Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta ; 1822(11): 1727-34, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22885111

ABSTRACT

The RecQ family of helicases has been shown to play an important role in maintaining genomic stability. In humans, this family has five members and mutations in three of these helicases, BLM, WRN and RECQL4, are associated with disease. Alterations in RECQL4 are associated with three diseases, Rothmund-Thomson syndrome, Baller-Gerold syndrome, and RAPADILINO syndrome. One of the more common mutations found in RECQL4 is the RAPADILINO mutation, c.1390+2delT which is a splice-site mutation leading to an in-frame skipping of exon 7 resulting in 44 amino acids being deleted from the protein (p.Ala420-Ala463del). In order to characterize the RAPADILINO RECQL4 mutant protein, it was expressed in bacteria and purified using an established protocol. Strand annealing, helicase, and ATPase assays were conducted to characterize the protein's activities relative to WT RECQL4. Here we show that strand annealing activity in the absence of ATP is unchanged from that of WT RECQL4. However, the RAPADILINO protein variant lacks helicase and ssDNA-stimulated ATPase activity. These observations help explain the underlying molecular etiology of the disease and our findings provide insight into the genotype and phenotype association among RECQL4 syndromes.


Subject(s)
Dwarfism , Heart Septal Defects, Atrial , Limb Deformities, Congenital , Mutation/genetics , RNA Splice Sites/genetics , RecQ Helicases/genetics , Rothmund-Thomson Syndrome , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Anal Canal/abnormalities , Anal Canal/metabolism , Craniosynostoses/genetics , Dwarfism/etiology , Dwarfism/genetics , Dwarfism/metabolism , Exons , Genetic Association Studies , Genomic Instability , Heart Septal Defects, Atrial/etiology , Heart Septal Defects, Atrial/genetics , Heart Septal Defects, Atrial/metabolism , Humans , Limb Deformities, Congenital/etiology , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/metabolism , Patella/abnormalities , Patella/metabolism , Radius/abnormalities , Radius/metabolism , RecQ Helicases/metabolism , Rothmund-Thomson Syndrome/etiology , Rothmund-Thomson Syndrome/genetics , Rothmund-Thomson Syndrome/metabolism
2.
PLoS One ; 7(5): e36960, 2012.
Article in English | MEDLINE | ID: mdl-22615856

ABSTRACT

XPB, also known as ERCC3 and RAD25, is a 3' → 5' DNA repair helicase belonging to the superfamily 2 of helicases. XPB is an essential core subunit of the eukaryotic basal transcription factor complex TFIIH. It has two well-established functions: in the context of damaged DNA, XPB facilitates nucleotide excision repair by unwinding double stranded DNA (dsDNA) surrounding a DNA lesion; while in the context of actively transcribing genes, XPB facilitates initiation of RNA polymerase II transcription at gene promoters. Human and other eukaryotic XPB homologs are relatively well characterized compared to conserved homologs found in mycobacteria and archaea. However, more insight into the function of bacterial helicases is central to understanding the mechanism of DNA metabolism and pathogenesis in general. Here, we characterized Mycobacterium tuberculosis XPB (Mtb XPB), a 3'→5' DNA helicase with DNA-dependent ATPase activity. Mtb XPB efficiently catalyzed DNA unwinding in the presence of significant excess of enzyme. The unwinding activity was fueled by ATP or dATP in the presence of Mg(2+)/Mn(2+). Consistent with the 3'→5' polarity of this bacterial XPB helicase, the enzyme required a DNA substrate with a 3' overhang of 15 nucleotides or more. Although Mtb XPB efficiently unwound DNA model substrates with a 3' DNA tail, it was not active on substrates containing a 3' RNA tail. We also found that Mtb XPB efficiently catalyzed ATP-independent annealing of complementary DNA strands. These observations significantly enhance our understanding of the biological roles of Mtb XPB.


Subject(s)
DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mycobacterium tuberculosis/genetics , Substrate Specificity/genetics , Adenosine Triphosphatases/metabolism , DNA Damage/genetics , DNA Repair , DNA Replication/genetics , DNA, Bacterial/genetics , Mycobacterium tuberculosis/metabolism , Protein Structure, Tertiary/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism , Transcription, Genetic/genetics
3.
Nucleic Acids Res ; 40(14): 6632-48, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22544709

ABSTRACT

Bacteria and yeast possess one RecQ helicase homolog whereas humans contain five RecQ helicases, all of which are important in preserving genome stability. Three of these, BLM, WRN and RECQL4, are mutated in human diseases manifesting in premature aging and cancer. We are interested in determining to which extent these RecQ helicases function cooperatively. Here, we report a novel physical and functional interaction between BLM and RECQL4. Both BLM and RECQL4 interact in vivo and in vitro. We have mapped the BLM interacting site to the N-terminus of RECQL4, comprising amino acids 361-478, and the region of BLM encompassing amino acids 1-902 interacts with RECQL4. RECQL4 specifically stimulates BLM helicase activity on DNA fork substrates in vitro. The in vivo interaction between RECQL4 and BLM is enhanced during the S-phase of the cell cycle, and after treatment with ionizing radiation. The retention of RECQL4 at DNA double-strand breaks is shortened in BLM-deficient cells. Further, depletion of RECQL4 in BLM-deficient cells leads to reduced proliferative capacity and an increased frequency of sister chromatid exchanges. Together, our results suggest that BLM and RECQL4 have coordinated activities that promote genome stability.


Subject(s)
Genomic Instability , RecQ Helicases/metabolism , Cell Line , DNA/metabolism , DNA Damage , Guanine/analogs & derivatives , Guanine/metabolism , HeLa Cells , Humans , Protein Interaction Domains and Motifs , RecQ Helicases/chemistry , S Phase , Sister Chromatid Exchange , Thymine/analogs & derivatives , Thymine/metabolism
4.
Aging Cell ; 11(3): 456-66, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22296597

ABSTRACT

RECQL4 is associated with Rothmund-Thomson Syndrome (RTS), a rare autosomal recessive disorder characterized by premature aging, genomic instability, and cancer predisposition. RECQL4 is a member of the RecQ helicase family, and has many similarities to WRN protein, which is also implicated in premature aging. There is no information about whether any of the RecQ helicases play roles in mitochondrial biogenesis, which is strongly implicated in the aging process. Here, we used microscopy to visualize RECQL4 in mitochondria. Fractionation of human and mouse cells also showed that RECQL4 was present in mitochondria. Q-PCR amplification of mitochondrial DNA demonstrated that mtDNA damage accumulated in RECQL4-deficient cells. Microarray analysis suggested that mitochondrial bioenergetic pathways might be affected in RTS. Measurements of mitochondrial bioenergetics showed a reduction in the mitochondrial reserve capacity after lentiviral knockdown of RECQL4 in two different primary cell lines. Additionally, biochemical assays with RECQL4, mitochondrial transcription factor A, and mitochondrial DNA polymerase γ showed that the polymerase inhibited RECQL4's helicase activity. RECQL4 is the first 3'-5' RecQ helicase to be found in both human and mouse mitochondria, and the loss of RECQL4 alters mitochondrial integrity.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Mitochondria/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Age Factors , Aged, 80 and over , Animals , Cell Fractionation/methods , Cell Line, Tumor , DNA Damage , Genomic Instability , HeLa Cells , Humans , Mice
5.
J Biol Chem ; 287(1): 196-209, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-22039056

ABSTRACT

Telomeres are structures at the ends of chromosomes and are composed of long tracks of short tandem repeat DNA sequences bound by a unique set of proteins (shelterin). Telomeric DNA is believed to form G-quadruplex and D-loop structures, which presents a challenge to the DNA replication and repair machinery. Although the RecQ helicases WRN and BLM are implicated in the resolution of telomeric secondary structures, very little is known about RECQL4, the RecQ helicase mutated in Rothmund-Thomson syndrome (RTS). Here, we report that RTS patient cells have elevated levels of fragile telomeric ends and that RECQL4-depleted human cells accumulate fragile sites, sister chromosome exchanges, and double strand breaks at telomeric sites. Further, RECQL4 localizes to telomeres and associates with shelterin proteins TRF1 and TRF2. Using recombinant proteins we showed that RECQL4 resolves telomeric D-loop structures with the help of shelterin proteins TRF1, TRF2, and POT1. We also found a novel functional synergistic interaction of this protein with WRN during D-loop unwinding. These data implicate RECQL4 in telomere maintenance.


Subject(s)
Mutant Proteins/metabolism , Mutation , RecQ Helicases/metabolism , Rothmund-Thomson Syndrome/genetics , Telomere/metabolism , Aphidicolin/pharmacology , Base Sequence , DNA/biosynthesis , DNA/chemistry , DNA/metabolism , DNA Replication/drug effects , Exodeoxyribonucleases/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Mutant Proteins/genetics , Nucleic Acid Conformation/drug effects , Protein Transport/drug effects , RNA, Small Interfering/genetics , RecQ Helicases/deficiency , RecQ Helicases/genetics , Rothmund-Thomson Syndrome/metabolism , Rothmund-Thomson Syndrome/pathology , Telomere/drug effects , Telomere/genetics , Telomeric Repeat Binding Protein 2/metabolism , Tumor Suppressor p53-Binding Protein 1 , Werner Syndrome Helicase
6.
DNA Repair (Amst) ; 9(7): 796-804, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20451470

ABSTRACT

Humans have five members of the well conserved RecQ helicase family: RecQ1, Bloom syndrome protein (BLM), Werner syndrome protein (WRN), RecQ4, and RecQ5, which are all known for their roles in maintaining genome stability. BLM, WRN, and RecQ4 are associated with premature aging and cancer predisposition. Of the three, RecQ4's biological and cellular roles have been least thoroughly characterized. Here we tested the helicase activity of purified human RecQ4 on various substrates. Consistent with recent results, we detected ATP-dependent RecQ4 unwinding of forked duplexes. However, our results provide the first evidence that human RecQ4's unwinding is independent of strand annealing, and that it does not require the presence of excess ssDNA. Moreover, we demonstrate that a point mutation of the conserved lysine in the Walker A motif abolished helicase activity, implying that not the N-terminal portion, but the helicase domain is solely responsible for the enzyme's unwinding activity. In addition, we demonstrate a novel stimulation of RecQ4's helicase activity by replication protein A, similar to that of RecQ1, BLM, WRN, and RecQ5. Together, these data indicate that specific biochemical activities and protein partners of RecQ4 are conserved with those of the other RecQ helicases.


Subject(s)
DNA Repair , RecQ Helicases/metabolism , Amino Acid Motifs/genetics , Cell Nucleus/enzymology , Conserved Sequence , DNA/chemistry , DNA/genetics , Humans , Lysine/genetics , Lysine/metabolism , Nucleic Acid Conformation , Point Mutation , Protein Structure, Tertiary/genetics , RecQ Helicases/chemistry , RecQ Helicases/genetics , Substrate Specificity
7.
DNA Repair (Amst) ; 9(3): 331-44, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20075015

ABSTRACT

Werner syndrome protein (WRN) is one of a family of five human RecQ helicases implicated in the maintenance of genome stability. The conserved RecQ family also includes RecQ1, Bloom syndrome protein (BLM), RecQ4, and RecQ5 in humans, as well as Sgs1 in Saccharomyces cerevisiae, Rqh1 in Schizosaccharomyces pombe, and homologs in Caenorhabditis elegans, Xenopus laevis, and Drosophila melanogaster. Defects in three of the RecQ helicases, RecQ4, BLM, and WRN, cause human pathologies linked with cancer predisposition and premature aging. Mutations in the WRN gene are the causative factor of Werner syndrome (WS). WRN is one of the best characterized of the RecQ helicases and is known to have roles in DNA replication and repair, transcription, and telomere maintenance. Studies both in vitro and in vivo indicate that the roles of WRN in a variety of DNA processes are mediated by post-translational modifications, as well as several important protein-protein interactions. In this work, we will summarize some of the early studies on the cellular roles of WRN and highlight the recent findings that shed some light on the link between the protein with its cellular functions and the disease pathology.


Subject(s)
DNA Repair , DNA/metabolism , Genome , RecQ Helicases/metabolism , Animals , DNA Replication , Humans , RecQ Helicases/genetics , Werner Syndrome/enzymology , Werner Syndrome/genetics
8.
J Biol Chem ; 284(45): 31074-84, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19734539

ABSTRACT

8-Oxo-2'-deoxyguanosine (8-oxodG) is one of the most important oxidative DNA lesions, and G-rich telomeric DNA is especially susceptible to oxidative DNA damage. RecQ helicases WRN and BLM and telomere-binding protein POT1 are thought to play roles in telomere maintenance. This study examines the ability of WRN, BLM, and RecQ5 to unwind and POT1 to bind telomeric D-loops containing 8-oxodG. The results demonstrate that WRN and BLM preferentially unwind telomeric D-loops containing 8-oxodG and that POT1 binds with higher affinity to telomeric D-loops with 8-oxodG but shows no preference for telomeric single-stranded DNA with 8-oxodG. We speculate that telomeric D-loops with 8-oxodG may have a greater tendency to form G-quadruplex DNA structures than telomeric DNA lacking 8-oxodG.


Subject(s)
Deoxyguanosine/analogs & derivatives , Exodeoxyribonucleases/metabolism , RecQ Helicases/metabolism , Telomere-Binding Proteins/metabolism , Telomere/chemistry , 8-Hydroxy-2'-Deoxyguanosine , Animals , Base Sequence , Cell Line , Deoxyguanosine/metabolism , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/genetics , Humans , Molecular Sequence Data , Protein Binding , RecQ Helicases/chemistry , RecQ Helicases/genetics , Shelterin Complex , Substrate Specificity , Telomere/genetics , Telomere/metabolism , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/genetics , Werner Syndrome Helicase
9.
J Biol Chem ; 283(41): 27483-27493, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18689797

ABSTRACT

Eukaryotic Okazaki fragment maturation requires complete removal of the initiating RNA primer before ligation occurs. Polymerase delta (Pol delta) extends the upstream Okazaki fragment and displaces the 5'-end of the downstream primer into a single nucleotide flap, which is removed by FEN1 nuclease cleavage. This process is repeated until all RNA is removed. However, a small fraction of flaps escapes cleavage and grows long enough to be coated with RPA and requires the consecutive action of the Dna2 and FEN1 nucleases for processing. Here we tested whether RPA inhibits FEN1 cleavage of long flaps as proposed. Surprisingly, we determined that RPA binding to long flaps made dynamically by polymerase delta only slightly inhibited FEN1 cleavage, apparently obviating the need for Dna2. Therefore, we asked whether other relevant proteins promote long flap cleavage via the Dna2 pathway. The Pif1 helicase, implicated in Okazaki maturation from genetic studies, improved flap displacement and increased RPA inhibition of long flap cleavage by FEN1. These results suggest that Pif1 accelerates long flap growth, allowing RPA to bind before FEN1 can act, thereby inhibiting FEN1 cleavage. Therefore, Pif1 directs long flaps toward the two-nuclease pathway, requiring Dna2 cleavage for primer removal.


Subject(s)
DNA Helicases/metabolism , DNA Replication/physiology , DNA, Fungal/biosynthesis , DNA/metabolism , Oligoribonucleotides/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Acetyltransferases , DNA/genetics , DNA Helicases/genetics , DNA Polymerase III/genetics , DNA Polymerase III/metabolism , DNA, Fungal/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oligoribonucleotides/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
10.
J Biol Chem ; 281(36): 26051-61, 2006 Sep 08.
Article in English | MEDLINE | ID: mdl-16837458

ABSTRACT

Eukaryotic Okazaki fragments are initiated by an RNA/DNA primer and extended by DNA polymerase delta (pol delta) and the replication clamp proliferating cell nuclear antigen (PCNA). Joining of the fragments by DNA ligase I to generate the continuous double-stranded DNA requires complete removal of the RNA/DNA primer. Pol delta extends the upstream Okazaki fragment and displaces the downstream RNA/DNA primer into a flap removed by nuclease cleavage. One proposed pathway for flap removal involves pol delta displacement of long flaps, coating of those flaps by replication protein A (RPA), and sequential cleavage of the flap by Dna2 nuclease followed by flap endonuclease 1 (FEN1). A second pathway involves reiterative single nucleotide or short oligonucleotide displacement by pol delta and cleavage by FEN1. We measured the length of FEN1 cleavage products on flaps strand-displaced by pol delta in an oligonucleotide system reconstituted with Saccharomyces cerevisiae proteins. Results showed that in the presence of PCNA and FEN1, pol delta displacement synthesis favors formation and cleavage of primarily short flaps, up to eight nucleotides in length; still, a portion of flaps grows to 20-30 nucleotides. The proportion of long flaps can be altered by mutations in the relevant proteins, sequence changes in the DNA, and reaction conditions. These results suggest that FEN1 is sufficient to remove a majority of Okazaki fragment primers. However, some flaps become long and require the two-nuclease pathway. It appears that both pathways, operating in parallel, are required for processing of all flaps.


Subject(s)
DNA Polymerase III/metabolism , DNA Primers/metabolism , DNA Replication/physiology , DNA/metabolism , DNA/chemistry , DNA/genetics , Flap Endonucleases/metabolism , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides/metabolism , Proliferating Cell Nuclear Antigen/metabolism , RNA/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Salts/chemistry
12.
Proc Natl Acad Sci U S A ; 101(48): 16762-7, 2004 Nov 30.
Article in English | MEDLINE | ID: mdl-15556996

ABSTRACT

The toroidal damage checkpoint complex Rad9-Rad1-Hus1 (9-1-1) has been characterized as a sensor of DNA damage. Flap endonuclease 1 (FEN1) is a structure-specific nuclease involved both in removing initiator RNA from Okazaki fragments and in DNA repair pathways. FEN1 activity is stimulated by proliferating cell nuclear antigen (PCNA), a toroidal sliding clamp that acts as a platform for DNA replication and repair complexes. We show that 9-1-1 also binds and stimulates FEN1. Stimulation is observed on a variety of flap, nick, and gapped substrates simulating repair intermediates. Blocking 9-1-1 entry to the double strands prevents a portion of the stimulation. Like PCNA stimulation, 9-1-1 stimulation cannot circumvent the tracking mechanism by which FEN1 enters the substrate; however, 9-1-1 does not substitute for PCNA in the stimulation of DNA polymerase beta. This suggests that 9-1-1 is a damage-specific activator of FEN1.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Endonucleases/physiology , Flap Endonucleases/metabolism , Base Sequence , DNA Damage , DNA Primers , Enzyme Activation , Humans , Hydrolysis , Schizosaccharomyces pombe Proteins
13.
J Biol Chem ; 278(44): 42854-66, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-12902352

ABSTRACT

We previously developed a system to investigate the mechanism of repeat sequence expansion during eukaryotic Okazaki fragment processing. Upstream and downstream primers were annealed to a complementary template to overlap across a CAG repeat region. Annealing by the competing primers lead to structural intermediates that ligated to expand the repeat segment. When an equal number of repeats overlapped on the upstream and downstream primers, a 2-fold expansion was expected, but no expansion occurred. We show here that such substrates do not expand irrespective of their repeat length. To reveal mechanism, we tested different hairpin loop intermediates expected to form and facilitate ligation. Substrates configured to form large loops in either the upstream or downstream primer alone allowed expansion. Large or small fixed position single loops allowed expansion when located at least six nucleotides up- or downstream of the nick. Fixed loops in both primers, simulating a double loop intermediate, allowed expansion as long as each loop was nine nucleotides from the nick. Thus, neither the double loop configuration required to form with equal length overlaps nor the large single loop configuration are fundamental structural impediments to expansion. We propose a model for the expansion mechanism based on the relative stabilities of single loop, double loop, hairpin, and flap intermediates that is consistent with the observed expansion efficiency of equal and unequal overlap substrates. The model suggests that the equilibrium concentration of double loop intermediates is so vanishingly small that they are not likely contributors to sequence expansion.


Subject(s)
DNA Replication , DNA/chemistry , Trinucleotide Repeats , Base Sequence , DNA Ligases/chemistry , Escherichia coli/metabolism , Models, Genetic , Molecular Sequence Data , Nucleic Acid Conformation , Oligonucleotides/chemistry , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...