Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cell ; 175(1): 6-9, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30217360

ABSTRACT

This year's Albert Lasker Basic Medical Research Award honors David Allis and Michael Grunstein for their pioneering research that highlighted the importance of histones and their post-translational modifications in the direct control of gene expression.


Subject(s)
Chromatin/physiology , Histones/physiology , Protein Processing, Post-Translational/physiology , Awards and Prizes , Biomedical Research , Gene Expression , Histone Code , Histones/history , History, 21st Century , Humans
2.
Mol Cell ; 83(3): 352-372, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36640769

ABSTRACT

Errors occurring during DNA replication can result in inaccurate replication, incomplete replication, or re-replication, resulting in genome instability that can lead to diseases such as cancer or disorders such as autism. A great deal of progress has been made toward understanding the entire process of DNA replication in eukaryotes, including the mechanism of initiation and its control. This review focuses on the current understanding of how the origin recognition complex (ORC) contributes to determining the location of replication initiation in the multiple chromosomes within eukaryotic cells, as well as methods for mapping the location and temporal patterning of DNA replication. Origin specification and configuration vary substantially between eukaryotic species and in some cases co-evolved with gene-silencing mechanisms. We discuss the possibility that centromeres and origins of DNA replication were originally derived from a common element and later separated during evolution.


Subject(s)
Centromere , DNA Replication , Replication Origin , Centromere/metabolism , Origin Recognition Complex/genetics , Replication Origin/genetics , Saccharomyces cerevisiae/genetics
3.
Mol Cell ; 81(9): 1951-1969.e6, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33761311

ABSTRACT

The initiation of DNA replication involves cell cycle-dependent assembly and disassembly of protein complexes, including the origin recognition complex (ORC) and CDC6 AAA+ ATPases. We report that multiple short linear protein motifs (SLiMs) within intrinsically disordered regions (IDRs) in ORC1 and CDC6 mediate cyclin-CDK-dependent and independent protein-protein interactions, conditional on the cell cycle phase. A domain within the ORC1 IDR is required for interaction between the ORC1 and CDC6 AAA+ domains in G1, whereas the same domain prevents CDC6-ORC1 interaction during mitosis. Then, during late G1, this domain facilitates ORC1 destruction by a SKP2-cyclin A-CDK2-dependent mechanism. During G1, the CDC6 Cy motif cooperates with cyclin E-CDK2 to promote ORC1-CDC6 interactions. The CDC6 IDR regulates self-interaction by ORC1, thereby controlling ORC1 protein levels. Protein phosphatase 1 binds directly to a SLiM in the ORC1 IDR, causing ORC1 de-phosphorylation upon mitotic exit, increasing ORC1 protein, and promoting pre-RC assembly.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , DNA Replication , Intrinsically Disordered Proteins/metabolism , Mitosis , Nuclear Proteins/metabolism , Origin Recognition Complex/metabolism , AAA Domain , ATPases Associated with Diverse Cellular Activities/genetics , Cell Cycle Proteins/genetics , Cell Nucleus/genetics , Cyclin A/genetics , Cyclin A/metabolism , Cyclin E/genetics , Cyclin E/metabolism , G1 Phase , HeLa Cells , Humans , Intrinsically Disordered Proteins/genetics , Nuclear Proteins/genetics , Origin Recognition Complex/genetics , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Stability , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 121(23): e2400667121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38758693

ABSTRACT

In the mid-1950s, Arthur Kornberg elucidated the enzymatic synthesis of DNA by DNA polymerase, for which he was recognized with the 1959 Nobel Prize in Physiology or Medicine. He then identified many of the proteins that cooperate with DNA polymerase to replicate duplex DNA of small bacteriophages. However, one major unanswered problem was understanding the mechanism and control of the initiation of chromosome replication in bacteria. In a seminal paper in 1981, Fuller, Kaguni, and Kornberg reported the development of a cell-free enzyme system that could replicate DNA that was dependent on the bacterial origin of DNA replication, oriC. This advance opened the door to a flurry of discoveries and important papers that elucidated the process and control of initiation of chromosome replication in bacteria.


Subject(s)
Chromosomes, Bacterial , DNA Replication , Chromosomes, Bacterial/genetics , Chromosomes, Bacterial/metabolism , History, 20th Century , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/genetics , Bacteria/genetics , Bacteria/metabolism , DNA, Bacterial/metabolism , DNA, Bacterial/genetics
5.
Cell ; 147(7): 1525-36, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22177093

ABSTRACT

Although replication-coupled chromatin assembly is known to be important for the maintenance of patterns of gene expression through sequential cell divisions, the role of replication-coupled chromatin assembly in controlling cell differentiation during animal development remains largely unexplored. Here we report that the CAF-1 protein complex, an evolutionarily conserved histone chaperone that deposits histone H3-H4 proteins onto replicating DNA, is required to generate a bilateral asymmetry in the C. elegans nervous system. A mutation in 1 of 24 C. elegans histone H3 genes specifically eliminates this aspect of neuronal asymmetry by causing a defect in the formation of a histone H3-H4 tetramer and the consequent inhibition of CAF-1-mediated nucleosome formation. Our results reveal that replication-coupled nucleosome assembly is necessary to generate a bilateral asymmetry in C. elegans neuroanatomy and suggest that left-right asymmetric epigenetic regulation can establish bilateral asymmetry in the nervous system.


Subject(s)
Caenorhabditis elegans/embryology , Caenorhabditis elegans/metabolism , Chromatin Assembly and Disassembly , DNA Replication , Epigenomics , Amino Acid Sequence , Animals , Body Patterning , Caenorhabditis elegans Proteins/metabolism , Histones/chemistry , Histones/metabolism , Molecular Sequence Data , Nervous System/embryology , Neurons/metabolism , Nucleosomes/metabolism , Sequence Alignment
6.
Mol Cell ; 59(2): 139-41, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26186286

ABSTRACT

The distribution of DNA polymerase activities at the eukaryotic DNA replication fork was "established," but recent genetic studies in this issue of Molecular Cell raise questions about which polymerases are copying the leading and lagging strand templates (Johnson et al, 2015).


Subject(s)
DNA Polymerase III/metabolism , DNA Replication , DNA, Fungal/metabolism , Saccharomyces cerevisiae Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 117(30): 17747-17756, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32669428

ABSTRACT

DNA replication origins serve as sites of replicative helicase loading. In all eukaryotes, the six-subunit origin recognition complex (Orc1-6; ORC) recognizes the replication origin. During late M-phase of the cell-cycle, Cdc6 binds to ORC and the ORC-Cdc6 complex loads in a multistep reaction and, with the help of Cdt1, the core Mcm2-7 helicase onto DNA. A key intermediate is the ORC-Cdc6-Cdt1-Mcm2-7 (OCCM) complex in which DNA has been already inserted into the central channel of Mcm2-7. Until now, it has been unclear how the origin DNA is guided by ORC-Cdc6 and inserted into the Mcm2-7 hexamer. Here, we truncated the C-terminal winged-helix-domain (WHD) of Mcm6 to slow down the loading reaction, thereby capturing two loading intermediates prior to DNA insertion in budding yeast. In "semi-attached OCCM," the Mcm3 and Mcm7 WHDs latch onto ORC-Cdc6 while the main body of the Mcm2-7 hexamer is not connected. In "pre-insertion OCCM," the main body of Mcm2-7 docks onto ORC-Cdc6, and the origin DNA is bent and positioned adjacent to the open DNA entry gate, poised for insertion, at the Mcm2-Mcm5 interface. We used molecular simulations to reveal the dynamic transition from preloading conformers to the loaded conformers in which the loading of Mcm2-7 on DNA is complete and the DNA entry gate is fully closed. Our work provides multiple molecular insights into a key event of eukaryotic DNA replication.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Replication , Replication Origin , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Binding Sites , Cryoelectron Microscopy , Minichromosome Maintenance Complex Component 6/chemistry , Minichromosome Maintenance Complex Component 6/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Origin Recognition Complex , Protein Binding , Protein Conformation , Structure-Activity Relationship
8.
Genes Dev ; 28(20): 2291-303, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25319829

ABSTRACT

Eukaryotic cells license each DNA replication origin during G1 phase by assembling a prereplication complex that contains a Mcm2-7 (minichromosome maintenance proteins 2-7) double hexamer. During S phase, each Mcm2-7 hexamer forms the core of a replicative DNA helicase. However, the mechanisms of origin licensing and helicase activation are poorly understood. The helicase loaders ORC-Cdc6 function to recruit a single Cdt1-Mcm2-7 heptamer to replication origins prior to Cdt1 release and ORC-Cdc6-Mcm2-7 complex formation, but how the second Mcm2-7 hexamer is recruited to promote double-hexamer formation is not well understood. Here, structural evidence for intermediates consisting of an ORC-Cdc6-Mcm2-7 complex and an ORC-Cdc6-Mcm2-7-Mcm2-7 complex are reported, which together provide new insights into DNA licensing. Detailed structural analysis of the loaded Mcm2-7 double-hexamer complex demonstrates that the two hexamers are interlocked and misaligned along the DNA axis and lack ATP hydrolysis activity that is essential for DNA helicase activity. Moreover, we show that the head-to-head juxtaposition of the Mcm2-7 double hexamer generates a new protein interaction surface that creates a multisubunit-binding site for an S-phase protein kinase that is known to activate DNA replication. The data suggest how the double hexamer is assembled and how helicase activity is regulated during DNA licensing, with implications for cell cycle control of DNA replication and genome stability.


Subject(s)
Minichromosome Maintenance Proteins/chemistry , Minichromosome Maintenance Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Adenosine Triphosphate/metabolism , Binding Sites , Enzyme Activation , Hydrolysis , Microscopy, Electron , Minichromosome Maintenance Proteins/isolation & purification , Molecular Conformation , Protein Binding
9.
Proc Natl Acad Sci U S A ; 114(45): E9529-E9538, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078375

ABSTRACT

During replication initiation, the core component of the helicase-the Mcm2-7 hexamer-is loaded on origin DNA as a double hexamer (DH). The two ring-shaped hexamers are staggered, leading to a kinked axial channel. How the origin DNA interacts with the axial channel is not understood, but the interaction could provide key insights into Mcm2-7 function and regulation. Here, we report the cryo-EM structure of the Mcm2-7 DH on dsDNA and show that the DNA is zigzagged inside the central channel. Several of the Mcm subunit DNA-binding loops, such as the oligosaccharide-oligonucleotide loops, helix 2 insertion loops, and presensor 1 (PS1) loops, are well defined, and many of them interact extensively with the DNA. The PS1 loops of Mcm 3, 4, 6, and 7, but not 2 and 5, engage the lagging strand with an approximate step size of one base per subunit. Staggered coupling of the two opposing hexamers positions the DNA right in front of the two Mcm2-Mcm5 gates, with each strand being pressed against one gate. The architecture suggests that lagging-strand extrusion initiates in the middle of the DH that is composed of the zinc finger domains of both hexamers. To convert the Mcm2-7 DH structure into the Mcm2-7 hexamer structure found in the active helicase, the N-tier ring of the Mcm2-7 hexamer in the DH-dsDNA needs to tilt and shift laterally. We suggest that these N-tier ring movements cause the DNA strand separation and lagging-strand extrusion.


Subject(s)
DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , DNA/chemistry , Minichromosome Maintenance Proteins/chemistry , DNA Replication/genetics , Oligosaccharides/chemistry , Protein Domains/genetics , Zinc Fingers/genetics
10.
Genes Dev ; 26(16): 1797-810, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22855792

ABSTRACT

Like DNA replication, centrosomes are licensed to duplicate once per cell division cycle to ensure genetic stability. In addition to regulating DNA replication, the Orc1 subunit of the human origin recognition complex controls centriole and centrosome copy number. Here we report that Orc1 harbors a PACT centrosome-targeting domain and a separate domain that differentially inhibits the protein kinase activities of Cyclin E-CDK2 and Cyclin A-CDK2. A cyclin-binding motif (Cy motif) is required for Orc1 to bind Cyclin A and inhibit Cyclin A-CDK2 kinase activity but has no effect on Cyclin E-CDK2 kinase activity. In contrast, Orc1 inhibition of Cyclin E-CDK2 kinase activity occurs by a different mechanism that is affected by Orc1 mutations identified in Meier-Gorlin syndrome patients. The cyclin/CDK2 kinase inhibitory domain of Orc1, when tethered to the PACT domain, localizes to centrosomes and blocks centrosome reduplication. Meier-Gorlin syndrome mutations that disrupt Cyclin E-CDK2 kinase inhibition also allow centrosome reduplication. Thus, Orc1 contains distinct domains that control centrosome copy number and DNA replication. We suggest that the Orc1 mutations present in some Meier-Gorlin syndrome patients contribute to the pronounced microcephaly and dwarfism observed in these individuals by altering centrosome duplication in addition to DNA replication defects.


Subject(s)
Centrosome/pathology , Growth Disorders/genetics , Micrognathism/genetics , Origin Recognition Complex/genetics , Amino Acid Sequence , Cell Line, Tumor , Centrioles/metabolism , Congenital Microtia , Cyclin-Dependent Kinases/metabolism , Ear/abnormalities , Growth Disorders/enzymology , HEK293 Cells , Humans , Micrognathism/enzymology , Molecular Sequence Data , Mutation , Patella/abnormalities , Patella/enzymology , Protein Structure, Tertiary , RNA-Binding Proteins/metabolism , Sequence Alignment
11.
Genome Res ; 26(3): 315-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26733669

ABSTRACT

Eukaryotic chromosomes initiate DNA synthesis from multiple replication origins in a temporally specific manner during S phase. The replicative helicase Mcm2-7 functions in both initiation and fork progression and thus is an important target of regulation. Mcm4, a helicase subunit, possesses an unstructured regulatory domain that mediates control from multiple kinase signaling pathways, including the Dbf4-dependent Cdc7 kinase (DDK). Following replication stress in S phase, Dbf4 and Sld3, an initiation factor and essential target of Cyclin-Dependent Kinase (CDK), are targets of the checkpoint kinase Rad53 for inhibition of initiation from origins that have yet to be activated, so-called late origins. Here, whole-genome DNA replication profile analysis is used to access under various conditions the effect of mutations that alter the Mcm4 regulatory domain and the Rad53 targets, Sld3 and Dbf4. Late origin firing occurs under genotoxic stress when the controls on Mcm4, Sld3, and Dbf4 are simultaneously eliminated. The regulatory domain of Mcm4 plays an important role in the timing of late origin firing, both in an unperturbed S phase and in dNTP limitation. Furthermore, checkpoint control of Sld3 impacts fork progression under replication stress. This effect is parallel to the role of the Mcm4 regulatory domain in monitoring fork progression. Hypomorph mutations in sld3 are suppressed by a mcm4 regulatory domain mutation. Thus, in response to cellular conditions, the functions executed by Sld3, Dbf4, and the regulatory domain of Mcm4 intersect to control origin firing and replication fork progression, thereby ensuring genome stability.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Minichromosome Maintenance Complex Component 4/metabolism , Replication Origin , Saccharomyces cerevisiae Proteins/metabolism , Alkylating Agents/pharmacology , Alleles , Checkpoint Kinase 2/metabolism , Chromosomes, Fungal , Cyclin-Dependent Kinases/metabolism , DNA Replication/drug effects , Hydroxyurea/pharmacology , Minichromosome Maintenance Complex Component 4/genetics , Mutation , Phenotype , Phosphorylation , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Sequence Deletion , Signal Transduction
12.
Mol Cell ; 42(1): 127-36, 2011 Apr 08.
Article in English | MEDLINE | ID: mdl-21474074

ABSTRACT

Telomere-associated position-effect variegation (TPEV) in budding yeast has been used as a model for understanding epigenetic inheritance and gene silencing. A widely used assay to identify mutants with improper TPEV employs the URA3 gene at the telomere of chromosome VII-L that can be counterselected with 5-fluoroorotic acid (5-FOA). 5-FOA resistance has been inferred to represent lack of transcription of URA3 and therefore to represent heterochromatin-induced gene silencing. For two genes implicated in telomere silencing, POL30 and DOT1, we show that the URA3 telomere reporter assay does not reflect their role in heterochromatin formation. Rather, an imbalance in ribonucleotide reductase (RNR), which is induced by 5-FOA, and the specific promoter of URA3 fused to ADH4 at telomere VII-L are jointly responsible for the variegated phenotype. We conclude that metabolic changes caused by the drug employed and certain mutants being studied are incompatible with the use of certain prototrophic markers for TPEV.


Subject(s)
Gene Silencing , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere/genetics , Telomere/metabolism , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Chromosomal Position Effects , Genes, Fungal , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Models, Genetic , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Orotic Acid/analogs & derivatives , Orotic Acid/metabolism , Proliferating Cell Nuclear Antigen , Ribonucleotide Reductases/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
13.
Genes Dev ; 25(13): 1359-64, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21724829

ABSTRACT

Histone H3 variant H3.3, while differing from canonical H3 (H3.1) by only five amino acids, is assembled into nucleosomes, along with histone H4, at genic regions by the histone chaperone HIRA, whereas H3.1 is assembled into nucleosomes in a CAF-1-dependent reaction. Here, we show that phosphorylation of histone H4 Ser 47 (H4S47ph), catalyzed by the PAK2 kinase, promotes nucleosome assembly of H3.3-H4 and inhibits nucleosome assembly of H3.1-H4 by increasing the binding affinity of HIRA to H3.3-H4 and reducing association of CAF-1 with H3.1-H4. These results reveal a mechanism whereby H4S47ph distinctly regulates nucleosome assembly of H3.1 and H3.3.


Subject(s)
Cell Cycle Proteins/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Nucleosomes/metabolism , Serine/metabolism , Transcription Factors/metabolism , Chromatin/metabolism , HEK293 Cells , HeLa Cells , Humans , Molecular Chaperones , Phosphorylation , Protein Binding , p21-Activated Kinases
14.
Genes Dev ; 24(11): 1133-44, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20516198

ABSTRACT

Break-induced replication (BIR) is an efficient homologous recombination (HR) pathway employed to repair a DNA double-strand break (DSB) when homology is restricted to one end. All three major replicative DNA polymerases are required for BIR, including the otherwise nonessential Pol32 subunit. Here we show that BIR requires the replicative DNA helicase (Cdc45, the GINS, and Mcm2-7 proteins) as well as Cdt1. In contrast, both subunits of origin recognition complex (ORC) and Cdc6, which are required to create a prereplication complex (pre-RC), are dispensable. The Cdc7 kinase, required for both initiation of DNA replication and post-replication repair (PRR), is also required for BIR. Ubiquitination and sumoylation of the DNA processivity clamp PCNA play modest roles; in contrast, PCNA alleles that suppress pol32Delta's cold sensitivity fail to suppress its role in BIR, and are by themselves dominant inhibitors of BIR. These results suggest that origin-independent BIR involves cross-talk between normal DNA replication factors and PRR.


Subject(s)
DNA Breaks, Double-Stranded , DNA Replication/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Origin Recognition Complex , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Alleles , Cell Cycle Proteins/metabolism , Cold Temperature , DNA Helicases/metabolism , DNA Repair/genetics , DNA Repair/physiology , DNA Replication/genetics , Mutation , Origin Recognition Complex/genetics , Protein Serine-Threonine Kinases/metabolism , SUMO-1 Protein/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitination
15.
Proc Natl Acad Sci U S A ; 111(18): E1899-908, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24740181

ABSTRACT

Eukaryotic DNA synthesis initiates from multiple replication origins and progresses through bidirectional replication forks to ensure efficient duplication of the genome. Temporal control of initiation from origins and regulation of replication fork functions are important aspects for maintaining genome stability. Multiple kinase-signaling pathways are involved in these processes. The Dbf4-dependent Cdc7 kinase (DDK), cyclin-dependent kinase (CDK), and Mec1, the yeast Ataxia telangiectasia mutated/Ataxia telangiectasia mutated Rad3-related checkpoint regulator, all target the structurally disordered N-terminal serine/threonine-rich domain (NSD) of mini-chromosome maintenance subunit 4 (Mcm4), a subunit of the mini-chromosome maintenance (MCM) replicative helicase complex. Using whole-genome replication profile analysis and single-molecule DNA fiber analysis, we show that under replication stress the temporal pattern of origin activation and DNA replication fork progression are altered in cells with mutations within two separate segments of the Mcm4 NSD. The proximal segment of the NSD residing next to the DDK-docking domain mediates repression of late-origin firing by checkpoint signals because in its absence late origins become active despite an elevated DNA damage-checkpoint response. In contrast, the distal segment of the NSD at the N terminus plays no role in the temporal pattern of origin firing but has a strong influence on replication fork progression and on checkpoint signaling. Both fork progression and checkpoint response are regulated by the phosphorylation of the canonical CDK sites at the distal NSD. Together, our data suggest that the eukaryotic MCM helicase contains an intrinsic regulatory domain that integrates multiple signals to coordinate origin activation and replication fork progression under stress conditions.


Subject(s)
DNA Replication/physiology , DNA, Fungal/biosynthesis , DNA, Fungal/chemistry , Minichromosome Maintenance Complex Component 4/chemistry , Minichromosome Maintenance Complex Component 4/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Genome, Fungal , Intracellular Signaling Peptides and Proteins/metabolism , Minichromosome Maintenance Complex Component 4/genetics , Mutation , Nucleic Acid Conformation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein Subunits , Replication Origin , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
16.
J Biol Chem ; 290(19): 12355-69, 2015 May 08.
Article in English | MEDLINE | ID: mdl-25784553

ABSTRACT

Replication of eukaryotic chromosomes occurs once every cell division cycle in normal cells and is a tightly controlled process that ensures complete genome duplication. The origin recognition complex (ORC) plays a key role during the initiation of DNA replication. In human cells, the level of Orc1, the largest subunit of ORC, is regulated during the cell division cycle, and thus ORC is a dynamic complex. Upon S phase entry, Orc1 is ubiquitinated and targeted for destruction, with subsequent dissociation of ORC from chromosomes. Time lapse and live cell images of human cells expressing fluorescently tagged Orc1 show that Orc1 re-localizes to condensing chromatin during early mitosis and then displays different nuclear localization patterns at different times during G1 phase, remaining associated with late replicating regions of the genome in late G1 phase. The initial binding of Orc1 to mitotic chromosomes requires C-terminal amino acid sequences that are similar to mitotic chromosome-binding sequences in the transcriptional pioneer protein FOXA1. Depletion of Orc1 causes concomitant loss of the mini-chromosome maintenance (Mcm2-7) helicase proteins on chromatin. The data suggest that Orc1 acts as a nucleating center for ORC assembly and then pre-replication complex assembly by binding to mitotic chromosomes, followed by gradual removal from chromatin during the G1 phase.


Subject(s)
Mitosis , Origin Recognition Complex/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Cell Division , Cell Line, Tumor , Cell Nucleus/metabolism , Chromosomes/ultrastructure , DNA Replication , Epitopes/chemistry , Escherichia coli/metabolism , Fluorescent Dyes/chemistry , G1 Phase/genetics , Genome, Human , HeLa Cells , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Molecular Sequence Data , Origin Recognition Complex/genetics , Protein Structure, Tertiary , RNA Interference , Sequence Homology, Amino Acid
17.
Nature ; 463(7277): 113-7, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-20054399

ABSTRACT

Eukaryotic DNA replication uses kinase regulatory pathways to facilitate coordination with other processes during cell division cycles and response to environmental cues. At least two cell cycle-regulated protein kinase systems, the S-phase-specific cyclin-dependent protein kinases (S-CDKs) and the Dbf4-Cdc7 kinase (DDK, Dbf4-dependent protein kinase) are essential activators for initiation of DNA replication. Although the essential mechanism of CDK activation of DNA replication in Saccharomyces cerevisiae has been established, exactly how DDK acts has been unclear. Here we show that the amino terminal serine/threonine-rich domain (NSD) of Mcm4 has both inhibitory and facilitating roles in DNA replication control and that the sole essential function of DDK is to relieve an inhibitory activity residing within the NSD. By combining an mcm4 mutant lacking the inhibitory activity with mutations that bypass the requirement for CDKs for initiation of DNA replication, we show that DNA synthesis can occur in G1 phase when CDKs and DDK are limited. However, DDK is still required for efficient S phase progression. In the absence of DDK, CDK phosphorylation at the distal part of the Mcm4 NSD becomes crucial. Moreover, DDK-null cells fail to activate the intra-S-phase checkpoint in the presence of hydroxyurea-induced DNA damage and are unable to survive this challenge. Our studies establish that the eukaryote-specific NSD of Mcm4 has evolved to integrate several protein kinase regulatory signals for progression through S phase.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , S Phase/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , DNA Damage , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , G1 Phase/drug effects , Genes, Essential , Hydroxyurea/pharmacology , Microbial Viability/drug effects , Minichromosome Maintenance Complex Component 4 , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , S Phase/drug effects , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Deletion , Substrate Specificity
18.
Mol Cell ; 30(3): 259-60, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18471969

ABSTRACT

The Kunkel laboratory has recently assigned polymerase (Pol) epsilon as the leading strand polymerase. In a recent issue of Molecular Cell, they now assign Pol delta as the lagging strand polymerase.


Subject(s)
DNA Polymerase III/metabolism , DNA Polymerase II/metabolism , DNA Replication , Eukaryotic Cells/physiology , Nucleic Acid Conformation , DNA Polymerase I/metabolism , Genes, Reporter , Simian virus 40/genetics
19.
Cancer Cell ; 11(5): 431-45, 2007 May.
Article in English | MEDLINE | ID: mdl-17482133

ABSTRACT

Systemic administration of chemotherapeutic agents results in indiscriminate drug distribution and severe toxicity. Here we report a technology potentially overcoming these shortcomings through encapsulation and cancer cell-specific targeting of chemotherapeutics in bacterially derived 400 nm minicells. We discovered that minicells can be packaged with therapeutically significant concentrations of chemotherapeutics of differing charge, hydrophobicity, and solubility. Targeting of minicells via bispecific antibodies to receptors on cancer cell membranes results in endocytosis, intracellular degradation, and drug release. This affects highly significant tumor growth inhibition and regression in mouse xenografts and case studies of lymphoma in dogs despite administration of minute amounts of drug and antibody; a factor critical for limiting systemic toxicity that should allow the use of complex regimens of combination chemotherapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Bacteria , Drug Delivery Systems , Animals , Antibodies/administration & dosage , Cell Line, Tumor , Dogs , Drug Compounding , Humans , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Swine
20.
Proc Natl Acad Sci U S A ; 108(17): 7113-8, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21482754

ABSTRACT

RNAi has revolutionized loss-of-function genetics by enabling sequence-specific suppression of virtually any gene. Furthermore, tetracycline response elements (TRE) can drive expression of short hairpin RNAs (shRNAs) for inducible and reversible target gene suppression. Here, we demonstrate the feasibility of transgenic inducible RNAi for suppression of essential genes. We set out to directly target cell proliferation by screening an RNAi library against DNA replication factors and identified multiple shRNAs against Replication Protein A, subunit 3 (RPA3). We generated transgenic mice with TRE-driven Rpa3 shRNAs whose expression enforced a reversible cell cycle arrest. In adult mice, the block in cell proliferation caused rapid atrophy of the intestinal epithelium which led to weight loss and lethality within 8-11 d of shRNA induction. Upon shRNA withdrawal, villus atrophy and weight loss were fully reversible. Thus, shRpa3 transgenic mice provide an interesting tool to study tissue maintenance and regeneration. Overall, we have established a robust system that serves the purpose of temperature-sensitive alleles in other model organisms, enabling inducible and reversible suppression of essential genes in a mammalian system.


Subject(s)
Alleles , Cell Cycle/physiology , DNA Replication/physiology , RNA Interference , Response Elements/physiology , Animals , Female , Male , Mice , Mice, Transgenic , Replication Protein A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL