Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Genes Cells ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38924305

ABSTRACT

Interhomolog recombination in meiosis is mediated by the Dmc1 recombinase. The Mei5-Sae3 complex of Saccharomyces cerevisiae promotes Dmc1 assembly and functions with Dmc1 for homology-mediated repair of meiotic DNA double-strand breaks. How Mei5-Sae3 facilitates Dmc1 assembly remains poorly understood. In this study, we created and characterized several mei5 mutants featuring the amino acid substitutions of basic residues. We found that Arg97 of Mei5, conserved in its ortholog, SFR1 (complex with SWI5), RAD51 mediator, in humans and other organisms, is critical for complex formation with Sae3 for Dmc1 assembly. Moreover, the substitution of either Arg117 or Lys133 with Ala in Mei5 resulted in the production of a C-terminal truncated Mei5 protein during yeast meiosis. Notably, the shorter Mei5-R117A protein was observed in meiotic cells but not in mitotic cells when expressed, suggesting a unique regulation of Dmc1-mediated recombination by posttranslational processing of Mei5-Sae3.

2.
Sci Rep ; 14(1): 9550, 2024 04 25.
Article in English | MEDLINE | ID: mdl-38664461

ABSTRACT

DNA double-strand breaks (DSBs) activate DNA damage responses (DDRs) in both mitotic and meiotic cells. A single-stranded DNA (ssDNA) binding protein, Replication protein-A (RPA) binds to the ssDNA formed at DSBs to activate ATR/Mec1 kinase for the response. Meiotic DSBs induce homologous recombination monitored by a meiotic DDR called the recombination checkpoint that blocks the pachytene exit in meiotic prophase I. In this study, we further characterized the essential role of RPA in the maintenance of the recombination checkpoint during Saccharomyces cerevisiae meiosis. The depletion of an RPA subunit, Rfa1, in a recombination-defective dmc1 mutant, fully alleviates the pachytene arrest with the persistent unrepaired DSBs. RPA depletion decreases the activity of a meiosis-specific CHK2 homolog, Mek1 kinase, which in turn activates the Ndt80 transcriptional regulator for pachytene exit. These support the idea that RPA is a sensor of ssDNAs for the activation of meiotic DDR. Rfa1 depletion also accelerates the prophase I delay in the zip1 mutant defective in both chromosome synapsis and the recombination, consistent with the notion that the accumulation of ssDNAs rather than defective synapsis triggers prophase I delay in the zip1 mutant.


Subject(s)
DNA Breaks, Double-Stranded , Meiosis , Replication Protein A , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription Factors , Replication Protein A/metabolism , Replication Protein A/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Recombination, Genetic , Homologous Recombination , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 1/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics
3.
Genes Cells ; 29(1): 86-98, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37968127

ABSTRACT

The mitotic cohesin complex necessary for sister chromatid cohesion and chromatin loop formation shows local and global association to chromosomes in response to DNA double-strand breaks (DSBs). Here, by genome-wide binding analysis of the meiotic cohesin with Rec8, we found that the Rec8-localization profile along chromosomes is altered from middle to late meiotic prophase I with cleavage-independent dissociation. Each Rec8-binding site on the chromosome axis follows a unique alternation pattern with dissociation and probably association. Centromeres showed altered Rec8 binding in late prophase I relative to mid-prophase I, implying chromosome remodeling of the regions. Rec8 dissociation ratio per chromosome is correlated well with meiotic DSB density. Indeed, the spo11 mutant deficient in meiotic DSB formation did not change the distribution of Rec8 along chromosomes in late meiotic prophase I. These suggest the presence of a meiosis-specific regulatory pathway for the global binding of Rec8-cohesin in response to DSBs.


Subject(s)
Meiosis , Saccharomyces cerevisiae , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cohesins , DNA/metabolism , DNA Breaks, Double-Stranded , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
4.
DNA Repair (Amst) ; 134: 103613, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38142595

ABSTRACT

RAD51 recombinase plays a central role in homologous recombination (HR) by forming a nucleoprotein filament on single-stranded DNA (ssDNA) to catalyze homology search and strand exchange between the ssDNA and a homologous double-stranded DNA (dsDNA). The catalytic activity of RAD51 assembled on ssDNA is critical for the DNA-homology-mediated repair of DNA double-strand breaks in somatic and meiotic cells and restarting stalled replication forks during DNA replication. The RAD51-ssDNA complex also plays a structural role in protecting the regressed/reversed replication fork. Two types of regulators control RAD51 filament formation, stability, and dynamics, namely positive regulators, including mediators, and negative regulators, so-called remodelers. The appropriate balance of action by the two regulators assures genome stability. This review describes the roles of positive and negative RAD51 regulators in HR and DNA replication and its meiosis-specific homolog DMC1 in meiotic recombination. We also provide future study directions for a comprehensive understanding of RAD51/DMC1-mediated regulation in maintaining and inheriting genome integrity.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , DNA-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Homologous Recombination , Rad51 Recombinase/metabolism , DNA Replication , Meiosis , DNA, Single-Stranded
5.
Nat Commun ; 14(1): 6857, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37891173

ABSTRACT

The formation of RAD51/DMC1 filaments on single-stranded (ss)DNAs essential for homology search and strand exchange in DNA double-strand break (DSB) repair is tightly regulated. FIGNL1 AAA+++ ATPase controls RAD51-mediated recombination in human cells. However, its role in gametogenesis remains unsolved. Here, we characterized a germ line-specific conditional knockout (cKO) mouse of FIGNL1. Fignl1 cKO male mice showed defective chromosome synapsis and impaired meiotic DSB repair with the accumulation of RAD51/DMC1 on meiotic chromosomes, supporting a positive role of FIGNL1 in homologous recombination at a post-assembly stage of RAD51/DMC1 filaments. Fignl1 cKO spermatocytes also accumulate RAD51/DMC1 on chromosomes in pre-meiotic S-phase. These RAD51/DMC1 assemblies are independent of meiotic DSB formation. We also showed that purified FIGNL1 dismantles RAD51 filament on double-stranded (ds)DNA as well as ssDNA. These results suggest an additional role of FIGNL1 in limiting the non-productive assembly of RAD51/DMC1 on native dsDNAs during pre-meiotic S-phase and meiotic prophase I.


Subject(s)
Meiosis , Rad51 Recombinase , Male , Humans , Animals , Mice , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , DNA Breaks, Double-Stranded , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Homologous Recombination , DNA , DNA Replication , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics
6.
Genes Genet Syst ; 98(1): 45-52, 2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37225456

ABSTRACT

Meiotic recombination between homologous chromosomes is promoted by the collaborative action of two RecA homologs, Rad51 and meiosis-specific Dmc1. The filament assembly of Dmc1 is promoted by meiosis-specific Mei5-Sae3 in budding yeast. Mei5-Sae3 shows sequence similarity to fission yeast Sfr1-Swi5, which stimulates DNA strand exchanges by Rad51 as well as Dmc1. Sae3 and Swi5 share a conserved motif with the amino acid sequence YNEI/LK/RD. In this study, we analyzed the role of the YNEL residues in the Sae3 sequence in meiotic recombination and found that these residues are critical for Sae3 function in Dmc1 assembly. L59 substitution in the Sae3 protein disrupts complex formation with Mei5, while Y56 and N57 substitutions do not. These observations reveal the differential contribution of conserved YNEL residues to Sae3 activities in meiotic recombination.


Subject(s)
Chromosomal Proteins, Non-Histone , Meiosis , Saccharomyces cerevisiae Proteins , Amino Acids/genetics , Amino Acids/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Meiosis/genetics , Rad51 Recombinase/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
7.
Front Cell Dev Biol ; 10: 1097446, 2022.
Article in English | MEDLINE | ID: mdl-36684419

ABSTRACT

Meiocytes organize higher-order chromosome structures comprising arrays of chromatin loops organized at their bases by linear axes. As meiotic prophase progresses, the axes of homologous chromosomes align and synapse along their lengths to form ladder-like structures called synaptonemal complexes (SCs). The entire process of meiotic recombination, from initiation via programmed DNA double-strand breaks (DSBs) to completion of DSB repair with crossover or non-crossover outcomes, occurs in the context of chromosome axes and SCs. These meiosis-specific chromosome structures provide specialized environments for the regulation of DSB formation and crossing over. In this review, we summarize insights into the importance of chromosome architecture in the regulation of meiotic recombination, focusing on cohesin-mediated axis formation, DSB regulation via tethered loop-axis complexes, inter-homolog template bias facilitated by axial proteins, and crossover regulation in the context of the SCs. We also discuss emerging evidence that the SUMO and the ubiquitin-proteasome system function in the organization of chromosome structure and regulation of meiotic recombination.

9.
Elife ; 102021 01 27.
Article in English | MEDLINE | ID: mdl-33502312

ABSTRACT

Protein modification by SUMO helps orchestrate the elaborate events of meiosis to faithfully produce haploid gametes. To date, only a handful of meiotic SUMO targets have been identified. Here, we delineate a multidimensional SUMO-modified meiotic proteome in budding yeast, identifying 2747 conjugation sites in 775 targets, and defining their relative levels and dynamics. Modified sites cluster in disordered regions and only a minority match consensus motifs. Target identities and modification dynamics imply that SUMOylation regulates all levels of chromosome organization and each step of meiotic prophase I. Execution-point analysis confirms these inferences, revealing functions for SUMO in S-phase, the initiation of recombination, chromosome synapsis and crossing over. K15-linked SUMO chains become prominent as chromosomes synapse and recombine, consistent with roles in these processes. SUMO also modifies ubiquitin, forming hybrid oligomers with potential to modulate ubiquitin signaling. We conclude that SUMO plays diverse and unanticipated roles in regulating meiotic chromosome metabolism.


Most mammalian, yeast and other eukaryote cells have two sets of chromosomes, one from each parent, which contain all the cell's DNA. Sex cells ­ like the sperm and egg ­ however, have half the number of chromosomes and are formed by a specialized type of cell division known as meiosis. At the start of meiosis, each cell replicates its chromosomes so that it has twice the amount of DNA. The cell then undergoes two rounds of division to form sex cells which each contain only one set of chromosomes. Before the cell divides, the two duplicated sets of chromosomes pair up and swap sections of their DNA. This exchange allows each new sex cell to have a unique combination of DNA, resulting in offspring that are genetically distinct from their parents. This complex series of events is tightly regulated, in part, by a protein called the 'small ubiquitin-like modifier' (or SUMO for short), which attaches itself to other proteins and modifies their behavior. This process, known as SUMOylation, can affect a protein's stability, where it is located in the cell and how it interacts with other proteins. However, despite SUMO being known as a key regulator of meiosis, only a handful of its protein targets have been identified. To gain a better understanding of what SUMO does during meiosis, Bhagwat et al. set out to find which proteins are targeted by SUMO in budding yeast and to map the specific sites of modification. The experiments identified 2,747 different sites on 775 different proteins, suggesting that SUMO regulates all aspects of meiosis. Consistently, inactivating SUMOylation at different times revealed SUMO plays a role at every stage of meiosis, including the replication of DNA and the exchanges between chromosomes. In depth analysis of the targeted proteins also revealed that SUMOylation targets different groups of proteins at different stages of meiosis and interacts with other protein modifications, including the ubiquitin system which tags proteins for destruction. The data gathered by Bhagwat et al. provide a starting point for future research into precisely how SUMO proteins control meiosis in yeast and other organisms. In humans, errors in meiosis are the leading cause of pregnancy loss and congenital diseases. Most of the proteins identified as SUMO targets in budding yeast are also present in humans. So, this research could provide a platform for medical advances in the future. The next step is to study mammalian models, such as mice, to confirm that the regulation of meiosis by SUMO is the same in mammals as in yeast.


Subject(s)
Meiosis , SUMO-1 Protein/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/physiology , Sumoylation , Chromosome Pairing , Prophase , SUMO-1 Protein/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
10.
J Emerg Med ; 60(3): e39-e44, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33353810

ABSTRACT

BACKGROUND: Pseudomembranous tracheobronchitis (PMTB) is a rare condition characterized by the formation of endobronchial pseudomembranes. PMTB overlaps with necrotizing tracheobronchitis or plastic bronchitis. The reported infectious etiology mainly includes invasive aspergillosis. PMTB can cause serious airway obstruction; however, urgent tracheotomy is rarely required. CASE REPORT: A 46-year-old woman was transferred to the emergency department (ED) with a 1-week history of progressive dyspnea and cough that was preceded by fever and sore throat. She was previously healthy except for a 20-year history of mild palmoplantar pustulosis. Stridor was evident. Nasolaryngoscopy performed in the ED revealed severe tracheal stenosis caused primarily by mucosal edema and secondarily by pseudomembranes. Initially, tracheitis was considered the sole cause of dyspnea. Although she underwent urgent tracheotomy to prevent asphyxia, her respiration deteriorated progressively. Bronchoscopy revealed massive pseudomembranes obstructing the bilateral bronchi, which led to the clinical diagnosis of PMTB. Subsequent toilet bronchoscopy markedly improved her ventilation. The causative pathogen was not identified despite extensive work-up, including molecular biological testing. Histopathologic examination of the pseudomembranes revealed fibrin with abundant neutrophils, which was consistent with PMTB. Associated conditions, including immunodeficiency, were not found. Her condition improved with antibiotics and repeated toilet bronchoscopy. WHY SHOULD AN EMERGENCY PHYSICIANS BE AWARE OF THIS?: PMTB is an important differential diagnosis of airway emergencies. PMTB can present with critical edematous tracheal stenosis and masked bronchial pseudomembranous obstruction. Emergency physicians should include PMTB in the differential diagnosis in adult patients with acute central airway obstruction because it requires prompt multimodal treatment.


Subject(s)
Airway Obstruction , Aspergillosis , Bronchitis , Tracheal Stenosis , Tracheitis , Adult , Airway Obstruction/etiology , Bronchitis/complications , Bronchitis/diagnosis , Bronchoscopy , Female , Humans , Middle Aged , Tracheal Stenosis/complications , Tracheal Stenosis/diagnosis , Tracheitis/complications , Tracheitis/diagnosis
11.
Methods Mol Biol ; 2153: 267-286, 2021.
Article in English | MEDLINE | ID: mdl-32840786

ABSTRACT

Crossing-over between homologous chromosomes is essential for accurate chromosome segregation at anaphase-I of meiosis. Defective crossing-over is associated with infertility, pregnancy miscarriage, and congenital disease. This chapter presents optimized protocols for the analysis of meiotic crossovers at the cytological level in spermatocytes and oocytes from mouse. The first approach employs immunocytology to detect MLH1, a DNA mismatch-repair protein that specifically marks crossover sites in the pachytene stage of meiotic prophase-I. These immunocytological methods have general utility for the analysis of other recombination steps, such as initiation and DNA strand exchange. The second approach visualizes chiasmata, the points of physical exchange between homologous chromosomes that are present during the diakinesis and metaphase-I stages. Both approaches are readily adaptable to the analysis of crossing over in other vertebrate species.


Subject(s)
Crossing Over, Genetic , MutL Protein Homolog 1/metabolism , Oocytes/cytology , Spermatocytes/cytology , Aneuploidy , Animals , Cells, Cultured , Chromosomes, Mammalian/metabolism , Female , Immunohistochemistry , Male , Mice , Oocytes/metabolism , Pachytene Stage , Spermatocytes/metabolism
12.
Nature ; 586(7830): 623-627, 2020 10.
Article in English | MEDLINE | ID: mdl-32814343

ABSTRACT

During meiosis, crossover recombination connects homologous chromosomes to direct their accurate segregation1. Defective crossing over causes infertility, miscarriage and congenital disease. Each pair of chromosomes attains at least one crossover via the formation and biased resolution of recombination intermediates known as double Holliday junctions2,3. A central principle of crossover resolution is that the two Holliday junctions are resolved in opposite planes by targeting nuclease incisions to specific DNA strands4. The endonuclease activity of the MutLγ complex has been implicated in the resolution of crossovers5-10, but the mechanisms that activate and direct strand-specific cleavage remain unknown. Here we show that the sliding clamp PCNA is important for crossover-biased resolution. In vitro assays with human enzymes show that PCNA and its loader RFC are sufficient to activate the MutLγ endonuclease. MutLγ is further stimulated by a co-dependent activity of the pro-crossover factors EXO1 and MutSγ, the latter of which binds Holliday junctions11. MutLγ also binds various branched DNAs, including Holliday junctions, but does not show canonical resolvase activity, implying that the endonuclease incises adjacent to junction branch points to achieve resolution. In vivo, RFC facilitates MutLγ-dependent crossing over in budding yeast. Furthermore, PCNA localizes to prospective crossover sites along synapsed chromosomes. These data highlight similarities between crossover resolution and the initiation steps of DNA mismatch repair12,13 and evoke a novel model for crossover-specific resolution of double Holliday junctions during meiosis.


Subject(s)
Crossing Over, Genetic , Endonucleases/metabolism , Meiosis , MutL Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Adenosine Triphosphate/metabolism , Animals , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , Enzyme Activation , Humans , Hydrolysis , Male , Mice , MutS Proteins/metabolism , Protein Binding , Replication Protein C/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
13.
Respirol Case Rep ; 8(6): e00603, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32547768

ABSTRACT

We describe the case of a 29-year-old female non-smoker who was treated with steroid therapy for a subacute exacerbation of multisystem Langerhans cell histiocytosis (MS-LCH) with worsening lung, skin, and oral mucosal lesions. The patient developed pneumonia, and computed tomography (CT) showed multiple thin-walled cavities. Transbronchial lung cryobiopsy (TBLC) specimens revealed Langerhans cells, which were positive for CD1a and S-100 expression. Similar histological findings were detected in the submandibular gland, skin, and tooth. On the basis of these findings, the patient was diagnosed with MS-LCH and subsequently treated with steroid therapy. From the literature review, case reports of non-smokers with pulmonary lesions that worsened and required treatment are rare. Almost all cases recurred and needed additional treatment. This case study contributes to our understanding of the potential role of steroid therapy in MS-LCH treatment. Additionally, TBLC is a novel, potentially safer, diagnostic tool that has not been previously described for LCH.

14.
Respirol Case Rep ; 8(6): e00611, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32587701

ABSTRACT

Organizing pneumonia (OP) is a common interstitial lung disease, pathologically characterized by polypoid granulation tissue in the alveolar ducts and alveoli. In clinical practice, OP occasionally presents as non-resolving pneumonia. The typical radiographic pattern of OP is characterized by dense consolidation with ground-glass opacities. Diffuse micronodular pattern of OP (MNOP) is a rare radiographic manifestation that mimics non-resolving bronchiolar diseases such as pulmonary tuberculosis or hypersensitivity pneumonitis. Steroid therapy is usually effective for MNOP; however, spontaneous remission in MNOP has never been reported. Herein, we report a case of a diffuse micronodular form of cryptogenic OP (COP) that was diagnosed via transbronchial biopsy (TBB) and resolved spontaneously within a few months. Our case highlights that MNOP may resolve spontaneously similar to other forms of OP, and mild cases may be under-recognized. Furthermore, careful observation could be an option for managing MNOP with mild and non-progressive symptoms.

15.
Nat Commun ; 11(1): 3101, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32555348

ABSTRACT

Orderly chromosome segregation is enabled by crossovers between homologous chromosomes in the first meiotic division. Crossovers arise from recombination-mediated repair of programmed DNA double-strand breaks (DSBs). Multiple DSBs initiate recombination, and most are repaired without crossover formation, although one or more generate crossovers on each chromosome. Although the underlying mechanisms are ill-defined, the differentiation and maturation of crossover-specific recombination intermediates requires the cyclin-like CNTD1. Here, we identify PRR19 as a partner of CNTD1. We find that, like CNTD1, PRR19 is required for timely DSB repair and the formation of crossover-specific recombination complexes. PRR19 and CNTD1 co-localise at crossover sites, physically interact, and are interdependent for accumulation, indicating a PRR19-CNTD1 partnership in crossing over. Further, we show that CNTD1 interacts with a cyclin-dependent kinase, CDK2, which also accumulates in crossover-specific recombination complexes. Thus, the PRR19-CNTD1 complex may enable crossover differentiation by regulating CDK2.


Subject(s)
Crossing Over, Genetic/genetics , Cyclins/genetics , DNA Breaks, Double-Stranded , Meiosis/genetics , Animals , Chromosomes/genetics , Cyclin-Dependent Kinase 2/genetics , DNA Damage/genetics , DNA Repair/genetics , Female , Homologous Recombination/genetics , Male , Mice
16.
Biointerphases ; 15(2): 021010, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32272844

ABSTRACT

With regard to life sciences, it is important to understand biological functions such as metabolic reactions at the cellular level. Time-of-flight secondary ion mass spectrometry (TOF-SIMS) that can provide chemical mappings at 100 nm lateral resolutions is useful for obtaining three-dimensional maps of biological molecules in cells and tissues. TOF-SIMS spectra generally contain several hundred to several thousand secondary ion peaks that provide detailed chemical information. In order to manage such a large number of peaks, data analysis methods such as multivariate analysis techniques have been applied to TOF-SIMS data of complex samples. However, the interpretation of the data analysis results is sometimes still difficult, especially for biological samples. In this study, TOF-SIMS data of resin-embedded plant samples were analyzed using one of the sparse modeling methods, least absolute shrinkage and selection operator (LASSO), to directly select secondary ions related to biological structures such as cell walls and nuclei. The same sample was measured by optical microscopy and the same measurement area as TOF-SIMS was extracted in order to prepare a target image for LASSO. The same area of the TOF-SIMS and microscope data were fused to evaluate the influence of the image fusion on the TOF-SIMS spectrum information using principal component analysis. Specifically, the authors examined onion mycorrhizal root colonized with Gigaspora margarita (an arbuscular mycorrhizal fungus). The results showed that by employing this approach using LASSO, important secondary ions from biological samples were effectively selected and could be clearly distinguished from the embedding resin.


Subject(s)
Algorithms , Fungi/chemistry , Spectrometry, Mass, Secondary Ion , Image Processing, Computer-Assisted , Ions , Principal Component Analysis
18.
Asia Pac J Clin Oncol ; 15(2): e3-e10, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29316288

ABSTRACT

AIM: In some developed countries, a proportion of nonsquamous non-small cell lung cancer (NSq NSCLC) patients are aged over 70 years when they are diagnosed. However, evidence of lung cancer chemotherapy usually comes from randomized controlled trials that only recruit younger patients with good performance status. In daily practice, less-toxic carboplatin + pemetrexed regimen is often used for elderly patients, although this regimen is not sufficiently supported by rigid evidence for elderly cases. METHODS: The protocol was registered on PROSPERO website (42017058508). Any phase trial that evaluated the efficacy and safety of carboplatin + pemetrexed in the elderly (aged 70 or higher) was included. Binary data were meta-analyzed with the random-model generic inverse variance method. Median survival duration was pooled after logarithmic transformation. RESULTS: Eight studies consisting of 285 patients were included among 882 articles that met the preliminary criteria. The pooled median overall survival and progression-free survivals were 14.9 months (95% confidence interval [95% CI], 12.0-18.4) and 5.4 months (95% CI, 4.5-6.4), respectively. The pooled response rate was 34.0% (95% CI, 27.5-40.5). Hematological adverse events such as neutropenia (≥grade 3; 48.3%; 95% CI, 40.1-56.6), thrombocytopenia (≥grade 3; 27.9%; 95% CI, 15.8-39.9), and anemia (≥grade 3; 17.1%; 95% CI, 8.3-25.8) were frequently observed. However, febrile neutropenia (6.8%; 95% CI, 0.2-13.3), nausea (≥grade 3; 0%; 95% CI, 0.0-4.4%) and treatment-related death (0.6%; 95% CI, 0-5.4%) were rare. CONCLUSION: Carboplatin + pemetrexed can be a good option for the treatment of the elderly with NSq NSCLC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Pemetrexed/therapeutic use , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carboplatin/pharmacology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Pemetrexed/pharmacology , Survival Analysis , Treatment Outcome
19.
Front Robot AI ; 6: 142, 2019.
Article in English | MEDLINE | ID: mdl-33501157

ABSTRACT

We developed a system to evaluate the skill of operating a hydraulic excavator. The system employs a remotely controlled (RC) excavator and virtual reality (VR) technology. We remodeled the RC excavator so that it can be operated in the same manner as a real excavator and proceeded to measure the excavator's state. To evaluate the skill of operating this system, we calculated several indices from the data recorded during excavation work and compared the indices obtained for expert and non-expert operators. The results revealed that it is possible to distinguish whether an expert or non-expert is operating the RC excavator. We calculated the same indices from the data recorded during excavation with a real excavator and verified that there exists a high correlation between the indices of the RC excavator and those of the real excavator. Thus, we confirmed that the indices of the real excavator and those of the simulator exhibited similar trends. This suggests that it is possible to partly evaluate the operation characteristics of a real excavator by using an RC excavator with different dynamics compared with a real excavator.

20.
Genetics ; 207(4): 1313-1333, 2017 12.
Article in English | MEDLINE | ID: mdl-28986445

ABSTRACT

Saccharomyces cerevisiae Mek1 is a CHK2/Rad53-family kinase that regulates meiotic recombination and progression upon its activation in response to DNA double-strand breaks (DSBs). The full catalog of direct Mek1 phosphorylation targets remains unknown. Here, we show that phosphorylation of histone H3 on threonine 11 (H3 T11ph) is induced by meiotic DSBs in S. cerevisiae and Schizosaccharomyces pombe Molecular genetic experiments in S. cerevisiae confirmed that Mek1 is required for H3 T11ph and revealed that phosphorylation is rapidly reversed when Mek1 kinase is no longer active. Reconstituting histone phosphorylation in vitro with recombinant proteins demonstrated that Mek1 directly catalyzes H3 T11 phosphorylation. Mutating H3 T11 to nonphosphorylatable residues conferred no detectable defects in otherwise unperturbed meiosis, although the mutations modestly reduced spore viability in certain strains where Rad51 is used for strand exchange in place of Dmc1. H3 T11ph is therefore mostly dispensable for Mek1 function. However, H3 T11ph provides an excellent marker of ongoing Mek1 kinase activity in vivo Anti-H3 T11ph chromatin immunoprecipitation followed by deep sequencing demonstrated that H3 T11ph was highly enriched at presumed sites of attachment of chromatin to chromosome axes, gave a more modest signal along chromatin loops, and was present at still lower levels immediately adjacent to DSB hotspots. These localization patterns closely tracked the distribution of Red1 and Hop1, axis proteins required for Mek1 activation. These findings provide insight into the spatial disposition of Mek1 kinase activity and the higher order organization of recombining meiotic chromosomes.


Subject(s)
DNA-Binding Proteins/genetics , Histones/genetics , MAP Kinase Kinase 1/genetics , Saccharomyces cerevisiae Proteins/genetics , Cell Cycle Proteins/genetics , Chromosomes, Fungal/genetics , DNA Breaks, Double-Stranded , Meiosis/genetics , Mutation , Phosphorylation , Rad51 Recombinase/genetics , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Spores/genetics , Spores/growth & development , Threonine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...