Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Rep ; 42(11): 113427, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37950866

ABSTRACT

Abasic sites are common DNA lesions stalling polymerases and threatening genome stability. When located in single-stranded DNA (ssDNA), they are shielded from aberrant processing by 5-hydroxymethyl cytosine, embryonic stem cell (ESC)-specific (HMCES) via a DNA-protein crosslink (DPC) that prevents double-strand breaks. Nevertheless, HMCES-DPCs must be removed to complete DNA repair. Here, we find that DNA polymerase α inhibition generates ssDNA abasic sites and HMCES-DPCs. These DPCs are resolved with a half-life of approximately 1.5 h. HMCES can catalyze its own DPC self-reversal reaction, which is dependent on glutamate 127 and is favored when the ssDNA is converted to duplex DNA. When the self-reversal mechanism is inactivated in cells, HMCES-DPC removal is delayed, cell proliferation is slowed, and cells become hypersensitive to DNA damage agents that increase AP (apurinic/apyrimidinic) site formation. In these circumstances, proteolysis may become an important mechanism of HMCES-DPC resolution. Thus, HMCES-DPC formation followed by self-reversal is an important mechanism for ssDNA AP site management.


Subject(s)
DNA Damage , Proteins , Proteins/genetics , DNA Replication , DNA Repair , DNA/genetics , DNA, Single-Stranded
2.
Proc Natl Acad Sci U S A ; 120(39): e2303752120, 2023 09 26.
Article in English | MEDLINE | ID: mdl-37722039

ABSTRACT

Isochromosomes are mirror-imaged chromosomes with simultaneous duplication and deletion of genetic material which may contain two centromeres to create isodicentric chromosomes. Although isochromosomes commonly occur in cancer and developmental disorders and promote genome instability, mechanisms that prevent isochromosomes are not well understood. We show here that the tumor suppressor and methyltransferase SETD2 is essential to prevent these errors. Using cellular and cytogenetic approaches, we demonstrate that loss of SETD2 or its epigenetic mark, histone H3 lysine 36 trimethylation (H3K36me3), results in the formation of isochromosomes as well as isodicentric and acentric chromosomes. These defects arise during DNA replication and are likely due to faulty homologous recombination by RAD52. These data provide a mechanism for isochromosome generation and demonstrate that SETD2 and H3K36me3 are essential to prevent the formation of this common mutable chromatin structure known to initiate a cascade of genomic instability in cancer.


Subject(s)
Isochromosomes , Humans , Centromere , Chromosome Aberrations , Cytogenetics , DNA Replication , Genomic Instability
3.
bioRxiv ; 2023 Jun 14.
Article in English | MEDLINE | ID: mdl-37398432

ABSTRACT

Abasic sites are common DNA lesions that stall polymerases and threaten genome stability. When located in single-stranded DNA (ssDNA), they are shielded from aberrant processing by HMCES via a DNA-protein crosslink (DPC) that prevents double-strand breaks. Nevertheless, the HMCES-DPC must be removed to complete DNA repair. Here, we found that DNA polymerase α inhibition generates ssDNA abasic sites and HMCES-DPCs. These DPCs are resolved with a half-life of approximately 1.5 hours. Resolution does not require the proteasome or SPRTN protease. Instead, HMCES-DPC self-reversal is important for resolution. Biochemically, self-reversal is favored when the ssDNA is converted to duplex DNA. When the self-reversal mechanism is inactivated, HMCES-DPC removal is delayed, cell proliferation is slowed, and cells become hypersensitive to DNA damage agents that increase AP site formation. Thus, HMCES-DPC formation followed by self-reversal is an important mechanism for ssDNA AP site management.

4.
Genes Dev ; 34(19-20): 1392-1405, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32883681

ABSTRACT

TRF1 facilitates the replication of telomeric DNA in part by recruiting the BLM helicase, which can resolve G-quadruplexes on the lagging-strand template. Lagging-strand telomeres lacking TRF1 or BLM form fragile telomeres-structures that resemble common fragile sites (CFSs)-but how they are formed is not known. We report that analogous to CFSs, fragile telomeres in BLM-deficient cells involved double-strand break (DSB) formation, in this case by the SLX4/SLX1 nuclease. The DSBs were repaired by POLD3/POLD4-dependent break-induced replication (BIR), resulting in fragile telomeres containing conservatively replicated DNA. BIR also promoted fragile telomere formation in cells with FokI-induced telomeric DSBs and in alternative lengthening of telomeres (ALT) cells, which have spontaneous telomeric damage. BIR of telomeric DSBs competed with PARP1-, LIG3-, and XPF-dependent alternative nonhomologous end joining (alt-NHEJ), which did not generate fragile telomeres. Collectively, these findings indicate that fragile telomeres can arise from BIR-mediated repair of telomeric DSBs.


Subject(s)
Chromosome Fragile Sites/genetics , DNA Breaks, Double-Stranded , DNA Replication , RecQ Helicases/genetics , RecQ Helicases/metabolism , Telomere/pathology , Animals , Cell Line , Cells, Cultured , DNA Repair , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Fibroblasts , Humans , Mice , Recombinases/genetics , Recombinases/metabolism
5.
Cell Rep ; 31(9): 107705, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492421

ABSTRACT

5-Hydroxymethylcytosine (5hmC) binding, ES-cell-specific (HMCES) crosslinks to apurinic or apyrimidinic (AP, abasic) sites in single-strand DNA (ssDNA). To determine whether HMCES responds to the ssDNA abasic site in cells, we exploited the activity of apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3A (APOBEC3A). APOBEC3A preferentially deaminates cytosines to uracils in ssDNA, which are then converted to abasic sites by uracil DNA glycosylase. We find that HMCES-deficient cells are hypersensitive to nuclear APOBEC3A localization. HMCES relocalizes to chromatin in response to nuclear APOBEC3A and protects abasic sites from processing into double-strand breaks (DSBs). Abasic sites induced by APOBEC3A slow both leading and lagging strand synthesis, and HMCES prevents further slowing of the replication fork by translesion synthesis (TLS) polymerases zeta (Polζ) and kappa (Polκ). Thus, our study provides direct evidence that HMCES responds to ssDNA abasic sites in cells to prevent DNA cleavage and balance the engagement of TLS polymerases.


Subject(s)
Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , Cytidine Deaminase/genetics , DNA Replication , DNA, Single-Stranded/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Deamination , Endonucleases/antagonists & inhibitors , Endonucleases/genetics , Endonucleases/metabolism , Humans , Multifunctional Enzymes/antagonists & inhibitors , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Uracil/metabolism , Uracil-DNA Glycosidase/metabolism
6.
PLoS Biol ; 18(1): e3000594, 2020 01.
Article in English | MEDLINE | ID: mdl-31895940

ABSTRACT

Alpha thalassemia/mental retardation syndrome X-linked chromatin remodeler (ATRX), a DAXX (death domain-associated protein) interacting protein, is often lost in cells using the alternative lengthening of telomeres (ALT) pathway, but it is not known how ATRX loss leads to ALT. We report that ATRX deletion from mouse cells altered the repair of telomeric double-strand breaks (DSBs) and induced ALT-like phenotypes, including ALT-associated promyelocytic leukemia (PML) bodies (APBs), telomere sister chromatid exchanges (T-SCEs), and extrachromosomal telomeric signals (ECTSs). Mechanistically, we show that ATRX affects telomeric DSB repair by promoting cohesion of sister telomeres and that loss of ATRX in ALT cells results in diminished telomere cohesion. In addition, we document a role for DAXX in the repair of telomeric DSBs. Removal of telomeric cohesion in combination with DAXX deficiency recapitulates all telomeric DSB repair phenotypes associated with ATRX loss. The data reveal that ATRX has an effect on telomeric DSB repair and that this role involves both telomere cohesion and a DAXX-dependent pathway.


Subject(s)
Co-Repressor Proteins/physiology , DNA Breaks, Double-Stranded , DNA Repair/genetics , Molecular Chaperones/physiology , Sister Chromatid Exchange/genetics , Telomere/genetics , X-linked Nuclear Protein/physiology , Animals , Cells, Cultured , Embryo, Mammalian , Female , HeLa Cells , Humans , Male , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Mice , Mice, Knockout , Signal Transduction/genetics , Telomere/metabolism , Telomere Homeostasis/genetics , alpha-Thalassemia/genetics , alpha-Thalassemia/pathology
7.
Proc Natl Acad Sci U S A ; 112(48): 14864-9, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26578802

ABSTRACT

The SMARCAL1 (SWI/SNF related, matrix-associated, actin-dependent, regulator of chromatin, subfamily A-like 1) DNA translocase is one of several related enzymes, including ZRANB3 (zinc finger, RAN-binding domain containing 3) and HLTF (helicase-like transcription factor), that are recruited to stalled replication forks to promote repair and restart replication. These enzymes can perform similar biochemical reactions such as fork reversal; however, genetic studies indicate they must have unique cellular activities. Here, we present data showing that SMARCAL1 has an important function at telomeres, which present an endogenous source of replication stress. SMARCAL1-deficient cells accumulate telomere-associated DNA damage and have greatly elevated levels of extrachromosomal telomere DNA (C-circles). Although these telomere phenotypes are often found in tumor cells using the alternative lengthening of telomeres (ALT) pathway for telomere elongation, SMARCAL1 deficiency does not yield other ALT phenotypes such as elevated telomere recombination. The activity of SMARCAL1 at telomeres can be separated from its genome-maintenance activity in bulk chromosomal replication because it does not require interaction with replication protein A. Finally, this telomere-maintenance function is not shared by ZRANB3 or HLTF. Our results provide the first identification, to our knowledge, of an endogenous source of replication stress that requires SMARCAL1 for resolution and define differences between members of this class of replication fork-repair enzymes.


Subject(s)
Chromosomes, Human/metabolism , DNA Helicases/metabolism , DNA Replication/physiology , Telomere Homeostasis/physiology , Telomere/metabolism , Animals , Chromosomes, Human/genetics , DNA Damage/physiology , DNA Helicases/genetics , HeLa Cells , Humans , Mice , Recombination, Genetic/physiology , Telomere/genetics
8.
PLoS Genet ; 8(7): e1002772, 2012.
Article in English | MEDLINE | ID: mdl-22829774

ABSTRACT

The Alternative Lengthening of Telomeres (ALT) pathway is a telomerase-independent pathway for telomere maintenance that is active in a significant subset of human cancers and in vitro immortalized cell lines. ALT is thought to involve templated extension of telomeres through homologous recombination, but the genetic or epigenetic changes that unleash ALT are not known. Recently, mutations in the ATRX/DAXX chromatin remodeling complex and histone H3.3 were found to correlate with features of ALT in pancreatic neuroendocrine cancers, pediatric glioblastomas, and other tumors of the central nervous system, suggesting that these mutations might contribute to the activation of the ALT pathway in these cancers. We have taken a comprehensive approach to deciphering ALT by applying genomic, molecular biological, and cell biological approaches to a panel of 22 ALT cell lines, including cell lines derived in vitro. Here we show that loss of ATRX protein and mutations in the ATRX gene are hallmarks of ALT-immortalized cell lines. In addition, ALT is associated with extensive genome rearrangements, marked micronucleation, defects in the G2/M checkpoint, and altered double-strand break (DSB) repair. These attributes will facilitate the diagnosis and treatment of ALT positive human cancers.


Subject(s)
DNA Helicases/genetics , Histones , Nuclear Proteins/genetics , Telomere Homeostasis/genetics , Telomere/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Chromatin Assembly and Disassembly/genetics , Co-Repressor Proteins , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA Helicases/metabolism , DNA Repair/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Genomic Instability , HeLa Cells , Histones/genetics , Histones/metabolism , Homologous Recombination , Humans , Molecular Chaperones , Nuclear Proteins/metabolism , Signal Transduction , Telomerase/genetics , Telomere/metabolism , X-linked Nuclear Protein
9.
J Biol Chem ; 285(19): 14565-71, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20233726

ABSTRACT

DNA damage repair and checkpoint responses prevent genome instability and provide a barrier to the development of cancer. Inherited mutations in DNA damage response (DDR) genes such as those that encode the homologous recombination (HR) proteins BRCA1 and BRCA2 cause cancer predisposition syndromes. PARP inhibitors are an exciting new class of targeted therapy for treating patients with HR repair-defective tumors. In this study, we use an RNAi screen to identify genes that when silenced cause synthetic lethality with the PARP inhibitor AZD2281. This screen identified the deubiquitylating enzyme USP11 as a participant in HR repair of DNA double-strand breaks. Silencing USP11 with siRNA leads to spontaneous DDR activation in otherwise undamaged cells and hypersensitivity to PARP inhibition, ionizing radiation, and other genotoxic stress agents. Moreover, we demonstrate that HR repair is defective in USP11-silenced cells. Finally, the recruitment of a subset of double-strand break repair proteins including RAD51 and 53BP1 to repair foci is misregulated in the absence of USP11 catalytic activity. Thus, our synthetic lethal approach identified USP11 as a component of the HR double-strand break repair pathway.


Subject(s)
DNA Breaks, Double-Stranded , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/genetics , RNA, Small Interfering/pharmacology , Thiolester Hydrolases/metabolism , Blotting, Western , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Repair , Fluorescent Antibody Technique , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Poly(ADP-ribose) Polymerases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Radiation, Ionizing , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Thiolester Hydrolases/genetics , Tumor Cells, Cultured , Tumor Stem Cell Assay , Tumor Suppressor p53-Binding Protein 1
10.
Proc Natl Acad Sci U S A ; 106(46): 19304-9, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19889979

ABSTRACT

The DNA damage response (DDR) has a critical role in maintaining genome integrity and serves as a barrier to tumorigenesis by promoting cell-cycle arrest, DNA repair, and apoptosis. The DDR is activated not only by genotoxic agents that induce DNA damage, but also during aberrant cell-division cycles caused by activated oncogenes and inactivated tumor suppressors. Here we use RNAi and cDNA overexpression screens in human cells to identify genes that, when deregulated, lead to activation of the DDR. The RNAi screen identified 73 genes that, when silenced in at least two cell types, cause DDR activation. Silencing several of these genes also caused an increased frequency of micronuclei, a marker of genetically unstable cells. The cDNA screen identified 97 genes that when overexpressed induce DDR activation in the absence of any exogenous genotoxic agent, with an overrepresentation of genes linked to cancer. Secondary RNAi screens identified CDK2-interacting protein (CINP) as a cell-cycle checkpoint protein. CINP interacts with ATR-interacting protein and regulates ATR-dependent signaling, resistance to replication stress, and G2 checkpoint integrity.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Genome, Human , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/genetics , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Genomics , HeLa Cells , Humans , Two-Hybrid System Techniques
11.
Genes Dev ; 23(20): 2405-14, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19793861

ABSTRACT

Mutations in SMARCAL1 (HARP) cause Schimke immunoosseous dysplasia (SIOD). The mechanistic basis for this disease is unknown. Using functional genomic screens, we identified SMARCAL1 as a genome maintenance protein. Silencing and overexpression of SMARCAL1 leads to activation of the DNA damage response during S phase in the absence of any genotoxic agent. SMARCAL1 contains a Replication protein A (RPA)-binding motif similar to that found in the replication stress response protein TIPIN (Timeless-Interacting Protein), which is both necessary and sufficient to target SMARCAL1 to stalled replication forks. RPA binding is critical for the cellular function of SMARCAL1; however, it is not necessary for the annealing helicase activity of SMARCAL1 in vitro. An SIOD-associated SMARCAL1 mutant fails to prevent replication-associated DNA damage from accumulating in cells in which endogenous SMARCAL1 is silenced. Ataxia-telangiectasia mutated (ATM), ATM and Rad3-related (ATR), and DNA-dependent protein kinase (DNA-PK) phosphorylate SMARCAL1 in response to replication stress. Loss of SMARCAL1 activity causes increased RPA loading onto chromatin and persistent RPA phosphorylation after a transient exposure to replication stress. Furthermore, SMARCAL1-deficient cells are hypersensitive to replication stress agents. Thus, SMARCAL1 is a replication stress response protein, and the pleiotropic phenotypes of SIOD are at least partly due to defects in genome maintenance during DNA replication.


Subject(s)
DNA Helicases/metabolism , DNA Replication , Genomic Instability , Cell Line , Chromatin , Gene Expression Regulation , HeLa Cells , Humans , Mutation , Osteochondrodysplasias/genetics , Osteochondrodysplasias/physiopathology , Phosphorylation , Protein Binding , Replication Protein A/metabolism
12.
DNA Repair (Amst) ; 8(9): 1004-8, 2009 Sep 02.
Article in English | MEDLINE | ID: mdl-19464237

ABSTRACT

Kinases in the phosphoinositide three-kinase-related kinase (PIKK) family include ATM (ataxia-telangiectasia mutated), ATR (ATM- and Rad3-related), DNA-PKcs (DNA-dependent protein kinase catalytic subunit), mTOR (mammalian target of rapamycin), and SMG1 (suppressor with morphological effect on genitalia family member). These atypical protein kinases regulate DNA damage responses, nutrient-dependent signaling, and nonsense-mediated mRNA decay. This review focuses on the mechanisms regulating the PIKK family with a strong emphasis on the DNA damage regulated kinases. We outline common regulatory themes and suggest how discoveries about the regulation of one PIKK can be informative for the other family members.


Subject(s)
Protein Kinases/metabolism , Animals , Enzyme Activators/metabolism , Humans , Protein Kinases/chemistry , Protein Processing, Post-Translational , Protein Transport
13.
Mol Cell Biol ; 26(21): 7977-90, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16940174

ABSTRACT

DDB1, a component of a Cul4A ubiquitin ligase complex, promotes nucleotide excision repair (NER) and regulates DNA replication. We have investigated the role of human DDB1 in maintaining genome stability. DDB1-depleted cells accumulate DNA double-strand breaks in widely dispersed regions throughout the genome and have activated ATM and ATR cell cycle checkpoints. Depletion of Cul4A yields similar phenotypes, indicating that an E3 ligase function of DDB1 is important for genome maintenance. In contrast, depletion of DDB2, XPA, or XPC does not cause activation of DNA damage checkpoints, indicating that defects in NER are not involved. One substrate of DDB1-Cul4A that is crucial for preventing genome instability is Cdt1. DDB1-depleted cells exhibit increased levels of Cdt1 protein and rereplication, despite containing other Cdt1 regulatory mechanisms. The rereplication, accumulation of DNA damage, and activation of checkpoint responses in DDB1-depleted cells require entry into S phase and are partially, but not completely, suppressed by codepletion of Cdt1. Therefore, DDB1 prevents DNA lesions from accumulating in replicating human cells, in part by regulating Cdt1 degradation.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Genome, Human , Genomic Instability , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Line , Chromosomes, Human , Cullin Proteins/metabolism , DNA Repair , DNA Replication , DNA-Binding Proteins/genetics , Genes, cdc , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xeroderma Pigmentosum , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum Group A Protein/metabolism
14.
Exp Cell Res ; 296(2): 285-93, 2004 Jun 10.
Article in English | MEDLINE | ID: mdl-15149858

ABSTRACT

The v-ErbB retroviral oncogene is a transduced, mutated copy of the avian EGF receptor gene, and its expression is sufficient to induce tumor formation in vivo. The structural alterations that release the oncogenic potential of the v-ErbB oncogene are similar to EGFR gene mutations described in human tumors. Thus, the study of v-ErbB tumor biology offers a useful model through which we can gain insight into the mechanism of EGFR-induced malignancies. Despite years of study, however, questions remain regarding the domains of v-ErbB required for oncogenicity. We sought to clarify the role of the transmembrane domain of v-ErbB during transformation using S3-v-ErbB, an acutely transforming retroviral oncogene isolated from avian sarcomas. Infection of primary fibroblasts with a retroviral vector containing S3-v-ErbB results in the formation of a transformation-associated phosphoprotein signaling complex, soft agar colony formation, and the rapid induction of highly vascularized sarcomas in vivo. To address contribution of the transmembrane domain of S3-v-ErbB during these processes, we constructed a mutant version of this oncogene with a precise deletion in this domain. Specifically, the S3-v-ErbB-TM- mutant was created through an in-frame deletion of the entire transmembrane domain. Primary fibroblasts expressing this S3-v-ErbB-TM- mutant fail to form a characteristic transformation-associated phosphoprotein complex and do not grow in an anchorage-independent manner. In addition, day-old chicks injected with a helper-independent retrovirus expressing the S3-v-ErbB-TM- mutant exhibit only limited tumor formation in vivo. These results demonstrate that the transmembrane domain and, consequently membrane localization, are essential for S3-v-ErbB-mediated transformation.


Subject(s)
Cell Membrane/metabolism , Cell Transformation, Neoplastic , Oncogene Proteins v-erbB/metabolism , Oncogene Proteins v-erbB/physiology , Animals , Animals, Newborn , Cell Line , Chick Embryo , Chickens , Fibroblasts , Ligands , Mutagenesis, Site-Directed , Mutation , Neoplasms/etiology , Oncogene Proteins v-erbB/administration & dosage , Protein Transport , Transformation, Genetic
15.
Oncogene ; 22(43): 6679-89, 2003 Oct 02.
Article in English | MEDLINE | ID: mdl-14555981

ABSTRACT

Mutations within members of the EGF/ErbB receptor family frequently release the oncogenic potential of these receptors, resulting in the activation of downstream signaling events independent of ligand regulatory constraints. We previously have demonstrated that the signal transduction events originating from S3-v-ErbB, a ligand-independent, oncogenic EGF receptor mutant, are qualitatively distinct from the ligand-dependent mitogenic signaling pathways associated with the wild-type EGF receptor. Specifically, expression of S3-v-ErbB in primary fibroblasts results in anchorage-independent growth, increased invasive potential, and the formation of a transformation-specific phosphoprotein signaling complex, all in a Ras-independent manner. Here we demonstrate the transformation-specific interaction between two components of this complex: the adaptor protein Grb2 and the cytoskeletal regulatory protein caldesmon. This interaction is mediated via both the amino-terminal SH3 and central SH2 domains of Grb2, and the amino-terminal (myosin-binding) domain of caldesmon. Expression of a dominant-negative Grb2 deletion mutant, which lacks the carboxy-terminal SH3 domain, in fibroblasts expressing S3-v-ErbB results in a reduction in phosphoprotein complex formation, the loss of anchorage-independent growth, and a reduction in invasive potential. Together, these results demonstrate a Ras-independent role for Grb2 in modulating cytoskeletal function during ligand-independent EGF receptor-mediated transformation, and provide further support for the hypothesis that ligand-independent oncogenic signaling is qualitatively distinct from ligand-dependent mitogenic signaling by the EGF receptor.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing , Cytoskeleton/metabolism , ErbB Receptors/metabolism , Proteins/metabolism , Actins/chemistry , Agar/chemistry , Animals , Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/metabolism , Cell Line, Transformed , Cell Transformation, Neoplastic , Cells, Cultured , Chick Embryo , Fibroblasts/metabolism , GRB2 Adaptor Protein , Glutathione Transferase/metabolism , Ligands , Microscopy, Fluorescence , Models, Biological , Mutation , Neoplasm Invasiveness , Phosphorylation , Phosphotyrosine/chemistry , Precipitin Tests , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Signal Transduction , Tyrosine/metabolism , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL