Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cell ; 187(14): 3638-3651.e18, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38838667

ABSTRACT

Telomere maintenance requires the extension of the G-rich telomeric repeat strand by telomerase and the fill-in synthesis of the C-rich strand by Polα/primase. At telomeres, Polα/primase is bound to Ctc1/Stn1/Ten1 (CST), a single-stranded DNA-binding complex. Like mutations in telomerase, mutations affecting CST-Polα/primase result in pathological telomere shortening and cause a telomere biology disorder, Coats plus (CP). We determined cryogenic electron microscopy structures of human CST bound to the shelterin heterodimer POT1/TPP1 that reveal how CST is recruited to telomeres by POT1. Our findings suggest that POT1 hinge phosphorylation is required for CST recruitment, and the complex is formed through conserved interactions involving several residues mutated in CP. Our structural and biochemical data suggest that phosphorylated POT1 holds CST-Polα/primase in an inactive, autoinhibited state until telomerase has extended the telomere ends. We propose that dephosphorylation of POT1 releases CST-Polα/primase into an active state that completes telomere replication through fill-in synthesis.


Subject(s)
DNA Polymerase I , Shelterin Complex , Telomere-Binding Proteins , Telomere , Humans , Cryoelectron Microscopy , DNA Polymerase I/metabolism , DNA Primase/metabolism , DNA Primase/genetics , Models, Molecular , Phosphorylation , Shelterin Complex/metabolism , Telomerase/metabolism , Telomere/metabolism , Telomere-Binding Proteins/metabolism
2.
Nature ; 627(8004): 664-670, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38418884

ABSTRACT

Telomerase adds G-rich telomeric repeats to the 3' ends of telomeres1, counteracting telomere shortening caused by loss of telomeric 3' overhangs during leading-strand DNA synthesis ('the end-replication problem'2). Here we report a second end-replication problem that originates from the incomplete duplication of the C-rich telomeric repeat strand (C-strand) by lagging-strand DNA synthesis. This problem is resolved by fill-in synthesis mediated by polymerase α-primase bound to Ctc1-Stn1-Ten1 (CST-Polα-primase). In vitro, priming for lagging-strand DNA replication does not occur on the 3' overhang and lagging-strand synthesis stops in a zone of approximately 150 nucleotides (nt) more than 26 nt from the end of the template. Consistent with the in vitro data, lagging-end telomeres of cells lacking CST-Polα-primase lost 50-60 nt of telomeric CCCTAA repeats per population doubling. The C-strands of leading-end telomeres shortened by around 100 nt per population doubling, reflecting the generation of 3' overhangs through resection. The measured overall C-strand shortening in the absence of CST-Polα-primase fill-in is consistent with the combined effects of incomplete lagging-strand synthesis and 5' resection at the leading ends. We conclude that canonical DNA replication creates two telomere end-replication problems that require telomerase to maintain the G-rich strand and CST-Polα-primase to maintain the C-strand.


Subject(s)
DNA Polymerase I , DNA Primase , DNA Replication , Telomere-Binding Proteins , Telomere , Humans , DNA Polymerase I/metabolism , DNA Primase/metabolism , Telomerase/metabolism , Telomere/genetics , Telomere/metabolism , Telomere-Binding Proteins/metabolism
3.
Cell ; 150(1): 39-52, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22748632

ABSTRACT

A 3' overhang is critical for the protection and maintenance of mammalian telomeres, but its synthesis must be regulated to avoid excessive resection of the 5' end, which could cause telomere shortening. How this balance is achieved in mammals has not been resolved. Here, we determine the mechanism for 3' overhang synthesis in mouse cells by evaluating changes in telomeric overhangs throughout the cell cycle and at leading- and lagging-end telomeres. Apollo, a nuclease bound to the shelterin subunit TRF2, initiates formation of the 3' overhang at leading-, but not lagging-end telomeres. Hyperresection by Apollo is blocked at both ends by the shelterin protein POT1b. Exo1 extensively resects both telomere ends, generating transient long 3' overhangs in S/G2. CST/AAF, a DNA polα.primase accessory factor, binds POT1b and shortens the extended overhangs produced by Exo1, likely through fill-in synthesis. 3' overhang formation is thus a multistep, shelterin-controlled process, ensuring functional telomeric overhangs at chromosome ends.


