Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
BMJ ; 385: e078876, 2024 05 28.
Article in English | MEDLINE | ID: mdl-38806195

ABSTRACT

OBJECTIVE: To evaluate the efficacy and safety of tislelizumab added to chemotherapy as first line (primary) treatment for advanced gastric or gastro-oesophageal junction adenocarcinoma compared with placebo plus chemotherapy. DESIGN: Randomised, double blind, placebo controlled, phase 3 study. SETTING: 146 medical centres across Asia, Europe, and North America, between 13 December 2018 and 28 February 2023. PARTICIPANTS: 1657 patients aged ≥18 years with human epidermal growth factor receptor 2 negative locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma, regardless of programmed death-ligand 1 (PD-L1) expression status, who had not received systemic anticancer therapy for advanced disease. INTERVENTIONS: Patients were randomly (1:1) assigned to receive either tislelizumab 200 mg or placebo intravenously every three weeks in combination with chemotherapy (investigator's choice of oxaliplatin and capecitabine, or cisplatin and 5-fluorouracil) and stratified by region, PD-L1 expression, presence or absence of peritoneal metastases, and investigator's choice of chemotherapy. Treatment continued until disease progression or unacceptable toxicity. MAIN OUTCOME MEASURES: The primary endpoint was overall survival, both in patients with a PD-L1 tumour area positivity (TAP) score of ≥5% and in all randomised patients. Safety was assessed in all those who received at least one dose of study treatment. RESULTS: Of 1657 patients screened between 13 December 2018 and 9 February 2021, 660 were ineligible due to not meeting the eligibility criteria, withdrawal of consent, adverse events, or other reasons. Overall, 997 were randomly assigned to receive tislelizumab plus chemotherapy (n=501) or placebo plus chemotherapy (n=496). Tislelizumab plus chemotherapy showed statistically significant improvements in overall survival versus placebo plus chemotherapy in patients with a PD-L1 TAP score of ≥5% (median 17.2 months v 12.6 months; hazard ratio 0.74 (95% confidence interval 0.59 to 0.94); P=0.006 (interim analysis)) and in all randomised patients (median 15.0 months v 12.9 months; hazard ratio 0.80 (0.70 to 0.92); P=0.001 (final analysis)). Grade 3 or worse treatment related adverse events were observed in 54% (268/498) of patients in the tislelizumab plus chemotherapy arm versus 50% (246/494) in the placebo plus chemotherapy arm. CONCLUSIONS: Tislelizumab added to chemotherapy as primary treatment for advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma provided superior overall survival with a manageable safety profile versus placebo plus chemotherapy in patients with a PD-L1 TAP score of ≥5%, and in all randomised patients. TRIAL REGISTRATION: ClinicalTrials.gov NCT03777657.


Subject(s)
Adenocarcinoma , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Esophageal Neoplasms , Esophagogastric Junction , Stomach Neoplasms , Humans , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Male , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/mortality , Female , Middle Aged , Double-Blind Method , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/mortality , Esophagogastric Junction/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aged , Adult , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Capecitabine/administration & dosage , Capecitabine/therapeutic use , Fluorouracil/administration & dosage , Fluorouracil/therapeutic use
2.
Biomark Res ; 11(1): 25, 2023 Mar 07.
Article in English | MEDLINE | ID: mdl-36879284

ABSTRACT

BACKGROUND: Activated immune cells (IC) in the tumor microenvironment (TME) are critical for anti-tumor efficacy. Greater understanding of the dynamic diversity and crosstalk between IC is needed to clarify their association with immune checkpoint inhibitor efficacy. METHODS: Patients from three tislelizumab monotherapy trials in solid tumors (NCT02407990, NCT04068519, NCT04004221) were retrospectively divided into subgroups by CD8+ T-cell and macrophage (Mφ) levels, assessed via multiplex immunohistochemistry (mIHC; n = 67) or gene expression profiling (GEP; n = 629). RESULTS: A trend of longer survival was observed in patients with both high CD8+ T-cell and Mφ levels versus other subgroups in the mIHC analysis (P = 0.11), which was confirmed with greater statistical significance in the GEP analysis (P = 0.0001). Co-existence of CD8+ T cells and Mφ was coupled with elevated CD8+ T-cell cytotoxicity, T-cell trafficking, MHC class I antigen presentation signatures/genes, and enrichment of the pro-inflammatory Mφ polarization pathway. Additionally, a high level of pro-inflammatory CD64+ Mφ density was associated with an immune-activated TME and survival benefit with tislelizumab (15.2 vs. 5.9 months for low density; P = 0.042). Spatial proximity analysis revealed that closer proximity between CD8+ T cells and CD64+ Mφ was associated with a survival benefit with tislelizumab (15.2 vs. 5.3 months for low proximity; P = 0.024). CONCLUSIONS: These findings support the potential role of crosstalk between pro-inflammatory Mφ and cytotoxic T cells in the clinical benefit of tislelizumab. TRIAL REGISTRATION: NCT02407990, NCT04068519, NCT04004221.

3.
Br J Cancer ; 128(8): 1418-1428, 2023 04.
Article in English | MEDLINE | ID: mdl-36797356

ABSTRACT

BACKGROUND: Many patients do not respond or eventually relapse on treatment with programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) checkpoint inhibitors due to secondary or acquired resistance; therefore, there is a need to investigate novel PD-1/PD-L1 inhibitors. METHODS: This open-label, non-randomised study investigated the safety and anti-tumour activity of BGB-A333, a PD-L1 inhibitor, alone and in combination with tislelizumab in patients with advanced solid tumours with progression during/after standard therapy. The primary objectives were to determine the recommended Phase 2 dose (RP2D), safety and tolerability for BGB-A333 alone and in combination with tislelizumab (Phase 1a/1b) and to determine the overall response rate (ORR) with BGB-A333 plus tislelizumab (Phase 2). RESULTS: Overall, 39 patients across Phase 1a (N = 15), 1b (N = 12) and 2 (N = 12) were enroled. In Phase 1a, an RP2D of 1350 mg was determined. In Phase 1a and 1b/2, serious treatment-emergent adverse events (TEAEs) were reported in five and eight patients, respectively. Two patients experienced TEAEs that led to death. In Phase 2, the ORR was 41.7% (n = 5/12; 95% confidence interval: 15.17%, 72.33%). CONCLUSIONS: TEAEs reported with BGB-A333 were consistent with other PD-L1 inhibitors. Encouraging preliminary anti-tumour activity was observed with BGB-A333 in combination with tislelizumab. CLINICAL TRIAL REGISTRATION: NCT03379259.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Humans , Programmed Cell Death 1 Receptor , Neoplasm Recurrence, Local/drug therapy , Antibodies, Monoclonal/adverse effects
4.
Front Immunol ; 13: 848113, 2022.
Article in English | MEDLINE | ID: mdl-35967453

ABSTRACT

Cancer driven by somatic mutations may express neoantigens that can trigger T-cell immune responses. Since T-cell receptor (TCR) repertoires play critical roles in anti-tumor immune responses for oncology, next-generation sequencing (NGS) was used to profile the hypervariable complementarity-determining region 3 (CDR3) of the TCR-beta chain in peripheral blood samples from 68 gastric cancer patients and 49 healthy controls. We found that most hyper-expanded CDR3 are individual-specific, and the gene usage of TRBV3-1 is more frequent in the tumor group regardless of tumor stage than in the healthy control group. We identified 374 hyper-expanded tumor-specific CDR3, which may play a vital role in anti-tumor immune responses. The patients with stage IV gastric cancer have higher EBV-specific CDR3 abundance than the control. In conclusion, analysis of the peripheral blood TCR repertoires may provide the biomarker for gastric cancer prognosis and guide future immunotherapy.


Subject(s)
Stomach Neoplasms , Complementarity Determining Regions/genetics , Genes, T-Cell Receptor beta , Humans , Receptors, Antigen, T-Cell, alpha-beta/genetics , Stomach Neoplasms/genetics , T-Lymphocytes
5.
Gastric Cancer ; 25(5): 943-955, 2022 09.
Article in English | MEDLINE | ID: mdl-35778636

ABSTRACT

BACKGROUND: In solid tumor Phase 1/2 trials (NCT02407990; NCT04068519), tislelizumab demonstrated clinical benefit, including in advanced gastroesophageal adenocarcinoma (GEA). However, the majority of patients with GEA did not respond, highlighting the need to understand mechanisms of resistance and identify predictive biomarkers for response. METHODS: All tislelizumab-treated patients with GEA from the Phase 1/2 trials were included (N = 105). Programmed death-ligand 1 (PD-L1) expression (Tumor Area Positivity [TAP] ≥ 5%), interferon gamma (IFNγ)-related gene signature, gene expression profile, tumor mutational burden (TMB), and gene hyperamplification (HA) were analyzed for correlation with tislelizumab. RESULTS: A moderate association was observed between PD-L1 TAP ≥ 5%, IFNγ gene signature, TMB-high and efficacy. A potential correlation between hyperamplification (HA +) and worse outcomes with programmed cell death protein 1 (PD-1) inhibition was identified. Hyperamplified genes were mainly enriched in cancer progression pathways, including cell cycle and RTK-RAS-PI3K pathways. Joint PD-L1 TAP ≥ 5% and lack of hyperamplification showed the most favorable benefit with an objective response rate of 29.4%, and median progression-free survival and overall survival of 4.1 and 14.7 months, respectively. Tumors with TAP ≥ 5% and HA - had inflamed immune signatures with increased immune cell infiltration, enhanced anti-tumor cytotoxic activity and antigen presentation signatures. Findings were validated in two independent gastric and gastrointestinal cancer cohorts treated with immune checkpoint inhibitors. CONCLUSIONS: In GEA, PD-L1 positivity, IFNγ-related gene signature and TMB-high status were positively associated with tislelizumab clinical benefit, whereas HA was associated with worse clinical outcomes. Combining PD-L1 positivity and HA - may help identify patients more likely to benefit from PD-1 blockade.


Subject(s)
Adenocarcinoma , Lung Neoplasms , Stomach Neoplasms , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Antibodies, Monoclonal, Humanized , B7-H1 Antigen , Biomarkers, Tumor/genetics , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Esophageal Neoplasms , Esophagogastric Junction/pathology , Humans , Lung Neoplasms/drug therapy , Mutation , Phosphatidylinositol 3-Kinases/genetics , Programmed Cell Death 1 Receptor/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
6.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32561638

ABSTRACT

BACKGROUND: Tislelizumab is an investigational, humanized, IgG4 monoclonal antibody with high affinity and binding specificity for programmed cell death-1 (PD-1) that was engineered to minimize binding to FcγR on macrophages in order to abrogate antibody-dependent phagocytosis, a mechanism of T-cell clearance and potential resistance to anti-PD-1 therapy. METHODS: The purpose of this phase 1/2, open-label, non-comparative study was to examine the safety, tolerability, and antitumor activity of tislelizumab in adult (≥18 years) Chinese patients with histologically or cytologically confirmed advanced solid tumors with measurable disease. The phase 1 portion of the study consisted of a dose-verification study and a pharmacokinetic (PK) substudy; phase 2 was an indication-expansion study including 11 solid tumor cohorts. Patients previously treated with therapies targeting PD-1 or its ligand, programmed cell death ligand-1 were excluded. During dose-verification, dose-limiting toxicities (DLTs) were monitored; safety and tolerability were examined and the previously determined recommended phase 2 dose (RP2D) was verified. The primary endpoint of phase 2 was investigator-assessed objective response rate per Response Evaluation Criteria in Solid Tumors V.1.1. RESULTS: As of December 1, 2018, 300 patients were treated with tislelizumab 200 mg intravenously once every 3 weeks (Q3W). Median duration of follow-up was 8.1 months (range 0.2-21.9). No DLTs were reported during the phase 1 dose-verification study and the RP2D was confirmed to be 200 mg intravenously Q3W. Most treatment-related adverse events (62%) were grade 1 or 2, with the most common being anemia (n=70; 23%) and increased aspartate aminotransferase (n=67; 22%). Of the 251 efficacy evaluable patients, 45 (18%) achieved a confirmed clinical response, including one patient from the PK substudy who achieved a complete response. Median duration of response was not reached for all except the nasopharyngeal carcinoma cohort (8.3 months). Antitumor responses were observed in multiple tumor types. CONCLUSIONS: Tislelizumab was generally well tolerated among Chinese patients. Antitumor activity was observed in patients with multiple solid tumors. TRIAL REGISTRATION NUMBER: CTR20160872.


Subject(s)
Anemia/epidemiology , Antibodies, Monoclonal, Humanized/adverse effects , Drugs, Investigational/adverse effects , Immune Checkpoint Inhibitors/adverse effects , Neoplasms/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Anemia/chemically induced , Anemia/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Aspartate Aminotransferases/blood , China , Cohort Studies , Dose-Response Relationship, Drug , Drugs, Investigational/administration & dosage , Drugs, Investigational/pharmacokinetics , Female , Follow-Up Studies , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/pharmacokinetics , Male , Middle Aged , Neoplasm Staging , Neoplasms/blood , Neoplasms/diagnosis , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Response Evaluation Criteria in Solid Tumors , Young Adult
7.
Nat Commun ; 10(1): 2612, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31197141

ABSTRACT

Primary microcephaly is caused by mutations in genes encoding centrosomal proteins including WDR62 and KIF2A. However, mechanisms underlying human microcephaly remain elusive. By creating mutant mice and human cerebral organoids, here we found that WDR62 deletion resulted in a reduction in the size of mouse brains and organoids due to the disruption of neural progenitor cells (NPCs), including outer radial glia (oRG). WDR62 ablation led to retarded cilium disassembly, long cilium, and delayed cell cycle progression leading to decreased proliferation and premature differentiation of NPCs. Mechanistically, WDR62 interacts with and promotes CEP170's localization to the basal body of primary cilium, where CEP170 recruits microtubule-depolymerizing factor KIF2A to disassemble cilium. WDR62 depletion reduced KIF2A's basal body localization, and enhanced KIF2A expression partially rescued deficits in cilium length and NPC proliferation. Thus, modeling microcephaly with cerebral organoids and mice reveals a WDR62-CEP170-KIF2A pathway promoting cilium disassembly, disruption of which contributes to microcephaly.


Subject(s)
Cell Cycle Proteins/metabolism , Kinesins/metabolism , Microcephaly/pathology , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Culture Techniques , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Line , Cell Proliferation , Cilia/metabolism , Disease Models, Animal , Female , Gene Knockout Techniques , Humans , Male , Mice , Mice, Knockout , Microcephaly/genetics , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neural Stem Cells/pathology , Neuroglia/cytology , Neuroglia/pathology , Organoids/pathology , Phosphoproteins/genetics , RNA, Small Interfering/metabolism
8.
Development ; 143(22): 4127-4136, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27729407

ABSTRACT

Zika virus (ZIKV) infection of pregnant women can result in fetal brain abnormalities. It has been established that ZIKV disrupts neural progenitor cells (NPCs) and leads to embryonic microcephaly. However, the fate of other cell types in the developing brain and their contributions to ZIKV-associated brain abnormalities remain largely unknown. Using intracerebral inoculation of embryonic mouse brains, we found that ZIKV infection leads to postnatal growth restriction including microcephaly. In addition to cell cycle arrest and apoptosis of NPCs, ZIKV infection causes massive neuronal death and axonal rarefaction, which phenocopy fetal brain abnormalities in humans. Importantly, ZIKV infection leads to abnormal vascular density and diameter in the developing brain, resulting in a leaky blood-brain barrier (BBB). Massive neuronal death and BBB leakage indicate brain damage, which is further supported by extensive microglial activation and astrogliosis in virally infected brains. Global gene analyses reveal dysregulation of genes associated with immune responses in virus-infected brains. Thus, our data suggest that ZIKV triggers a strong immune response and disrupts neurovascular development, resulting in postnatal microcephaly with extensive brain damage.


Subject(s)
Brain/blood supply , Brain/embryology , Microcephaly/virology , Neovascularization, Physiologic , Neurogenesis , Zika Virus Infection/embryology , Aedes , Animals , Blood-Brain Barrier/embryology , Blood-Brain Barrier/virology , Brain/virology , Central Nervous System Vascular Malformations/embryology , Central Nervous System Vascular Malformations/virology , Chlorocebus aethiops , Disease Models, Animal , Female , Fetal Growth Retardation/virology , Mice , Mice, Inbred C57BL , Microcephaly/embryology , Nervous System Malformations/embryology , Nervous System Malformations/virology , Neural Stem Cells/physiology , Neural Stem Cells/virology , Neurogenesis/physiology , Pregnancy , Vero Cells , Zika Virus/physiology
9.
Dev Biol ; 408(1): 140-50, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26441343

ABSTRACT

How neural progenitor cell (NPC) behaviors are temporally controlled in early developing embryos remains undefined. The in vivo functions of microRNAs (miRNAs) in early mammalian development remain largely unknown. Mir-302/367 is a miRNA cluster that encodes miR-367 and four miR-302 members (miR302a-d). We show that miR-302b is highly expressed in early neuroepithelium and its expression decline as development progresses. We generated a mir-302/367 knockout mouse model and found that deletion of mir-302/367 results in an early embryonic lethality and open neural tube defect (NTD). NPCs exhibit enhanced proliferation, precocious differentiation, and decreased cell survival in mutant embryos. Furthermore, we identified Fgf15, Cyclin D1, and D2 as direct targets of miR-302 in NPCs in vivo, and their expression is enhanced in mutant NPCs. Ectopic expression of Cyclin D1 and D2 increases NPC proliferation, while FGF19 (human ortholog of Fgf15) overexpression leads to an increase of NPC differentiation. Thus, these findings reveal essential roles of miR-302/367 in orchestrating gene expression and NPC behaviors in neurulation; they also point to miRNAs as critical genetic components associated with neural tube formation.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurulation/genetics , Animals , Apoptosis , Base Sequence , Cell Proliferation/genetics , Cell Survival/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin D2/genetics , Cyclin D2/metabolism , Embryo Loss/genetics , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Deletion , Gene Expression Regulation , Mice, Knockout , MicroRNAs/genetics , Molecular Sequence Data , Neural Tube Defects/genetics , Time Factors
10.
Development ; 142(9): 1616-27, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25922525

ABSTRACT

Neural progenitor cells (NPCs) have distinct proliferation capacities at different stages of brain development. Lin28 is an RNA-binding protein with two homologs in mice: Lin28a and Lin28b. Here we show that Lin28a/b are enriched in early NPCs and their expression declines during neural differentiation. Lin28a single-knockout mice show reduced NPC proliferation, enhanced cell cycle exit and a smaller brain, whereas mice lacking both Lin28a alleles and one Lin28b allele display similar but more severe phenotypes. Ectopic expression of Lin28a in mice results in increased NPC proliferation, NPC numbers and brain size. Mechanistically, Lin28a physically and functionally interacts with Imp1 (Igf2bp1) and regulates Igf2-mTOR signaling. The function of Lin28a/b in NPCs could be attributed, at least in part, to the regulation of their mRNA targets that encode Igf1r and Hmga2. Thus, Lin28a and Lin28b have overlapping functions in temporally regulating NPC proliferation during early brain development.


Subject(s)
Brain/embryology , Cell Proliferation/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Neural Stem Cells/physiology , RNA-Binding Proteins/metabolism , Animals , Brain/cytology , Bromodeoxyuridine , DNA-Binding Proteins/genetics , Electroporation , Gene Expression Regulation, Developmental/genetics , HMGA2 Protein/metabolism , Immunoprecipitation , Mice , Mice, Knockout , RNA-Binding Proteins/genetics , Real-Time Polymerase Chain Reaction
11.
IET Syst Biol ; 8(2): 33-40, 2014 Apr.
Article in English | MEDLINE | ID: mdl-25014223

ABSTRACT

Detecting associations between human genetic variants and their phenotypic effects is a significant problem in understanding genetic bases of human-inherited diseases. The focus is on a typical type of genetic variants called non-synonymous single nucleotide polymorphisms (nsSNPs), whose occurrence may potentially alter the structures of proteins, affecting functions of proteins, and thereby causing diseases. Most of the existing methods predict associations between nsSNPs and diseases based on features derived from only protein sequence and/or structure information, and give no information about which specific disease an nsSNP is associated with. To cope with these problems, the identification of nsSNPs that are associated with a specific disease from a set of candidate nsSNPs as a binary classification problem has been formulated. A new approach has been adopted for predicting associations between nsSNPs and diseases based on multiple nsSNP similarity networks and disease phenotype similarity networks. With a series of comprehensive validation experiments, it has been demonstrated that the proposed method is effective in both recovering the nsSNP-disease associations and inferring suspect disease-associated nsSNPs for both diseases with known genetic bases and diseases of unknown genetic bases.


Subject(s)
Computational Biology/methods , Genetic Diseases, Inborn/genetics , Polymorphism, Single Nucleotide , Algorithms , Area Under Curve , Binding Sites , Genetic Predisposition to Disease , Genetic Variation , Humans , Mutation , Phenotype , ROC Curve , Software
14.
Nucleic Acids Res ; 42(10): 6183-95, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24711369

ABSTRACT

In Neurospora crassa, the methionine synthase gene met-8 plays a key role in methionine synthesis. In this study, we found that MET-8 protein levels were compromised in several mutants defective in proper heterochromatin formation. Bioinformatics analysis revealed a 50-kb AT-rich region adjacent to the met-8 promoter. ChIP assays confirmed that trimethylated H3K9 was enriched in this region, indicating that heterochromatin may form upstream of met-8. In an H3K9R mutant strain, the output of met-8 was dramatically reduced, similar to what we observed in mutant strains that had defective heterochromatin formation. Furthermore, the production of ectopically expressed met-8 at the his-3 locus in the absence of a normal heterochromatin environment was inefficient, whereas ectopic expression of met-8 downstream of two other heterochromatin domains was efficient. In addition, our data show that the expression of mig-6 was also controlled by an upstream 4.2-kb AT-rich region similar to that of the met-8 gene, and we demonstrate that the AT-rich regions adjacent to met-8 or mig-6 are required for their peak expression. Our study indicates that met-8 and mig-6 may represent a novel type of gene, whose expression relies on the proper formation of a nearby heterochromatin region.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Heterochromatin/metabolism , Neurospora crassa/genetics , 5' Untranslated Regions , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Fungal Proteins/metabolism , Gene Deletion , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Neurospora crassa/enzymology , Neurospora crassa/metabolism , RNA Polymerase II/metabolism
15.
J Biol Chem ; 285(7): 4355-65, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-19948733

ABSTRACT

DNA methylation and H3K9 trimethylation are involved in gene silencing and heterochromatin assembly in mammals and fungi. In the filamentous fungus Neurospora crassa, it has been demonstrated that H3K9 trimethylation catalyzed by histone methyltransferase DIM-5 is essential for DNA methylation. Trimethylated H3K9 is recognized by HP1, which then recruits the DNA methyltransferase DIM-2 to methylate the DNA. Here, we show that in Neurospora, ubiquitin ligase components Cullin4 and DDB1 are essential for DNA methylation. These proteins regulate DNA methylation through their effects on the trimethylation of histone H3K9. In addition, we showed that the E3 ligase activity of the Cul4-based ubiquitin ligase is required for its function in histone H3K9 trimethylation in Neurospora. Furthermore, we demonstrated that Cul4 and DDB1 are associated with the histone methyltransferase DIM-5 protein in vivo. Together, these results suggest a mechanism for DNA methylation control that may be applicable in other eukaryotic organisms.


Subject(s)
Cullin Proteins/metabolism , DNA Methylation/physiology , DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Neurospora crassa/metabolism , Blotting, Western , Chromatin Immunoprecipitation , Cullin Proteins/genetics , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Fungal Proteins/genetics , Histones/genetics , Histones/metabolism , Mutagenesis , Neurospora crassa/genetics , Polymerase Chain Reaction , Protein Binding/genetics , Protein Binding/physiology
16.
Chemosphere ; 72(1): 122-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18191174

ABSTRACT

To evaluate the treatment capability of subsurface flow constructed wetland (SFCW) and the effect of salinity on the degradation of atrazine, the degradation of atrazine in SFCW was studied. Under the static condition, the degradation of atrazine in SFCW followed first-order kinetics: c=0.09679 exp(-0.0396t) (c, residue concentration, mg l(-1); t, retention time, d), with a half-life of approximately 17.5 days. The atrazine degradation kinetic functions were established for salinities of 1.5, 3.0, 5.0, 10.0 and 15.0 g l(-1), respectively, which appeared to approach first-order kinetics. The effect of salinity on the atrazine treatment efficiency showed an exponential inhibition: lnk=3.204+0.04991 C (k, degradation constant; C, NaCl concentration, mg l(-1)). The attenuation of atrazine in SFCW cannot be a result of hydrolysis or sorption process. It was considered that some bacteria in the wetland system degraded atrazine into deethylatrazine (DEA) and deisopropylatrazine (DIA) and sequentially into CO(2) and H(2)O. Salinity impacted on the growth of bacteria resulting in a switch of the microbial community. With the increase of salinity, Shannon-Wiener Diversity Index in the SFCW system declined. The relationship between atrazine degradation constant (k) and Shannon Index was established as shown in linear phase, y=-0.07286+0.0363x. The positive correlation between them indicated that microbial community played an important role in the atrazine degradation process.


Subject(s)
Atrazine/chemistry , Sodium Chloride/chemistry , Wetlands , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Half-Life , Kinetics , Polymerase Chain Reaction
17.
Bioresour Technol ; 99(11): 4990-6, 2008 Jul.
Article in English | MEDLINE | ID: mdl-17964141

ABSTRACT

The fate of dissolved organic matter (DOM) during horizontal subsurface-flow constructed wetlands (HSSF CWs) was examined. In several studies it had been demonstrated that factors such as vegetation and substrates type affected the treatment efficiency of DOM, while very few studies discerned their influence on the transformations of DOM. Thus three pilot-scale HSSF CWs, i.e. reed (Phragmites australis)/gravel bed (W1), hybrid vegetation{cattail (Typha latifolia), bulrush (Scirpus validus), reed}/gravel bed (W2) and reed/hybrid substrates bed (gravel, zeolite, slag) (W3), were designed, and were operated continuously to investigate soluble COD (SCOD) removal and DOM transformations affected by vegetation and substrate type, and to explore the correlation between SCOD and biodiversity. The results showed that cattail and bulrush contributed to higher SCOD removal than common reed, and that gravel, zeolite and slag did not show significant influence on SCOD removal. The composition of the dissolved organic carbon (DOC) could undergo a considerable shift in composition due to metabolism and senescence from plant and microorganism. Nonlabile aromatic hydrocarbons and alkyl hydrocarbons in the effluent were a significant portion compared with labile alcoholic and alkene in the influent. It was also observed that the type of vegetation and substrate had great influence on the structure of bacteria, and the Shannon-Wiener Index increased linearly with the decrease of SCOD concentration along water flow in W2 and W3 (R2=0.96).


Subject(s)
Organic Chemicals/isolation & purification , Plants/metabolism , Wetlands , Bacteria , Biodegradation, Environmental , Carbon/metabolism , DNA, Ribosomal/analysis , Electrophoresis, Agar Gel , Molecular Weight , Oxygen/metabolism , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...