Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38260523

ABSTRACT

Mammalian DNA replication employs several RecQ DNA helicases to orchestrate the faithful duplication of genetic information. Helicase function is often coupled to the activity of specific nucleases, but how helicase and nuclease activities are co-directed is unclear. Here we identify the inactive ubiquitin-specific protease, USP50, as a ubiquitin-binding and chromatin-associated protein required for ongoing replication, fork restart, telomere maintenance and cellular survival during replicative stress. USP50 supports WRN:FEN1 at stalled replication forks, suppresses MUS81-dependent fork collapse and restricts double-strand DNA breaks at GC-rich sequences. Surprisingly we find that cells depleted for USP50 and recovering from a replication block exhibit increased DNA2 and RECQL4 foci and that the defects in ongoing replication, poor fork restart and increased fork collapse seen in these cells are mediated by DNA2, RECQL4 and RECQL5. These data define a novel ubiquitin-dependent pathway that promotes the balance of helicase: nuclease use at ongoing and stalled replication forks.

2.
J Cell Sci ; 135(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35748225

ABSTRACT

Commonly applied super-resolution light microscopies have provided insight into subcellular processes at the nanoscale. However, imaging depth, speed, throughput and cost remain significant challenges, limiting the numbers of three-dimensional (3D) nanoscale processes that can be investigated and the number of laboratories able to undertake such analysis. Expansion microscopy (ExM) solves many of these limitations, but its application to imaging nuclear processes has been constrained by concerns of unequal nuclear expansion. Here, we demonstrate the conditions for isotropic expansion of the nucleus at a resolution equal to or better than 120-130 nm (pre-expansion). Using the DNA damage response proteins BRCA1, 53BP1 (also known as TP53BP1) and RAD51 as exemplars, we quantitatively describe the 3D nanoscale organisation of over 50,000 DNA damage response structures. We demonstrate the ability to assess chromatin-regulated events and show the simultaneous assessment of four elements. This study thus demonstrates how ExM can contribute to the investigation of nanoscale nuclear processes.


Subject(s)
Chromatin , Microscopy , Cell Nucleus , Microscopy/methods
3.
Mol Cell Oncol ; 6(6): e1656500, 2019.
Article in English | MEDLINE | ID: mdl-31692900

ABSTRACT

The breast cancer type-1 susceptibility protein (BRCA1) contributes to genome integrity through homologous recombinational DNA repair and by protecting stalled replication forks from nucleolytic degradation. We recently discovered that fork protection requires a conformational change of BRCA1 unimportant to homologous recombination repair, indicating separate roles for BRCA1 in these pathways.

4.
Front Mol Biosci ; 6: 79, 2019.
Article in English | MEDLINE | ID: mdl-31552267

ABSTRACT

DNA double-strand breaks (DSBs) occur in our cells in the context of chromatin. This type of lesion is toxic, entirely preventing genome continuity and causing cell death or terminal arrest. Several repair mechanisms can act on DNA surrounding a DSB, only some of which carry a low risk of mutation, so that which repair process is utilized is critical to the stability of genetic material of cells. A key component of repair outcome is the degree of DNA resection directed to either side of the break site. This in turn determines the subsequent forms of repair in which DNA homology plays a part. Here we will focus on chromatin and chromatin-bound complexes which constitute the "mountains" that block resection, with a particular focus on how the breast and ovarian cancer predisposition protein-1 (BRCA1) contributes to repair outcomes through overcoming these blocks.

5.
Nature ; 571(7766): 521-527, 2019 07.
Article in English | MEDLINE | ID: mdl-31270457

ABSTRACT

The integrity of genomes is constantly threatened by problems encountered by the replication fork. BRCA1, BRCA2 and a subset of Fanconi anaemia proteins protect stalled replication forks from degradation by nucleases, through pathways that involve RAD51. The contribution and regulation of BRCA1 in replication fork protection, and how this role relates to its role in homologous recombination, is unclear. Here we show that BRCA1 in complex with BARD1, and not the canonical BRCA1-PALB2 interaction, is required for fork protection. BRCA1-BARD1 is regulated by a conformational change mediated by the phosphorylation-directed prolyl isomerase PIN1. PIN1 activity enhances BRCA1-BARD1 interaction with RAD51, thereby increasing the presence of RAD51 at stalled replication structures. We identify genetic variants of BRCA1-BARD1 in patients with cancer that exhibit poor protection of nascent strands but retain homologous recombination proficiency, thus defining domains of BRCA1-BARD1 that are required for fork protection and associated with cancer development. Together, these findings reveal a BRCA1-mediated pathway that governs replication fork protection.


Subject(s)
BRCA1 Protein/chemistry , BRCA1 Protein/metabolism , DNA Replication , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , BRCA1 Protein/genetics , Cell Line, Tumor , DNA Replication/genetics , Genomic Instability/genetics , Humans , Isomerism , Mutation , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Rad51 Recombinase/metabolism
6.
Genes Dev ; 33(5-6): 333-347, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30796017

ABSTRACT

SUMOylation (small ubiquitin-like modifier) in the DNA double-strand break (DSB) response regulates recruitment, activity, and clearance of repair factors. However, our understanding of a role for deSUMOylation in this process is limited. Here we identify different mechanistic roles for deSUMOylation in homologous recombination (HR) and nonhomologous end joining (NHEJ) through the investigation of the deSUMOylase SENP2. We found that regulated deSUMOylation of MDC1 prevents excessive SUMOylation and its RNF4-VCP mediated clearance from DSBs, thereby promoting NHEJ. In contrast, we show that HR is differentially sensitive to SUMO availability and SENP2 activity is needed to provide SUMO. SENP2 is amplified as part of the chromosome 3q amplification in many cancers. Increased SENP2 expression prolongs MDC1 focus retention and increases NHEJ and radioresistance. Collectively, our data reveal that deSUMOylation differentially primes cells for responding to DSBs and demonstrates the ability of SENP2 to tune DSB repair responses.


Subject(s)
Cysteine Endopeptidases/metabolism , DNA End-Joining Repair/genetics , DNA Repair/genetics , Homologous Recombination/genetics , Sumoylation/genetics , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/radiation effects , Cysteine Endopeptidases/genetics , DNA Breaks, Double-Stranded , HEK293 Cells , HeLa Cells , Humans , Infrared Rays , Nuclear Proteins/metabolism , Radiation Tolerance/genetics , Signal Transduction/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Valosin Containing Protein/metabolism
7.
J Biol Chem ; 294(2): 424-436, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30373771

ABSTRACT

Ubiquitin-specific proteases (USPs) reverse ubiquitination and regulate virtually all cellular processes. Defined noncatalytic domains in USP4 and USP15 are known to interact with E3 ligases and substrate recruitment factors. No such interactions have been reported for these domains in the paralog USP11, a key regulator of DNA double-strand break repair by homologous recombination. We hypothesized that USP11 domains adjacent to its protease domain harbor unique peptide-binding sites. Here, using a next-generation phage display (NGPD) strategy, combining phage display library screening with next-generation sequencing, we discovered unique USP11-interacting peptide motifs. Isothermal titration calorimetry disclosed that the highest affinity peptides (KD of ∼10 µm) exhibit exclusive selectivity for USP11 over USP4 and USP15 in vitro Furthermore, a crystal structure of a USP11-peptide complex revealed a previously unknown binding site in USP11's noncatalytic ubiquitin-like (UBL) region. This site interacted with a helical motif and is absent in USP4 and USP15. Reporter assays using USP11-WT versus a binding pocket-deficient double mutant disclosed that this binding site modulates USP11's function in homologous recombination-mediated DNA repair. The highest affinity USP11 peptide binder fused to a cellular delivery sequence induced significant nuclear localization and cell cycle arrest in S phase, affecting the viability of different mammalian cell lines. The USP11 peptide ligands and the paralog-specific functional site in USP11 identified here provide a framework for the development of new biochemical tools and therapeutic agents. We propose that an NGPD-based strategy for identifying interacting peptides may be applied also to other cellular targets.


Subject(s)
Peptides/chemistry , Peptides/metabolism , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Crystallography, X-Ray , DNA Repair , Homologous Recombination , Humans , Kinetics , Ligands , Mice , Molecular Sequence Data , Peptides/genetics , Protein Domains , Thiolester Hydrolases/genetics , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitination
8.
Nat Commun ; 9(1): 229, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335415

ABSTRACT

BRCA1-BARD1-catalyzed ubiquitination of histone H2A is an important regulator of the DNA damage response, priming chromatin for repair by homologous recombination. However, no specific deubiquitinating enzymes (DUBs) are known to antagonize this function. Here we identify ubiquitin specific protease-48 (USP48) as a H2A DUB, specific for the C-terminal BRCA1 ubiquitination site. Detailed biochemical analysis shows that an auxiliary ubiquitin, an additional ubiquitin that itself does not get cleaved, modulates USP48 activity, which has possible implications for its regulation in vivo. In cells we reveal that USP48 antagonizes BRCA1 E3 ligase function and in BRCA1-proficient cells loss of USP48 results in positioning 53BP1 further from the break site and in extended resection lengths. USP48 repression confers a survival benefit to cells treated with camptothecin and its activity acts to restrain gene conversion and mutagenic single-strand annealing. We propose that USP48 promotes genome stability by antagonizing BRCA1 E3 ligase function.


Subject(s)
BRCA1 Protein/metabolism , Histones/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin/metabolism , Animals , BRCA1 Protein/genetics , Base Sequence , Cell Line, Tumor , Cells, Cultured , DNA Repair , HeLa Cells , Humans , Kinetics , Mice, Knockout , RNA Interference , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination
9.
Nucleus ; 8(2): 116-125, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28032817

ABSTRACT

The protein product of the breast and ovarian cancer gene, BRCA1, is part of an obligate heterodimer with BARD1. Together these RING bearing proteins act as an E3 ubiquitin ligase. Several functions have been attributed to BRCA1 that contribute to genome integrity but which of these, if any, require this enzymatic function was unclear. Here we review recent studies clarifying the role of BRCA1 E3 ubiquitin ligase in DNA repair. Perhaps the most surprising finding is the narrow range of BRCA1 functions this activity relates to. Remarkably ligase activity promotes chromatin remodelling and 53BP1 positioning through the remodeller SMARCAD1, but the activity is dispensable for the cellular survival in response to cisplatin or replication stressing agents. Implications for therapy response and tumor susceptibility are discussed.


Subject(s)
BRCA1 Protein/metabolism , DNA Repair , Ubiquitin-Protein Ligases/metabolism , Animals , BRCA1 Protein/chemistry , Chromatin/metabolism , Gene Silencing , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
10.
Nat Struct Mol Biol ; 23(7): 647-55, 2016 07.
Article in English | MEDLINE | ID: mdl-27239795

ABSTRACT

The opposing activities of 53BP1 and BRCA1 influence pathway choice in DNA double-strand-break repair. How BRCA1 counteracts the inhibitory effect of 53BP1 on DNA resection and homologous recombination is unknown. Here we identify the site of BRCA1-BARD1 required for priming ubiquitin transfer from E2∼ubiquitin and demonstrate that BRCA1-BARD1's ubiquitin ligase activity is required for repositioning 53BP1 on damaged chromatin. We confirm H2A ubiquitination by BRCA1-BARD1 and show that an H2A-ubiquitin fusion protein promotes DNA resection and repair in BARD1-deficient cells. BRCA1-BARD1's function in homologous recombination requires the chromatin remodeler SMARCAD1. SMARCAD1 binding to H2A-ubiquitin and optimal localization to sites of damage and activity in DNA repair requires its ubiquitin-binding CUE domains. SMARCAD1 is required for 53BP1 repositioning, and the need for SMARCAD1 in olaparib or camptothecin resistance is alleviated by 53BP1 loss. Thus, BRCA1-BARD1 ligase activity and subsequent SMARCAD1-dependent chromatin remodeling are critical regulators of DNA repair.


Subject(s)
BRCA1 Protein/genetics , Chromatin/metabolism , DNA Helicases/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Recombinational DNA Repair , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , BRCA1 Protein/metabolism , Binding Sites , Camptothecin/pharmacology , Chromatin/chemistry , Chromatin/drug effects , Cloning, Molecular , DNA Breaks, Double-Stranded , DNA Cleavage/drug effects , DNA Helicases/metabolism , DNA, Neoplasm/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Models, Molecular , Phthalazines/pharmacology , Piperazines/pharmacology , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
11.
EMBO Rep ; 14(11): 975-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24018422

ABSTRACT

SUMO conjugation is known to occur in response to double-stranded DNA breaks in mammalian cells, but whether SUMO deconjugation has a role remains unclear. Here, we show that the SUMO/Sentrin/Smt3-specific peptidase, SENP7, interacts with the chromatin repressive KRAB-associated protein 1 (KAP1) through heterochromatin protein 1 alpha (HP1α). SENP7 promotes the removal of SUMO2/3 from KAP1 and regulates the interaction of the chromatin remodeler CHD3 with chromatin. Consequently, in the presence of CHD3, SENP7 is required for chromatin relaxation in response to DNA damage, for homologous recombination repair and for cellular resistance to DNA-damaging agents. Thus, deSUMOylation by SENP7 is required to promote a permissive chromatin environment for DNA repair.


Subject(s)
Chromatin/metabolism , Endopeptidases/metabolism , Recombinational DNA Repair , Amino Acid Motifs , Amino Acid Sequence , Chromatin Assembly and Disassembly , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone , DNA Breaks, Double-Stranded , DNA Damage , DNA Helicases/metabolism , Endopeptidases/chemistry , HEK293 Cells , HeLa Cells , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Molecular Sequence Data , Protein Binding , SUMO-1 Protein/metabolism
12.
EMBO J ; 31(19): 3918-34, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-22909820

ABSTRACT

The regulation of Ubiquitin (Ub) conjugates generated by the complex network of proteins that promote the mammalian DNA double-strand break (DSB) response is not fully understood. We show here that the Ub protease POH1/rpn11/PSMD14 resident in the 19S proteasome regulatory particle is required for processing poly-Ub formed in the DSB response. Proteasome activity is required to restrict tudor domain-dependent 53BP1 accumulation at sites of DNA damage. This occurs both through antagonism of RNF8/RNF168-mediated lysine 63-linked poly-Ub and through the promotion of JMJD2A retention on chromatin. Consistent with this role POH1 acts in opposition to RNF8/RNF168 to modulate end-joining DNA repair. Additionally, POH1 acts independently of 53BP1 in homologous recombination repair to promote RAD51 loading. Accordingly, POH1-deficient cells are sensitive to DNA damaging agents. These data demonstrate that proteasomal POH1 is a key de-ubiquitinating enzyme that regulates ubiquitin conjugates generated in response to damage and that several aspects of the DSB response are regulated by the proteasome.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/physiology , Proteasome Endopeptidase Complex/metabolism , Trans-Activators/metabolism , Cell Line , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Polyubiquitin/metabolism , Rad51 Recombinase/metabolism , Tumor Suppressor p53-Binding Protein 1 , Ubiquitin-Protein Ligases/metabolism
13.
Nature ; 462(7275): 886-90, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-20016594

ABSTRACT

Mutations in BRCA1 are associated with a high risk of breast and ovarian cancer. BRCA1 participates in the DNA damage response and acts as a ubiquitin ligase. However, its regulation remains poorly understood. Here we report that BRCA1 is modified by small ubiquitin-like modifier (SUMO) in response to genotoxic stress, and co-localizes at sites of DNA damage with SUMO1, SUMO2/3 and the SUMO-conjugating enzyme Ubc9. PIAS SUMO E3 ligases co-localize with and modulate SUMO modification of BRCA1, and are required for BRCA1 ubiquitin ligase activity in cells. In vitro SUMO modification of the BRCA1/BARD1 heterodimer greatly increases its ligase activity, identifying it as a SUMO-regulated ubiquitin ligase (SRUbL). Further, PIAS SUMO ligases are required for complete accumulation of double-stranded DNA (dsDNA) damage-repair proteins subsequent to RNF8 accrual, and for proficient double-strand break repair. These data demonstrate that the SUMOylation pathway plays a significant role in mammalian DNA damage response.


Subject(s)
BRCA1 Protein/metabolism , DNA Damage , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , DNA Breaks, Double-Stranded , DNA Repair , HeLa Cells , Histones/metabolism , Humans , Protein Inhibitors of Activated STAT/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
14.
Mol Cell Biol ; 29(24): 6380-90, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19822666

ABSTRACT

As well as providing a structural framework, the actin cytoskeleton plays integral roles in cell death, survival, and proliferation. The disruption of the actin cytoskeleton results in the activation of the c-Jun N-terminal kinase (JNK) stress-activated protein kinase (SAPK) pathway; however, the sensor of actin integrity that couples to the JNK pathway has not been characterized in mammalian cells. We now report that the mammalian Ste20-like (MST) kinases mediate the activation of the JNK pathway in response to the disruption of the actin cytoskeleton. One consequence of actin disruption is the JNK-mediated stabilization of p21(Waf1/Cip1) (p21) via the phosphorylation of Thr57. The expression of MST1 or MST2 was sufficient to stabilize p21 in a JNK- and Thr57-dependent manner, while the stabilization of p21 by actin disruption required MST activity. These data indicate that, in addition to being components of the Salvador-Warts-Hippo tumor suppressor network and binding partners of c-Raf and the RASSF1A tumor suppressor, MST kinases serve to monitor cytoskeletal integrity and couple via the JNK SAPK pathway to the regulation of a key cell cycle regulatory protein.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoskeleton/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Protein Serine-Threonine Kinases/metabolism , Actins/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p21/genetics , Emetine/metabolism , Enzyme Activation , Enzyme Stability , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mitogen-Activated Protein Kinase 8/genetics , Mutagenesis, Site-Directed , NIH 3T3 Cells , Protein Serine-Threonine Kinases/genetics , Protein Synthesis Inhibitors/metabolism , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine-Threonine Kinase 3 , Signal Transduction/physiology
15.
Cell Signal ; 20(11): 1986-94, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18664382

ABSTRACT

The conditional kinase DeltaMEKK3:ER allows activation of JNK, p38 and ERK1/2 without overt cellular stress or damage and has proved useful in understanding how these pathways regulate apoptosis and cell cycle progression. We have previously shown that activation of DeltaMEKK3:ER causes a sustained G(1) cell cycle arrest which requires p21(CIP1), with ERK1/2 and p38 cooperating to promote p21(CIP1) expression. In cells lacking p21(CIP1), DeltaMEKK3:ER causes only a transient delay in cell cycle re-entry. We now show that this delay in cell cycle re-entry is due to a reduction in cyclin D1 levels. Activation of DeltaMEKK3:ER promotes the proteasome-dependent turnover of cyclin D1; this requires phosphorylation of threonine 286 (T(286)) and expression of cyclin D1T(286)A rescues the delay in G(1)/S progression. DeltaMEKK3:ER-dependent phosphorylation of T(286) does not appear to be mediated by GSK3beta but requires activation of the ERK1/2 and p38 pathways. ERK1/2 can physically associate with cyclin D1 but activation of ERK1/2 alone is not sufficient for phosphorylation of T(286). Rather, cyclin D1 phosphorylation appears to require coincident activation of ERK1/2 and p38. Thus activation of DeltaMEKK3:ER promotes a sustained G(1) cell cycle arrest by a bipartite mechanism involving the rapid destruction of cyclin D1 and the slower more prolonged expression of p21(CIP1). This has parallels with the bipartite response to ionizing radiation and p53-independent mechanisms of G(1) cell cycle arrest in simple organisms such as yeast.


Subject(s)
Cyclin D1/metabolism , G1 Phase , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Amino Acid Motifs , Animals , Cell Line , Cyclin D1/chemistry , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Enzyme Activation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mutation/genetics , Phosphorylation , Phosphothreonine/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , raf Kinases/metabolism
16.
Oncogene ; 23(19): 3284-95, 2004 Apr 22.
Article in English | MEDLINE | ID: mdl-14981547

ABSTRACT

To study the mechanisms by which mitogen- and stress-activated protein kinases regulate cell cycle re-entry, we have used a panel of conditional kinases that stimulate defined MAPK or SAPK cascades. Activation of DeltaMEKK3:ER* during serum restimulation of quiescent cells causes a strong activation of JNK1 and p38alpha but only a modest potentiation of serum-stimulated ERK1/2 activity. In CCl39 cells this promoted a sustained G1 arrest that correlated with decreased expression of cyclin D1 and Cdc25A, increased expression of p21CIP1 and inhibition of CDK2 activity. In Rat-1 cells, in which p21(CIP1) expression is silenced by methylation, DeltaMEKK3:ER* activation caused only a transient delay in the S phase entry rather than a sustained G1 arrest. Furthermore, p21CIP1-/- 3T3 cells were defective for the DeltaMEKK3:ER*-induced G1 cell cycle arrest compared to their wild-type counterparts. These results suggest that activated DeltaMEKK3:ER* inhibits the G1 --> S progression by two kinetically distinct mechanisms, with expression of p21CIP1 being required to ensure a sustained G1 cell cycle arrest. The ERK1/2 and p38alphabeta pathways cooperated to induce p21CIP1 expression and inhibition of p38alphabeta caused a partial reversal of the cell cycle arrest. In contrast, selective activation of ERK1/2 by DeltaRaf-1:ER* did not inhibit serum stimulated cell cycle re-entry. Finally, selective activation of JNK by DeltaMEKK1:ER* failed to inhibit cell cycle re-entry, even in cells that retained wild-type p53, arguing against a major role for JNK alone in antagonizing the G1 --> S transition.


Subject(s)
Cyclins/physiology , G1 Phase , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinases/physiology , Tamoxifen/analogs & derivatives , 3T3 Cells , Animals , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/pharmacology , Mice , Mitogen-Activated Protein Kinase 3 , Phosphorylation , RNA, Messenger/analysis , Rats , Retinoblastoma Protein/metabolism , S Phase , Tamoxifen/pharmacology , Tumor Suppressor Protein p53/physiology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...