Subject(s)
DNA-Binding Proteins/metabolism , Exodeoxyribonucleases/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Amino Acid Sequence , Animals , DNA Replication , Mice , Molecular Sequence Data , Sequence Alignment , Telomere Shortening
4.
Nature ; 560(7716): 112-116, 2018 08.
Article in English | MEDLINE | ID: mdl-30022158

ABSTRACT

In DNA repair, the resection of double-strand breaks dictates the choice between homology-directed repair-which requires a 3' overhang-and classical non-homologous end joining, which can join unresected ends1,2. BRCA1-mutant cancers show minimal resection of double-strand breaks, which renders them deficient in homology-directed repair and sensitive to inhibitors of poly(ADP-ribose) polymerase 1 (PARP1)3-8. When BRCA1 is absent, the resection of double-strand breaks is thought to be prevented by 53BP1, RIF1 and the REV7-SHLD1-SHLD2-SHLD3 (shieldin) complex, and loss of these factors diminishes sensitivity to PARP1 inhibitors4,6-9. Here we address the mechanism by which 53BP1-RIF1-shieldin regulates the generation of recombinogenic 3' overhangs. We report that CTC1-STN1-TEN1 (CST)10, a complex similar to replication protein A that functions as an accessory factor of polymerase-α (Polα)-primase11, is a downstream effector in the 53BP1 pathway. CST interacts with shieldin and localizes with Polα to sites of DNA damage in a 53BP1- and shieldin-dependent manner. As with loss of 53BP1, RIF1 or shieldin, the depletion of CST leads to increased resection. In BRCA1-deficient cells, CST blocks RAD51 loading and promotes the efficacy of PARP1 inhibitors. In addition, Polα inhibition diminishes the effect of PARP1 inhibitors. These data suggest that CST-Polα-mediated fill-in helps to control the repair of double-strand breaks by 53BP1, RIF1 and shieldin.


Subject(s)
DNA Breaks, Double-Stranded , DNA Polymerase I/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Telomere-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Animals , BRCA1 Protein/deficiency , Cell Line , DNA Primase/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Humans , Mice , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Recombinational DNA Repair , Telomere/genetics , Telomere/metabolism
5.
Genes Dev ; 30(7): 812-26, 2016 04 01.
Article in English | MEDLINE | ID: mdl-27013236

ABSTRACT

Coats plus (CP) can be caused by mutations in the CTC1 component of CST, which promotes polymerase α (polα)/primase-dependent fill-in throughout the genome and at telomeres. The cellular pathology relating to CP has not been established. We identified a homozygous POT1 S322L substitution (POT1(CP)) in two siblings with CP. POT1(CP)induced a proliferative arrest that could be bypassed by telomerase. POT1(CP)was expressed at normal levels, bound TPP1 and telomeres, and blocked ATR signaling. POT1(CP)was defective in regulating telomerase, leading to telomere elongation rather than the telomere shortening observed in other telomeropathies. POT1(CP)was also defective in the maintenance of the telomeric C strand, causing extended 3' overhangs and stochastic telomere truncations that could be healed by telomerase. Consistent with shortening of the telomeric C strand, metaphase chromosomes showed loss of telomeres synthesized by leading strand DNA synthesis. We propose that CP is caused by a defect in POT1/CST-dependent telomere fill-in. We further propose that deficiency in the fill-in step generates truncated telomeres that halt proliferation in cells lacking telomerase, whereas, in tissues expressing telomerase (e.g., bone marrow), the truncations are healed. The proposed etiology can explain why CP presents with features distinct from those associated with telomerase defects (e.g., dyskeratosis congenita).


