cells lacking GSK-3 beta with respect to wild-type cells and can be reduced upon reintroduction of GSK-3 beta by transfection. Moreover, p105 degradation in response to TNF-alpha is prevented in GSK-3 beta-/- fibroblasts and by a Ser to Ala point mutation on p105 at positions 903 or 907. Interestingly, the increased sensitiveness to TNF-alpha-induced death occurring in GSK-3 beta-/- fibroblasts, which is coupled to a perturbation of p50/105 ratio, can be reproduced by p105 silencing in wild-type fibroblasts.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , NF-kappa B/metabolism , Protein Precursors/metabolism , Animals , Binding Sites , Cells, Cultured , Drug Stability , Glycogen Synthase Kinase 3/deficiency , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , Mice , Mice, Knockout , Models, Biological , Mutagenesis, Site-Directed , NF-kappa B/chemistry , NF-kappa B/genetics , NF-kappa B p50 Subunit , Protein Precursors/chemistry , Protein Precursors/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL