Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Eng Ethics ; 28(4): 31, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35796841

ABSTRACT

In the last 20 years, there has been a sharp increase in the incidence of retractions of articles published in scientific journals, the majority of which are due to research misconduct. In some cases, researchers have revised and republished articles that were retracted due to misconduct, which raises some novel questions concerning authorship. Suppose that an article is retracted because one of the authors fabricated or falsified some data, but the researchers decide to salvage the useable data, make appropriate revisions, and resubmit the article for publication. If the person who committed misconduct has made a significant contribution to the research reported in the revised paper, should they be named as an author to recognize this contribution or should they be denied authorship because they committed misconduct? This is a challenging issue because it involves the confluence of two research ethics domains that are usually dealt with separately, i.e., resolution of authorship disputes and adjudication of misconduct findings, as well as potential conflicts among norms that underlie authorship practices and misconduct adjudication. In this paper, we (1) describe some actual cases involving articles that were retracted for misconduct and republished; (2) review policies from the International Committee of Medical Journal Editors, Committee on Publication Ethics, and top fifteen biomedical journals to determine whether they provide adequate guidance for cases like these; and (3) analyze the ethical and policy issues that may arise in these situations.


Subject(s)
Biomedical Research , Scientific Misconduct , Authorship , Ethics, Research , Humans
2.
Methods Mol Biol ; 1999: 307-318, 2019.
Article in English | MEDLINE | ID: mdl-31127587

ABSTRACT

Understanding the molecular dynamics of DNA replication in vivo has been a formidable challenge requiring the development of advanced technologies. Over the past 50 years or so, studies involving DNA autoradiography in bacterial cells have led to sophisticated DNA tract analyses in human cells to characterize replication dynamics at the single-molecule level. Our own lab has used DNA fiber analysis to characterize replication in helicase-deficient human cells. This work led us to propose a model in which the human DNA helicase RECQ1 acts as a governor of the single-stranded DNA binding protein RPA and regulates its bioavailability for DNA synthesis. We have also used the DNA fiber approach to investigate the interactive role of DDX11 helicase with a replication fork protection protein (Timeless) in human cells when they are under pharmacologically induced stress. In this methods chapter, we present a step-by-step protocol for the single-molecule DNA fiber assay. We describe experimental designs to study replication stress and staining patterns from pulse-chase labeling experiments to address the dynamics of replication forks in stressed cells.


Subject(s)
DNA Damage/genetics , DNA Replication/genetics , Single Molecule Imaging/methods , Cell Cycle Proteins/metabolism , DEAD-box RNA Helicases/metabolism , DNA Damage/drug effects , DNA Helicases/metabolism , DNA Replication/drug effects , DNA, Single-Stranded/metabolism , Deoxyuridine/analogs & derivatives , Deoxyuridine/toxicity , HeLa Cells , Humans , Idoxuridine/analogs & derivatives , Idoxuridine/toxicity , Intracellular Signaling Peptides and Proteins/metabolism , RecQ Helicases/metabolism , Replication Protein A/metabolism
3.
EMBO Rep ; 18(11): 2030-2050, 2017 11.
Article in English | MEDLINE | ID: mdl-28887320

ABSTRACT

Cancer-associated p53 missense mutants confer gain of function (GOF) and promote tumorigenesis by regulating crucial signaling pathways. However, the role of GOF mutant p53 in regulating DNA replication, a commonly altered pathway in cancer, is less explored. Here, we show that enhanced Cdc7-dependent replication initiation enables mutant p53 to confer oncogenic phenotypes. We demonstrate that mutant p53 cooperates with the oncogenic transcription factor Myb in vivo and transactivates Cdc7 in cancer cells. Moreover, mutant p53 cells exhibit enhanced levels of Dbf4, promoting the activity of Cdc7/Dbf4 complex. Chromatin enrichment of replication initiation factors and subsequent increase in origin firing confirm increased Cdc7-dependent replication initiation in mutant p53 cells. Further, knockdown of CDC7 significantly abrogates mutant p53-driven cancer phenotypes in vitro and in vivo Importantly, high CDC7 expression significantly correlates with p53 mutational status and predicts poor clinical outcome in lung adenocarcinoma patients. Collectively, this study highlights a novel functional interaction between mutant p53 and the DNA replication pathway in cancer cells. We propose that increased Cdc7-dependent replication initiation is a hallmark of p53 gain-of-function mutations.


Subject(s)
Adenocarcinoma/genetics , Cell Cycle Proteins/genetics , DNA Replication , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression Profiling , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Minichromosome Maintenance Complex Component 2/genetics , Minichromosome Maintenance Complex Component 2/metabolism , Neoplasm Staging , Neoplasm Transplantation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Survival Analysis , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism
4.
Genes (Basel) ; 7(7)2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27376332

ABSTRACT

Mounting evidence indicates that alternate DNA structures, which deviate from normal double helical DNA, form in vivo and influence cellular processes such as replication and transcription. However, our understanding of how the cellular machinery deals with unusual DNA structures such as G-quadruplexes (G4), triplexes, or hairpins is only beginning to emerge. New advances in the field implicate a direct role of the Fanconi Anemia Group J (FANCJ) helicase, which is linked to a hereditary chromosomal instability disorder and important for cancer suppression, in replication past unusual DNA obstacles. This work sets the stage for significant progress in dissecting the molecular mechanisms whereby replication perturbation by abnormal DNA structures leads to genomic instability. In this review, we focus on FANCJ and its role to enable efficient DNA replication when the fork encounters vastly abundant naturally occurring DNA obstacles, which may have implications for targeting rapidly dividing cancer cells.

5.
Methods ; 108: 130-41, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27064001

ABSTRACT

The growing number of DNA helicases implicated in hereditary disorders and cancer indicates that this particular class of enzymes plays key roles in genomic stability and cellular homeostasis. Indeed, a large body of work has provided molecular and cellular evidence that helicases act upon a variety of nucleic acid substrates and interact with numerous proteins to enact their functions in replication, DNA repair, recombination, and transcription. Understanding how helicases operate in unique and overlapping pathways is a great challenge to researchers. In this review, we describe a series of experimental approaches and methodologies to identify and characterize DNA helicase inhibitors which collectively provide an alternative and useful strategy to explore their biological significance in cell-based systems. These procedures were used in the discovery of biologically active compounds that inhibited the DNA unwinding function catalyzed by the human WRN helicase-nuclease defective in the premature aging disorder Werner syndrome. We describe in vitro and in vivo experimental approaches to characterize helicase inhibitors with WRN as the model, anticipating that these approaches may be extrapolated to other DNA helicases, particularly those implicated in DNA repair and/or the replication stress response.


Subject(s)
Biological Assay/methods , DNA Helicases/antagonists & inhibitors , DNA Replication/genetics , Enzyme Inhibitors/isolation & purification , DNA Helicases/chemistry , DNA Repair/genetics , Enzyme Inhibitors/chemistry , Humans , Substrate Specificity
6.
Curr Biol ; 25(21): 2830-2838, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26455304

ABSTRACT

Three (BLM, WRN, and RECQ4) of the five human RecQ helicases are linked to genetic disorders characterized by genomic instability, cancer, and accelerated aging [1]. RECQ1, the first human RecQ helicase discovered [2-4] and the most abundant [5], was recently implicated in breast cancer [6, 7]. RECQ1 is an ATP-dependent DNA-unwinding enzyme (helicase) [8, 9] with roles in replication [10-12] and DNA repair [13-16]. RECQ1 is highly expressed in various tumors and cancer cell lines (for review, see [17]), and its suppression reduces cancer cell proliferation [14], suggesting a target for anti-cancer drugs. RECQ1's assembly state plays a critical role in modulating its helicase, branch migration (BM), or strand annealing [18, 19]. The crystal structure of truncated RECQ1 [20, 21] resembles that of E. coli RecQ [22] with two RecA-like domains, a RecQ-specific zinc-binding domain and a winged-helix domain, the latter implicated in DNA strand separation and oligomer formation. In addition, a conserved aromatic loop (AL) is important for DNA unwinding by bacterial RecQ [23, 24] and truncated RECQ1 helicases [21]. To better understand the roles of RECQ1, two AL mutants (W227A and F231A) in full-length RECQ1 were characterized biochemically and genetically. The RECQ1 mutants were defective in helicase or BM but retained DNA binding, oligomerization, ATPase, and strand annealing. RECQ1-depleted HeLa cells expressing either AL mutant displayed reduced replication tract length, elevated dormant origin firing, and increased double-strand breaks that could be suppressed by exogenously expressed replication protein A (RPA). Thus, RECQ1 governs RPA's availability in order to maintain normal replication dynamics, suppress DNA damage, and preserve genome homeostasis.


Subject(s)
DNA Replication/physiology , RecQ Helicases/genetics , RecQ Helicases/metabolism , Stress, Physiological/physiology , Cell Line, Tumor , Cells, Cultured , DNA/genetics , DNA/metabolism , DNA Damage , DNA Repair , DNA Replication/genetics , Genomic Instability , Humans , Protein Binding , Protein Structure, Tertiary , Replication Protein A/metabolism , Stress, Physiological/genetics , Substrate Specificity
7.
Cell Cycle ; 14(22): 3540-3, 2015.
Article in English | MEDLINE | ID: mdl-26125302

ABSTRACT

Identifying and characterizing novel genetic risk factors for BRCA1/2 negative breast cancers is highly relevant for early diagnosis and development of a management plan. Mutations in a number of DNA repair genes have been associated with genomic instability and development of breast and various other cancers. Whole exome sequencing efforts by 2 groups have led to the discovery in distinct populations of multiple breast cancer susceptibility mutations in RECQL, a gene that encodes a DNA helicase involved in homologous recombination repair and response to replication stress. RECQL pathogenic mutations were identified that truncated or disrupted the RECQL protein or introduced missense mutations in its helicase domain. RECQL mutations may serve as a useful biomarker for breast cancer. Targeting RECQL associated tumors with novel DNA repair inhibitors may provide a new strategy for anti-cancer therapy.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Mutation , RecQ Helicases/genetics , BRCA1 Protein/deficiency , BRCA1 Protein/genetics , BRCA2 Protein/deficiency , BRCA2 Protein/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Exome , Female , Genomic Instability , High-Throughput Nucleotide Sequencing , Humans , Protein Structure, Tertiary , RecQ Helicases/metabolism , Recombinational DNA Repair , Signal Transduction
8.
PLoS One ; 9(11): e113293, 2014.
Article in English | MEDLINE | ID: mdl-25409515

ABSTRACT

8,5' cyclopurine deoxynucleosides (cPu) are locally distorting DNA base lesions corrected by nucleotide excision repair (NER) and proposed to play a role in neurodegeneration prevalent in genetically defined Xeroderma pigmentosum (XP) patients. In the current study, purified recombinant helicases from different classifications based on sequence homology were examined for their ability to unwind partial duplex DNA substrates harboring a single site-specific cPu adduct. Superfamily (SF) 2 RecQ helicases (RECQ1, BLM, WRN, RecQ) were inhibited by cPu in the helicase translocating strand, whereas helicases from SF1 (UvrD) and SF4 (DnaB) tolerated cPu in either strand. SF2 Fe-S helicases (FANCJ, DDX11 (ChlR1), DinG, XPD) displayed marked differences in their ability to unwind the cPu DNA substrates. Archaeal Thermoplasma acidophilum XPD (taXPD), homologue to the human XPD helicase involved in NER DNA damage verification, was impeded by cPu in the non-translocating strand, while FANCJ was uniquely inhibited by the cPu in the translocating strand. Sequestration experiments demonstrated that FANCJ became trapped by the translocating strand cPu whereas RECQ1 was not, suggesting the two SF2 helicases interact with the cPu lesion by distinct mechanisms despite strand-specific inhibition for both. Using a protein trap to simulate single-turnover conditions, the rate of FANCJ or RECQ1 helicase activity was reduced 10-fold and 4.5-fold, respectively, by cPu in the translocating strand. In contrast, single-turnover rates of DNA unwinding by DDX11 and UvrD helicases were only modestly affected by the cPu lesion in the translocating strand. The marked difference in effect of the translocating strand cPu on rate of DNA unwinding between DDX11 and FANCJ helicase suggests the two Fe-S cluster helicases unwind damaged DNA by distinct mechanisms. The apparent complexity of helicase encounters with an unusual form of oxidative damage is likely to have important consequences in the cellular response to DNA damage and DNA repair.


Subject(s)
DNA Helicases/metabolism , Deoxyadenosines/pharmacology , Deoxyguanosine/pharmacology , Archaea/enzymology , Archaeal Proteins/isolation & purification , Archaeal Proteins/metabolism , DNA/chemistry , DNA Damage , DNA Helicases/isolation & purification , DNA Repair , Deoxyadenosines/chemical synthesis , Deoxyguanosine/chemical synthesis , Humans , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
9.
J Biol Chem ; 289(29): 19928-41, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24895130

ABSTRACT

Understanding how cellular machinery deals with chromosomal genome complexity is an important question because protein bound to DNA may affect various cellular processes of nucleic acid metabolism. DNA helicases are at the forefront of such processes, yet there is only limited knowledge how they remodel protein-DNA complexes and how these mechanisms are regulated. We have determined that representative human RecQ and Fe-S cluster DNA helicases are potently blocked by a protein-DNA interaction. The Fanconi anemia group J (FANCJ) helicase partners with the single-stranded DNA-binding protein replication protein A (RPA) to displace BamHI-E111A bound to duplex DNA in a specific manner. Protein displacement was dependent on the ATPase-driven function of the helicase and unique properties of RPA. Further biochemical studies demonstrated that the shelterin proteins TRF1 and TRF2, which preferentially bind the telomeric repeat found at chromosome ends, effectively block FANCJ from unwinding the forked duplex telomeric substrate. RPA, but not the Escherichia coli single-stranded DNA-binding protein or shelterin factor Pot1, stimulated FANCJ ejection of TRF1 from the telomeric DNA substrate. FANCJ was also able to displace TRF2 from the telomeric substrate in an RPA-dependent manner. The stimulation of helicase-catalyzed protein displacement is also observed with the DNA helicase RECQ1, suggesting a conserved functional interaction of RPA-interacting helicases. These findings suggest that partnerships between RPA and interacting human DNA helicases may greatly enhance their ability to dislodge proteins bound to duplex DNA, an activity that is likely to be highly relevant to their biological roles in DNA metabolism.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , DNA/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , RecQ Helicases/metabolism , Replication Protein A/metabolism , Amino Acid Substitution , Base Sequence , DNA/chemistry , DNA/genetics , Deoxyribonuclease BamHI/metabolism , Exodeoxyribonucleases/metabolism , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Nucleic Acid Conformation , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Protein A/genetics , Substrate Specificity , Telomeric Repeat Binding Protein 1/metabolism , Werner Syndrome Helicase
10.
Mol Cell ; 54(4): 613-25, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24793694

ABSTRACT

Upon androgen stimulation, PKN1-mediated histone H3 threonine 11 phosphorylation (H3T11P) promotes AR target gene activation. However, the underlying mechanism is not completely understood. Here, we show that WDR5, a subunit of the SET1/MLL complex, interacts with H3T11P, and this interaction facilitates the recruitment of the MLL1 complex and subsequent H3K4 tri-methylation (H3K4me3). Using ChIP-seq, we find that androgen stimulation results in a 6-fold increase in the number of H3T11P-marked regions and induces WDR5 colocalization to one third of H3T11P-enriched promoters, thus establishing a genome-wide relationship between H3T11P and recruitment of WDR5. Accordingly, PKN1 knockdown or chemical inhibition severely blocks WDR5 chromatin association and H3K4me3 on AR target genes. Finally, WDR5 is critical in prostate cancer cell proliferation and is hyperexpressed in human prostate cancers. Together, these results identify WDR5 as a critical epigenomic integrator of histone phosphorylation and methylation and as a major driver of androgen-dependent prostate cancer cell proliferation.


Subject(s)
Androgens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Prostatic Neoplasms/metabolism , Protein Kinase C/metabolism , Receptors, Androgen/metabolism , Threonine/metabolism , Cell Line, Tumor , Cell Proliferation , Chromatin/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HeLa Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Intracellular Signaling Peptides and Proteins , Male , Methylation , Myeloid-Lymphoid Leukemia Protein/genetics , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase C/genetics , Receptors, Androgen/genetics , Signal Transduction , Threonine/genetics
11.
Cell Mol Life Sci ; 71(14): 2625-39, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24487782

ABSTRACT

In 2010, a new recessive cohesinopathy disorder, designated Warsaw breakage syndrome (WABS), was described. The individual with WABS displayed microcephaly, pre- and postnatal growth retardation, and abnormal skin pigmentation. Cytogenetic analysis revealed mitomycin C (MMC)-induced chromosomal breakage; however, an additional sister chromatid cohesion defect was also observed. WABS is genetically linked to bi-allelic mutations in the ChlR1/DDX11 gene which encodes a protein of the conserved family of Iron-Sulfur (Fe-S) cluster DNA helicases. Mutations in the budding yeast ortholog of ChlR1, known as Chl1, were known to cause sister chromatid cohesion defects, indicating a conserved function of the gene. In 2012, three affected siblings were identified with similar symptoms to the original WABS case, and found to have a homozygous mutation in the conserved Fe-S domain of ChlR1, confirming the genetic linkage. Significantly, the clinically relevant mutations perturbed ChlR1 DNA unwinding activity. In addition to its genetic importance in human disease, ChlR1 is implicated in papillomavirus genome maintenance and cancer. Although its precise functions in genome homeostasis are still not well understood, ongoing molecular studies of ChlR1 suggest the helicase plays a critically important role in cellular replication and/or DNA repair.


Subject(s)
Abnormalities, Multiple/genetics , DEAD-box RNA Helicases/genetics , DNA Helicases/genetics , DEAD-box RNA Helicases/physiology , DNA Breaks , DNA Helicases/physiology , G-Quadruplexes , Genomic Instability , Homeostasis , Humans , Mitomycin , Mutation , Neoplasms/genetics , Papillomaviridae/genetics , Phenotype , Substrate Specificity , Syndrome
12.
Cell Cycle ; 12(20): 3329-35, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24036544

ABSTRACT

Our recently published work suggests that DNA helicases such as the Werner syndrome helicase (WRN) represent a novel class of proteins to target for anticancer therapy. Specifically, pharmacological inhibition of WRN helicase activity in human cells defective in the Fanconi anemia (FA) pathway of interstrand cross-link (ICL) repair are sensitized to the DNA cross-linking agent and chemotherapy drug mitomycin C (MMC) by the WRN helicase inhibitor NSC 617145. (1) The mechanistic basis for the synergistic interaction between NSC 617145 and MMC is discussed in this paper and extrapolated to potential implications for genetic or chemically induced synthetic lethality provoked by cellular exposure to the WRN helicase inhibitor under the context of relevant DNA repair deficiencies associated with cancers or induced by small-molecule inhibitors. Experimental data are presented showing that small-molecule inhibition of WRN helicase elevates sensitivity to MMC-induced stress in human cells that are deficient in both FANCD2 and DNA protein kinase catalytic subunit (DNA-PKcs). These findings suggest a model in which drug-mediated inhibition of WRN helicase activity exacerbates the deleterious effects of MMC-induced DNA damage when both the FA and NHEJ pathways are defective. We conclude with a perspective for the FA pathway and synthetic lethality and implications for DNA repair helicase inhibitors that can be developed for anticancer strategies.


Subject(s)
Enzyme Inhibitors/pharmacology , Fanconi Anemia/enzymology , Neoplasms/enzymology , RecQ Helicases/antagonists & inhibitors , Cell Line , DNA End-Joining Repair/drug effects , Fanconi Anemia/pathology , Humans , Maleimides/pharmacology , Mitomycin/pharmacology , Models, Biological , Neoplasms/pathology , RecQ Helicases/metabolism , Signal Transduction/drug effects , Stress, Physiological/drug effects
13.
Cancer Res ; 73(17): 5497-507, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23867477

ABSTRACT

Werner syndrome is genetically linked to mutations in WRN that encodes a DNA helicase-nuclease believed to operate at stalled replication forks. Using a newly identified small-molecule inhibitor of WRN helicase (NSC 617145), we investigated the role of WRN in the interstrand cross-link (ICL) response in cells derived from patients with Fanconi anemia, a hereditary disorder characterized by bone marrow failure and cancer. In FA-D2(-/-) cells, NSC 617145 acted synergistically with very low concentrations of mitomycin C to inhibit proliferation in a WRN-dependent manner and induce double-strand breaks (DSB) and chromosomal abnormalities. Under these conditions, ataxia-telangiectasia mutated activation and accumulation of DNA-dependent protein kinase, catalytic subunit pS2056 foci suggested an increased number of DSBs processed by nonhomologous end-joining (NHEJ). Rad51 foci were also elevated in FA-D2(-/-) cells exposed to NSC 617145 and mitomycin C, suggesting that WRN helicase inhibition interferes with later steps of homologous recombination at ICL-induced DSBs. Thus, when the Fanconi anemia pathway is defective, WRN helicase inhibition perturbs the normal ICL response, leading to NHEJ activation. Potential implication for treatment of Fanconi anemia-deficient tumors by their sensitization to DNA cross-linking agents is discussed.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Enzyme Inhibitors/pharmacology , Exodeoxyribonucleases/antagonists & inhibitors , Fanconi Anemia/drug therapy , Maleimides/pharmacology , RecQ Helicases/antagonists & inhibitors , Alkylating Agents/pharmacology , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/metabolism , Blotting, Western , Cell Proliferation/drug effects , Chromatin/genetics , Chromosomal Instability , DNA Replication/drug effects , DNA-Activated Protein Kinase/metabolism , Drug Synergism , Drug Therapy, Combination , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/pathology , HCT116 Cells , HeLa Cells , Humans , Mitomycin/pharmacology , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , Rad51 Recombinase/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Werner Syndrome Helicase
16.
Mol Cell Biol ; 31(24): 4858-73, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22006017

ABSTRACT

Although discovered long ago, posttranslational phosphorylation of histones has been in the spotlight only recently. Information is accumulating almost daily on phosphorylation of histones and their roles in cellular physiology and human diseases. An extensive cross talk exists between phosphorylation and other posttranslational modifications, which together regulate various biological processes, including gene transcription, DNA repair, and cell cycle progression. Recent research on histone phosphorylation has demonstrated that nearly all histone types are phosphorylated at specific residues and that these modifications act as a critical intermediate step in chromosome condensation during cell division, transcriptional regulation, and DNA damage repair. As with all young fields, apparently conflicting and sometimes controversial observations about histone phosphorylations and their true functions in different species are found in the literature. Accumulating evidence suggests that instead of functioning strictly as part of a general code, histone phosphorylation probably functions by establishing cross talk with other histone modifications and serving as a platform for recruitment or release of effector proteins, leading to a downstream cascade of events. Here we extensively review published information on the complexities of histone phosphorylation, the roles of proteins recognizing these modifications and the resuting physiological outcome, and, importantly, future challenges and opportunities in this fast-moving field.


Subject(s)
Histones/chemistry , Histones/genetics , Animals , Cell Cycle , Cell Division , Chromatin/chemistry , Chromatin/genetics , DNA Repair , Gene Expression Regulation , Humans , Phosphorylation , Protein Processing, Post-Translational/genetics
17.
J Biol Chem ; 286(18): 15666-77, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21454660

ABSTRACT

The spindle assembly checkpoint (SAC) ensures accurate segregation of chromosomes by monitoring kinetochore attachment of spindles during mitosis. Proper progression of mitosis depends on orderly ubiquitination and subsequent degradation of various mitotic inhibitors. At the molecular level, upon removal of SAC, Cdc20 activates E3 ubiquitin ligase anaphase-promoting complex/cyclosome that, along with E2 ubiquitin-conjugating enzyme UbcH10, executes this function. Both Cdc20 and UbcH10 are overexpressed in many cancer types and are associated with defective SAC function leading to chromosomal instability. The precise mechanism of correlated overexpression of these two proteins remains elusive. We show that Cdc20 transcriptionally up-regulates UbcH10 expression. The WD40 domain of Cdc20 is required for this activity. Physical interaction between Cdc20 and anaphase-promoting complex/cyclosome-CBP/p300 complex and its subsequent recruitment to the UBCH10 promoter are involved in this transactivation process. This transcriptional regulatory function of Cdc20 was observed to be cell cycle-specific. We hypothesize that this co-regulated overexpression of both proteins contributes to chromosomal instability.


Subject(s)
Cell Cycle Proteins/metabolism , Gene Expression Regulation/physiology , Spindle Apparatus/metabolism , Transcription, Genetic/physiology , Ubiquitin-Conjugating Enzymes/biosynthesis , Anaphase-Promoting Complex-Cyclosome , Cdc20 Proteins , Cell Cycle Proteins/genetics , Chromosomal Instability/physiology , Chromosome Segregation/physiology , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , HeLa Cells , Hep G2 Cells , Humans , Mitosis/physiology , Spindle Apparatus/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligase Complexes/metabolism
18.
Nucleic Acids Res ; 37(8): 2688-98, 2009 May.
Article in English | MEDLINE | ID: mdl-19273532

ABSTRACT

CDC20 is a critical molecule in the Spindle Assembly Checkpoint (SAC). It activates the Anaphase promoting complex and helps a dividing cell to proceed towards Anaphase. CDC20 is overexpressed in many tumor cells which cause chromosomal instability. There have been limited reports on the mechanism of SAC's response to genotoxic stress. We show that ectopically expressed p53 or DNA damage induced endogenous p53 can downregulate Cdc20 transcriptionally. We have identified a consensus p53-binding site on the Cdc20 promoter and have shown that it is being used by p53 to bind the promoter and bring about chromatin remodeling thereby repressing Cdc20. Additionally, p53 also downregulates Cdc20 promoter through CDE/CHR element, but in a p21 independent manner. This CDE/CHR element-mediated downregulation occurs only under p53 overexpressed condition but not in the context of DNA damage. The present results suggest that the two CCAAT elements in the Cdc20 promoter are not used by p53 to downregulate its activity, as reported earlier.


Subject(s)
Cell Cycle Proteins/genetics , Chromatin Assembly and Disassembly , DNA Damage , Down-Regulation , Promoter Regions, Genetic , Tumor Suppressor Protein p53/metabolism , Binding Sites , Cdc20 Proteins , Cell Cycle Proteins/metabolism , Cell Line , Humans , Transcription, Genetic , Tumor Suppressor Protein p53/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...