Subject(s)
Ataxia/genetics , Brain Neoplasms/genetics , Calcinosis/genetics , Central Nervous System Cysts/genetics , Leukoencephalopathies/genetics , Muscle Spasticity/genetics , Mutation/genetics , Retinal Diseases/genetics , Seizures/genetics , Telomere Shortening/genetics , Telomere-Binding Proteins/genetics , Telomere/genetics , Telomere/pathology , Aminopeptidases/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Female , Humans , Metaphase , Protein Binding , Serine Proteases/metabolism , Shelterin Complex , Signal Transduction , Telomere/metabolism , Telomere Homeostasis/genetics
6.
Mol Cell ; 39(6): 839-50, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20864032

ABSTRACT

TEL2 interacts with and is essential for the stability of all phosphatidylinositol 3-kinase-related kinases (PIKKs), but its mechanism of action remains unclear. Here, we show that TEL2 is constitutively phosphorylated on conserved serines 487 and 491 by casein kinase 2 (CK2). Proteomic analyses establish that the CK2 phosphosite of TEL2 confers binding to the R2TP/prefoldin-like complex, which possesses chaperon/prefoldin activities required during protein complex assembly. The PIH1D1 subunit of the R2TP complex binds directly to the CK2 phosphosite of TEL2 in vitro and is required for the TEL2-R2TP/prefoldin-like complex interaction in vivo. Although the CK2 phosphosite mutant of TEL2 retains association with the PIKKs and HSP90 in cells, failure to interact with the R2TP/prefoldin-like complex results in instability of the PIKKs, principally mTOR and SMG1. We propose that TEL2 acts as a scaffold to coordinate the activities of R2TP/prefoldin-like and HSP90 chaperone complexes during the assembly of the PIKKs.


Subject(s)
Casein Kinase II/metabolism , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-ets/metabolism , TOR Serine-Threonine Kinases/metabolism , ATPases Associated with Diverse Cellular Activities , Animals , Apoptosis Regulatory Proteins , Ataxia Telangiectasia Mutated Proteins , Binding Sites/physiology , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cytoplasm/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Enzyme Stability , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Models, Biological , Molecular Chaperones/metabolism , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/metabolism , Serine/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism
7.
Genes Dev ; 24(18): 2019-30, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20801936

ABSTRACT

We reported previously that the stability of all mammalian phosphatidylinositol 3-kinase-related protein kinases (PIKKs) depends on their interaction with Tel2, the ortholog of yeast Tel2 and Caenorhabditis elegans Clk-2. Here we provide evidence that Tel2 acts with Hsp90 in the maturation of PIKK complexes. Quantitative immunoblotting showed that the abundance of Tel2 is low compared with the PIKKs, and Tel2 preferentially bound newly synthesized ATM, ATR, mTOR, and DNA-PKcs. Tel2 complexes contained, in addition to Tti1-Tti2, the Hsp90 chaperone, and inhibition of Hsp90 interfered with the interaction of Tel2 with the PIKKs. Analysis of in vivo labeled nascent protein complexes showed that Tel2 and Hsp90 mediate the formation of the mTOR TORC1 and TORC2 complexes and the association of ATR with ATRIP. The structure of yeast Tel2, reported here, shows that Tel2 consists of HEAT-like helical repeats that assemble into two separate α-solenoids. Through mutagenesis, we identify a surface patch of conserved residues involved in binding to the Tti1-Tti2 complex in vitro. In vivo, mutation of this conserved patch affects cell growth, levels of PIKKs, and ATM/ATR-mediated checkpoint signaling, highlighting the importance of Tti1-Tti2 binding to the function of Tel2. Taken together, our data suggest that the Tel2-Tti1-Tti2 complex is a PIKK-specific cochaperone for Hsp90.


Subject(s)
DNA Helicases/genetics , HSP90 Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/metabolism , ATPases Associated with Diverse Cellular Activities , Animals , Cells, Cultured , DNA Helicases/chemistry , HSP90 Heat-Shock Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship , TOR Serine-Threonine Kinases , Telomere-Binding Proteins/genetics
8.
J Cell Sci ; 126(Pt 14): 3095-104, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23857907

ABSTRACT

Spermatogenesis is a complex process that generates haploid germ cells or spores and implements meiosis, a succession of two special cell divisions that are required for homologous chromosome segregation. During prophase to the first meiotic division, homologous recombination (HR) repairs Spo11-dependent DNA double-strand breaks (DSBs) in the presence of telomere movements to allow for chromosome pairing and segregation at the meiosis I division. In contrast to HR, non-homologous end joining (NHEJ), the major DSB repair mechanism during the G1 cell cycle phase, is downregulated during early meiotic prophase. At somatic mammalian telomeres, the NHEJ factor Ku70/80 inhibits HR, as does the Rap1 component of the shelterin complex. Here, we investigated the role of Ku70 and Rap1 in meiotic telomere redistribution and genome protection in spermatogenesis by studying single and double knockout mice. Ku70(-/-) mice display reduced testis size and compromised spermatogenesis, whereas meiotic telomere dynamics and chromosomal bouquet formation occurred normally in Ku70(-/-) and Ku70(-/-)Rap1(Δ/Δ) knockout spermatocytes. Elevated mid-preleptotene frequencies were associated with significantly increased DNA damage in Ku-deficient B spermatogonia, and in differentiated Sertoli cells. Significantly elevated levels of γH2AX foci in Ku70(-/-) diplotene spermatocytes suggest compromised progression of DNA repair at a subset of DSBs. This might explain the elevated meiotic metaphase apoptosis that is present in Ku70-deficient stage XII testis tubules, indicating spindle assembly checkpoint activation. In summary, our data indicate that Ku70 is important for repairing DSBs in somatic cells and in late spermatocytes of the testis, thereby assuring the fidelity of spermatogenesis.


Subject(s)
Antigens, Nuclear/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Spermatogenesis , Testis/pathology , rap1 GTP-Binding Proteins/metabolism , Animals , Antigens, Nuclear/genetics , Apoptosis/genetics , Cell Cycle , Cells, Cultured , DNA Repair/genetics , DNA, Recombinant/genetics , DNA-Binding Proteins/genetics , Ku Autoantigen , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/genetics , Recombination, Genetic , rap1 GTP-Binding Proteins/genetics
9.
Circ J ; 78(6): 1342-8, 2014.
Article in English | MEDLINE | ID: mdl-24717234

ABSTRACT

BACKGROUND: Warfarin reduces the risk of stroke in patients with atrial fibrillation, but requires a moderate-to-high time in therapeutic range (TTR). We hypothesized that point-of-care (POC) testing for prothrombin time-internationalized normalized ratio (PT-INR) could improve the TTR in patients receiving warfarin. METHODS AND RESULTS: Eight outpatient clinics that introduced POC testing for PT-INR participated in this study. We identified 148 consecutive patients who received warfarin for at least 12 months before and after the introduction of POC testing. We compared the TTR before and after the introduction of POC testing for each patient. TTR after the introduction of POC testing was significantly higher than that beforehand (51.9%±33.0% vs. 69.3%±26.3%; P<0.0001). The improvement in TTR was statistically significant in patients who had low TTR (<70%) before the introduction of POC testing. After the introduction of POC, the time spent above the target INR showed no significant change (3.7%±10.6% vs. 3.3%±6.3%, P=0.7322), while that spent below the target INR improved significantly (44.4%±34.4% vs. 27.4%±27.6%, P<0.0001). CONCLUSIONS: The introduction of POC testing was associated with an improvement in TTR, mainly through a reduction in the time spent below the target INR.


Subject(s)
Ambulatory Care Facilities , Anticoagulants/administration & dosage , Point-of-Care Systems , Prothrombin Time , Warfarin/administration & dosage , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Time Factors
10.
bioRxiv ; 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-37961611

ABSTRACT

Telomerase adds G-rich telomeric repeats to the 3' ends of telomeres1, counteracting telomere shortening caused by loss of telomeric 3' overhangs during leading-strand DNA synthesis ("the end-replication problem"2). We report a second end-replication problem that originates from the incomplete duplication of the C-rich telomeric repeat strand by lagging-strand synthesis. This problem is solved by CST-Polymeraseα(Polα)-primase fill-in synthesis. In vitro, priming for lagging-strand DNA replication does not occur on the 3' overhang and lagging-strand synthesis stops in an ~150-nt zone more than 26 nt from the end of the template. Consistent with the in vitro data, lagging-end telomeres of cells lacking CST-Polα-primase lost ~50-60 nt of CCCTAA repeats per population doubling (PD). The C-strands of leading-end telomeres shortened by ~100 nt/PD, reflecting the generation of 3' overhangs through resection. The measured overall C-strand shortening in absence of CST-Polα-primase fill-in is consistent with the combined effects of incomplete lagging-strand synthesis and 5' resection at the leading-ends. We conclude that canonical DNA replication creates two telomere end-replication problems that require telomerase to maintain the G-strand and CST-Polα-primase to maintain the C-strand.

11.
bioRxiv ; 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37215005

ABSTRACT

Telomere maintenance requires extension of the G-rich telomeric repeat strand by telomerase and fill-in synthesis of the C-rich strand by Polα/Primase. Telomeric Polα/Primase is bound to Ctc1-Stn1-Ten1 (CST), a single-stranded DNA-binding complex. Like mutations in telomerase, mutations affecting CST-Polα/Primase result in pathological telomere shortening and cause a telomere biology disorder, Coats plus (CP). We determined cryogenic electron microscopy structures of human CST bound to the shelterin heterodimer POT1/TPP1 that reveal how CST is recruited to telomeres by POT1. Phosphorylation of POT1 is required for CST recruitment, and the complex is formed through conserved interactions involving several residues mutated in CP. Our structural and biochemical data suggest that phosphorylated POT1 holds CST-Polα/Primase in an inactive auto-inhibited state until telomerase has extended the telomere ends. We propose that dephosphorylation of POT1 releases CST-Polα/Primase into an active state that completes telomere replication through fill-in synthesis.

12.
Neurol Med Chir (Tokyo) ; 63(1): 9-16, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36223946

ABSTRACT

With the aging of the population, the number of people taking antithrombotic drugs is increasing. Few reports have described the clinical presentation, treatment, and outcomes of nontraumatic subarachnoid hemorrhage (SAH) in patients with preceding antithrombotic therapy. This study included 459 patients with nontraumatic SAH who had been treated between April 2009 and May 2021. Overall, 39 of the 459 patients with aneurysmal SAH were on antithrombotic therapy before ictus (8.5%). Therefore, we classified patients into two groups: Group A (n = 39), patients with preceding antithrombotic therapy and Group B (n = 420), patients without preceding antithrombotic therapy. Hunt and Kosnik (H&K) grade on admission was significantly higher in Group A than in Group B (p = 0.02). Patients in Group A more frequently received endovascular treatment. The rate of endovascular therapy for symptomatic vasospasm after SAH was significantly lower in Group A (2.6%) than in Group B (15.5%; p = 0.03). The outcomes at 3 months after onset were significantly poorer in Group A patients than in Group B patients (p = 0.03). Patients with preceding antithrombotic drugs tended to be at greater risk of unfavorable outcomes, but this difference was not significant in the univariate analysis. In the multivariate analysis, patient age, H&K grade ≥4, and subdural hematoma remained as risk factors for poor outcomes; however, preceding use of antithrombotic drugs was not a significant risk factor.


Subject(s)
Subarachnoid Hemorrhage , Humans , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/therapy , Fibrinolytic Agents/adverse effects , Prognosis , Hematoma, Subdural , Risk Factors , Treatment Outcome
13.
Neurol Med Chir (Tokyo) ; 61(2): 124-133, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33390558

ABSTRACT

We compared the rate of selective shunt and pattern of monitoring change between single and dual monitoring in patients undergoing carotid endarterectomy (CEA). A total of 121 patients underwent 128 consecutive CEA procedures. Excluding five procedures using internal shunts in a premeditated manner, we classified patients according to the monitoring: Group A (n = 72), patients with single somatosensory evoked potential (SSEP) monitoring; and Group B (n = 51), patients with dual SSEP and motor evoked potential (MEP). Among the 123 CEAs, an internal shunt was inserted in 12 procedures (9.8%) due to significant changes in monitoring (Group A 5.6%, Group B 15.7%, p = 0.07). The rate of shunt use was significantly higher in patients with the absence of contralateral proximal anterior cerebral artery (A1) on magnetic resonance angiography (MRA) than in patients with other types of MRA (p <0.001). Significant monitor changes were seen in 16 (12.5%) in both groups. In four of nine patients in Group B, SSEP and MEP changes were synchronized, and in the remaining five patients, a time lag was evident between SSEP and MEP changes. In conclusion, the rate of internal shunt use tended to be more frequent in patients with dual monitoring than in patients with single SSEP monitoring, but the difference was not significant. Contralateral A1 absence may predict the need for a shunt and care should be taken to monitor changes throughout the entire CEA procedure. Use of dual monitoring can capture ischemic changes due to the complementary relationship, and may reduce the rate of false-negative monitor changes during CEA.


Subject(s)
Carotid Artery, Internal/surgery , Carotid Stenosis/surgery , Endarterectomy, Carotid/adverse effects , Evoked Potentials, Motor , Evoked Potentials, Somatosensory , Intraoperative Neurophysiological Monitoring/methods , Aged , Arteriovenous Shunt, Surgical , Brain Ischemia/diagnostic imaging , Carotid Artery, Internal/pathology , Cerebral Arteries/diagnostic imaging , Endarterectomy, Carotid/methods , Female , Humans , Intraoperative Complications/diagnostic imaging , Japan , Magnetic Resonance Angiography , Male , Middle Aged , Retrospective Studies
14.
Neurol Med Chir (Tokyo) ; 60(8): 383-389, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32669526

ABSTRACT

Although carotid endarterectomy (CEA) is an established procedure, technical modifications are required when anatomical features are unusual. The present study aimed to determine the characteristics of diagnostic features, surgical management, and outcomes of patients with a twisted carotid bifurcation (TCB). We assessed 108 consecutive patients by cervical carotid echography (CCE) and black-blood magnetic resonance imaging (BB-MRI) before they underwent 115 CEA procedures. We classified carotid bifurcation (CB) anatomy based on anteroposterior findings of the internal carotid artery (ICA) and external carotid artery (ECA) determined by cerebral or three-dimensional computed tomographic angiography as follows. The ICA and ECA ran laterally and medially, respectively, in Type 1, overlapped in Type 2, and the ICA and ECA ran medially and laterally, respectively, in Type 3. We also classified the patients according to whether or not they had a TCB and compared their diagnostic findings, clinical characteristics, and surgical outcomes. The numbers of patients with Types 1, 2, and 3 were 74 (64.4%), 32 (27.8%), and 9 (7.8%), respectively, and 13 (11.3%) with a TCB included four patients with Type 2 and all nine patients with Type 3. The appearance of Type 3 differed from that of the other two types on CCE and BB-MR images. After correcting the anatomical location of a TCB, surgical duration and adverse event rates did not significantly differ between patients with and without a TCB. Patients with a TCB could safely undergo CEA after correcting the ICA to the normal position.


Subject(s)
Carotid Artery, External/pathology , Carotid Artery, Internal/pathology , Carotid Stenosis/diagnosis , Carotid Stenosis/surgery , Endarterectomy, Carotid , Aged , Carotid Artery, External/diagnostic imaging , Carotid Artery, Internal/diagnostic imaging , Carotid Stenosis/complications , Computed Tomography Angiography , Female , Humans , Magnetic Resonance Angiography , Male , Middle Aged , Ultrasonography
15.
Opt Express ; 17(8): 6414-20, 2009 Apr 13.
Article in English | MEDLINE | ID: mdl-19365466

ABSTRACT

Local wet etching technique was proposed to fabricate high-performance aspherical mirrors. In this process, only the limited area facing to the small nozzle is removed by etching on objective surface. The desired objective shape is deterministically fabricated by performing the numerically controlled scanning of the nozzle head. Using the technique, a plano-elliptical mirror to focus the neutron beam was successfully fabricated with the figure accuracy of less than 0.5 microm and the focusing gain of 6. The strong and thin focused neutron beam is expected to be a useful tool for the analyses of various material properties.


Subject(s)
Lenses , Computer-Aided Design , Equipment Design , Equipment Failure Analysis , Neutrons , Reproducibility of Results , Sensitivity and Specificity , Surface Properties
16.
Curr Biol ; 13(17): 1549-56, 2003 Sep 02.
Article in English | MEDLINE | ID: mdl-12956959

ABSTRACT

We report cytologic and genetic data indicating that telomere dysfunction induces a DNA damage response in mammalian cells. Dysfunctional, uncapped telomeres, created through inhibition of TRF2, became associated with DNA damage response factors, such as 53BP1, gamma-H2AX, Rad17, ATM, and Mre11. We refer to the domain of telomere-associated DNA damage factors as a Telomere Dysfunction-Induced Focus (TIF). The accumulation of 53BP1 on uncapped telomeres was reduced in the presence of the PI3 kinase inhibitors caffeine and wortmannin, which affect ATM, ATR, and DNA-PK. By contrast, Mre11 TIFs were resistant to caffeine, consistent with previous findings on the Mre11 response to ionizing radiation. A-T cells had a diminished 53BP1 TIF response, indicating that the ATM kinase is a major transducer of this pathway. However, in the absence of ATM, TRF2 inhibition still induced TIFs and senescence, pointing to a second ATM-independent pathway. We conclude that the cellular response to telomere dysfunction is governed by proteins that also control the DNA damage response. TIFs represent a new tool for evaluating telomere status in normal and malignant cells suspected of harboring dysfunctional telomeres. Furthermore, induction of TIFs through TRF2 inhibition provides an opportunity to study the DNA damage response within the context of well-defined, physically marked lesions.


Subject(s)
DNA Damage/genetics , Intracellular Signaling Peptides and Proteins , Phosphoproteins , Telomere/genetics , Telomeric Repeat Binding Protein 2/metabolism , Carrier Proteins/metabolism , Endodeoxyribonucleases/metabolism , Exodeoxyribonucleases/metabolism , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoblotting , Phosphoinositide-3 Kinase Inhibitors , Saccharomyces cerevisiae Proteins/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/antagonists & inhibitors , Transfection , Tumor Suppressor p53-Binding Protein 1
17.
Mol Cell Biol ; 23(18): 6533-41, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12944479

ABSTRACT

The human telomeric DNA binding factor TRF1 (hTRF1) and its interacting proteins TIN2, tankyrase 1 and 2, and PINX1 have been implicated in the regulation of telomerase-dependent telomere length maintenance. Here we show that targeted deletion of exon 1 of the mouse gene encoding Trf1 causes early (day 5 to 6 postcoitus) embryonic lethality. The absence of telomerase did not alter the Terf1(ex1Delta/ex1Delta) lethality, indicating that the phenotype was not due to inappropriate telomere elongation by telomerase. Terf1(ex1Delta/ex1Delta) blastocysts had a severe growth defect of the inner cell mass that was accompanied by apoptosis. However, no evidence was found for telomere uncapping causing this cell death; chromosome spreads of Terf1(ex1Delta/ex1Delta) blastocysts did not reveal chromosome end-to-end fusions, and p53 deficiency only briefly delayed Terf1(ex1Delta/ex1Delta) lethality. These data suggest that murine Trf1 has an essential function that is independent of telomere length regulation.


Subject(s)
Telomere/physiology , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Animals , Blastocyst/pathology , Cell Division , Cells, Cultured , Female , Fetal Death/genetics , Gene Targeting , Mice , Mice, Mutant Strains , Pregnancy , Sequence Deletion , Telomerase/deficiency , Telomerase/genetics , Tumor Suppressor Protein p53/genetics
18.
Mol Cell Biol ; 22(8): 2769-76, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11909969

ABSTRACT

A growing number of DNA polymerases have been identified, although their physiological function and relation to human disease remain mostly unknown. DNA polymerase lambda (Pol lambda; also known as Pol beta2) has recently been identified as a member of the X family of DNA polymerases and shares 32% amino acid sequence identity with DNA Pol beta within the polymerase domain. With the use of homologous recombination, we generated Pol lambda(-/-) mice. Pol lambda(-/-) mice develop hydrocephalus with marked dilation of the lateral ventricles and exhibit a high rate of mortality after birth, although embryonic development appears normal. Pol lambda(-/-) mice also show situs inversus totalis and chronic suppurative sinusitis. The surviving male, but not female, Pol lambda(-/-) mice are sterile as a result of spermatozoal immobility. Microinjection of sperm from male Pol lambda(-/-) mice into oocytes gives rise to normal offspring, suggesting that the meiotic process is not impaired. Ultrastructural analysis reveals that inner dynein arms of cilia from both the ependymal cell layer and respiratory epithelium are defective, which may underlie the pathogenesis of hydrocephalus, situs inversus totalis, chronic sinusitis, and male infertility. Sensitivity of Pol lambda(-/-) cells to various kinds of DNA damage is indistinguishable from that of Pol lambda(+/+) cells. Collectively, Pol lambda(-/-) mice may provide a useful model for clarifying the pathogenesis of immotile cilia syndrome.


Subject(s)
Ciliary Motility Disorders/etiology , DNA Polymerase beta/deficiency , DNA Polymerase beta/genetics , Hydrocephalus/enzymology , Hydrocephalus/genetics , Infertility, Male/enzymology , Infertility, Male/genetics , Sinusitis/enzymology , Sinusitis/genetics , Situs Inversus/enzymology , Situs Inversus/genetics , Animals , Chronic Disease , Ciliary Motility Disorders/enzymology , Ciliary Motility Disorders/genetics , Ciliary Motility Disorders/pathology , DNA Polymerase beta/physiology , Disease Models, Animal , Female , Humans , Hydrocephalus/pathology , Infertility, Male/pathology , Male , Mice , Mice, Knockout , Pregnancy , Sinusitis/pathology , Situs Inversus/pathology , Sperm Injections, Intracytoplasmic
19.
Am J Cardiol ; 96(8): 1037-41, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16214434

ABSTRACT

The ventricular septum receives its blood supply from the septal perforators of the left anterior descending (LAD) coronary artery and the right coronary artery. However, when the LAD artery extends to the inferior wall, beyond the apex (so-called wrapped LAD), the ventricular septum near the apex receives blood supply only from the LAD artery. As a consequence, ventricular septal rupture (VSR) would seem more likely in myocardial infarction with occlusion of this type of LAD artery. To test this hypothesis, we compared electrocardiographic findings in 21 patients who had anterior acute myocardial infarction that was complicated by VSR with those in 275 patients who had acute myocardial infarction that was not complicated by VSR. We observed ST-segment elevation in all inferior leads (II, III, and aVF) in addition to anterior leads in 42.9% of patients (9 of 21) who had VSR but in only 3.6% of those (10 of 275) who did not have VSR. Abnormal Q waves appeared in all 3 inferior leads in 44.4% of patients (8 of 18) who had VSR but in only 4.0% of those (10 of 250) who did not have VSR. Thus, the incidence of ST-segment elevation and abnormal Q waves in the inferior leads was significantly (p <0.001) greater in the VSR group. In addition, multivariate analysis of patient characteristics, including advanced age, female gender, and coronary morphology, showed VSR to be significantly correlated with ST-segment elevation (odds ratio 16.93, 95% confidence interval 4.13 to 69.30) and abnormal Q waves (odds ratio 13.64, 95% confidence interval 3.16 to 58.79) in the 3 inferior leads. In conclusion, these electrocardiographic findings can be useful predictors of complication by VSR.


Subject(s)
Electrocardiography , Myocardial Infarction/complications , Ventricular Septal Rupture/complications , Aged , Female , Hospital Mortality , Humans , Male , Middle Aged , Myocardial Infarction/diagnosis , Predictive Value of Tests , Ventricular Septal Rupture/classification , Ventricular Septal Rupture/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL