Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Ann Transl Med ; 7(18): 472, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31700908

ABSTRACT

BACKGROUND: Laminectomy and laminoplasty are popularly used in posterior cervical spine surgery but still have involved complications. We aimed to compare the clinical outcomes of microscope-assisted extensor muscle-preserving laminectomy (MA-EMPL) and open-door laminoplasty (ODLP) in treating multilevel cervical spondylotic myelopathy (MCSM). METHODS: A prospective study was designed to enroll twenty patients with MCSM underwent MA-EMPL, and recruit twenty-four patients with MCSM received ODLP (control). Radiographic measurements, outcome indicators including Japanese Orthopedic Association (JOA) score and visual analogue score (VAS) were used to evaluate technical effectiveness. Surgical complications were documented to assess technical safety. RESULTS: Postoperative cervical curvature index and range of neck motion (ROM) were not significantly changed except ROM in ODLP group. Postoperative JOA score and VAS in both groups showed improvements at final follow-up. There was no statistical difference in postoperative neurological recovery rates between two groups (67.6%±17.8% vs. 70.15%±19.6%, P=0.632). However, VAS was significantly lower at postoperative 1 month in MA-EMPL group compared with ODLP group (P<0.001). The incidences of C5 palsy were 0 vs. 16.7% between MA-EMPL group and ODLP group. There was no axial symptom occurred in MA-EMPL group while six patients in ODLP group (0 vs. 25%, P=0.049). In addition, the mean blood loss and hospital stay were lesser in MA-EMPL group compared with ODLP group (P<0.001, P=0.002, respectively). CONCLUSIONS: MA-EMPL is an effective, safe and minimally invasive method in treatment of MCSM. Compared with ODLP, MA-EMPL has advantage to decrease intraoperative blood loss, hospital stay, postoperative VAS and axial symptom, as well as preserve postoperative ROM.

2.
Blood ; 132(18): 1963-1973, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30150205

ABSTRACT

Long noncoding RNAs (lncRNAs) are increasingly being appreciated as participants in regulation of important cellular processes, including transcription. Because lncRNAs are highly cell type specific, they have the potential to contribute to the unique transcriptional repertoire of diverse cells, but underlying mechanisms are unclear. We studied BGLT3, an erythroid lncRNA encoded downstream of Aγ-globin (HBG1). BGLT3 and γ-globin genes are dynamically cotranscribed in erythroid cells in vivo. Deletion of BGLT3 using CRISPR/Cas9 editing shows that it specifically contributes to regulation of γ-globin genes. We used reduction or overexpression of the RNA and inhibition of transcription through the locus by CRISPRi to distinguish functions of the transcript vs the underlying sequence. Transcription of the BGLT3 locus is critical for looping between the γ-globin genes and BGLT3 sequences. In contrast, the BGLT3 transcript is dispensable for γ-globin/BGLT3 looping but interacts with the mediator complex on chromatin. Manipulation of the BGLT3 locus does not compromise γ-globin gene long-range looping interactions with the ß-globin locus control region (LCR). These data reveal that BGLT3 regulates γ-globin transcription in a developmental stage-specific fashion together with the LCR by serving as a separate means to increase RNA Pol II density at the γ-globin promoters.


Subject(s)
Locus Control Region , RNA, Long Noncoding/genetics , gamma-Globins/genetics , Animals , CRISPR-Cas Systems , Cell Line , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Mice, Transgenic
3.
Mol Cells ; 41(7): 695-702, 2018 Jul 31.
Article in English | MEDLINE | ID: mdl-30008200

ABSTRACT

The inner ear is a complex sensory organ responsible for hearing and balance. Formation of the inner ear is dependent on tight regulation of spatial and temporal expression of genes that direct a series of developmental processes. Recently, epigenetic regulation has emerged as a crucial regulator of the development of various organs. However, what roles higher-order chromatin organization and its regulator molecules play in inner ear development are unclear. CCCTC-binding factor (CTCF) is a highly conserved 11-zinc finger protein that regulates the three-dimensional architecture of chromatin, and is involved in various gene regulation processes. To delineate the role of CTCF in inner ear development, the present study investigated inner ear-specific Ctcf knockout mouse embryos (Pax2-Cre; Ctcffl/fl ). The loss of Ctcf resulted in multiple defects of inner ear development and severely compromised otic neurogenesis, which was partly due to a loss of Neurog1 expression. Furthermore, reduced Neurog1 gene expression by CTCF knockdown was found to be associated with changes in histone modification at the gene's promoter, as well as its upstream enhancer. The results of the present study demonstrate that CTCF plays an essential role in otic neurogenesis by modulating histone modification in the Neurog1 locus.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , CCCTC-Binding Factor/metabolism , Ear, Inner/innervation , Genetic Loci , Histones/metabolism , Nerve Tissue Proteins/genetics , Neurogenesis , Protein Processing, Post-Translational , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Ear, Inner/embryology , Ear, Inner/pathology , Embryo, Mammalian/metabolism , Gene Deletion , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Lysine/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Protein Processing, Post-Translational/drug effects , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Tretinoin/pharmacology
4.
Retina ; 38(2): 253-262, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28141749

ABSTRACT

PURPOSE: To determine the repeatability of measuring the thickness of the central macula, retinal nerve fiber layer, and ganglion cell-inner plexiform layer (GC-IPL) using spectral domain optical coherence tomography (Cirrus HD-OCT) in eyes with age-related macular degeneration. METHODS: One hundred and thirty-four eyes were included. The measurement repeatability was assessed by an experienced examiner who performed two consecutive measurements using a 512 × 128 macular cube scan and a 200 × 200 optic disk cube scan. To assess changes in macular morphology in patients with age-related macular degeneration, the patients were divided into the following three groups according to the central macular thickness (CMT): A group, CMT < 200 µm; B group, 200 µm ≤ CMT < 300 µm; and C group, CMT > 300 µm. RESULTS: Measurement repeatability was assessed using test-retest variability, a coefficient of variation, and an intraclass correlation coefficient. The mean measurement repeatability for the central macular, retinal nerve fiber layer, and GC-IPL thickness was high in the B group. The mean measurement repeatability for both the central macula and retinal nerve fiber layer thickness was high in the A and C groups, but was lower for the GC-IPL thickness. The measurement repeatability for GC-IPL thickness was high in the B group, but low in the A group and in the C group. CONCLUSION: The automated measurement repeatability for GC-IPL thickness was significantly lower in patients with age-related macular degeneration with out of normal CMT range. The effect of changes in macular morphology should be considered when analyzing GC-IPL thicknesses in a variety of ocular diseases.


Subject(s)
Macula Lutea/pathology , Nerve Fibers/pathology , Optic Disk/pathology , Retinal Ganglion Cells/pathology , Tomography, Optical Coherence/methods , Visual Acuity , Wet Macular Degeneration/diagnosis , Aged , Disease Progression , Female , Follow-Up Studies , Humans , Male , Prospective Studies , Time Factors , Wet Macular Degeneration/physiopathology
5.
Gastroenterology ; 154(4): 1047-1060, 2018 03.
Article in English | MEDLINE | ID: mdl-29229400

ABSTRACT

BACKGROUND AND AIMS: CD4+CD25+Foxp3+ T-regulatory (Treg) cells control immune responses and maintain immune homeostasis. However, under inflammatory conditions, Treg cells produce cytokines that promote inflammation. We investigated production of tumor necrosis factor (TNF) by Treg cells in patients with acute hepatitis A (AHA), and examined the characteristics of these cells and association with clinical factors. METHODS: We analyzed blood samples collected from 63 patients with AHA at the time of hospitalization (and some at later time points) and 19 healthy donors in South Korea. Liver tissues were collected from patients with fulminant AHA during liver transplantation. Peripheral blood mononuclear cells were isolated from whole blood and lymphocytes were isolated from liver tissues and analyzed by flow cytometry. Cytokine production from Treg cells (CD4+CD25+Foxp3+) was measured by immunofluorescence levels following stimulation with anti-CD3 and anti-CD28. Epigenetic stability of Treg cells was determined based on DNA methylation patterns. Phenotypes of Treg cells were analyzed by flow cytometry and an RORγt inhibitor, ML-209, was used to inhibit TNF production. Treg cell suppression assay was performed by co-culture of Treg-depleted peripheral blood mononuclear cells s and isolated Treg cells. RESULTS: A higher proportion of CD4+CD25+Foxp3+ Treg cells from patients with AHA compared with controls produced TNF upon stimulation with anti-CD3 and anti-CD28 (11.2% vs 2.8%). DNA methylation analysis confirmed the identity of the Treg cells. TNF-producing Treg cells had features of T-helper 17 cells, including up-regulation of RORγt, which was required for TNF production. The Treg cells had reduced suppressive functions compared with Treg cells from controls. The frequency of TNF-producing Treg cells in AHA patients' blood correlated with their serum level of alanine aminotransferase. CONCLUSIONS: Treg cells from patients with AHA have altered functions compared with Treg cells from healthy individuals. Treg cells from patients with AHA produce higher levels of TNF, gain features of T-helper 17 cells, and have reduced suppressive activity. The presence of these cells is associated with severe liver injury in patients with AHA.


Subject(s)
Hepatitis A/metabolism , Liver/metabolism , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acute Disease , Antigens, CD/immunology , Antigens, CD/metabolism , Apyrase/immunology , Apyrase/metabolism , Case-Control Studies , Cells, Cultured , DNA Methylation , Epigenesis, Genetic , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Hepatitis A/diagnosis , Hepatitis A/immunology , Hepatitis A/virology , Hepatitis A virus/immunology , Hepatitis A virus/pathogenicity , Host-Pathogen Interactions , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Liver/immunology , Liver/pathology , Liver/virology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Severity of Illness Index , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/virology , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/virology , Time Factors , Tumor Necrosis Factor-alpha/immunology
6.
Cell Rep ; 19(12): 2490-2502, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636938

ABSTRACT

Lineage-specific transcription factors are critical for long-range enhancer interactions, but direct or indirect contributions of architectural proteins such as CCCTC-binding factor (CTCF) to enhancer function remain less clear. The LDB1 complex mediates enhancer-gene interactions at the ß-globin locus through LDB1 self-interaction. We find that an LDB1-bound enhancer upstream of carbonic anhydrase 2 (Car2) activates its expression by interacting directly with CTCF at the gene promoter. Both LDB1 and CTCF are required for enhancer-Car2 looping, and the domain of LDB1 contacted by CTCF is necessary to rescue Car2 transcription in LDB1-deficient cells. Genome-wide studies and CRISPR/Cas9 genome editing indicate that LDB1-CTCF enhancer looping underlies activation of a substantial fraction of erythroid genes. Our results provide a mechanism by which long-range interactions of architectural protein CTCF can be tailored to achieve a tissue-restricted pattern of chromatin loops and gene expression.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA-Binding Proteins/physiology , Enhancer Elements, Genetic , Erythroid Cells/metabolism , LIM Domain Proteins/physiology , Animals , Cell Lineage , Gene Expression , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Protein Binding , Protein Interaction Domains and Motifs
7.
BMB Rep ; 49(10): 578-583, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27697109

ABSTRACT

Special AT-rich sequence binding protein 1 (SATB1) is a nuclear matrix-associated DNA-binding protein that functions as a chromatin organizer. SATB1 is highly expressed in aggressive breast cancer cells and promotes growth and metastasis by reprograming gene expression. Through genomewide cross-examination of gene expression and histone methylation, we identified SATB1 target genes for which expression is associated with altered epigenetic marks. Among the identified genes, long noncoding RNA urothelial carcinoma-associated 1 (UCA1) was upregulated by SATB1 depletion. Upregulation of UCA1 coincided with increased H3K4 trimethylation (H3K4me3) levels and decreased H3K27 trimethylation (H3K27me3) levels. Our study showed that SATB1 binds to the upstream region of UCA1 in vivo, and that its promoter activity increases with SATB1 depletion. Furthermore, simultaneous depletion of SATB1 and UCA1 potentiated suppression of tumor growth and cell survival. Thus, SATB1 repressed the expression of oncogenic UCA1, suppressing growth and survival of breast cancer cells. [BMB Reports 2016; 49(10): 578-583].


Subject(s)
Epigenesis, Genetic , Matrix Attachment Region Binding Proteins/metabolism , RNA, Long Noncoding/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Flow Cytometry , Genes, Reporter , Histones/metabolism , Humans , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Methylation , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Up-Regulation
8.
J Immunol ; 196(10): 4378-89, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27067007

ABSTRACT

IL-21, a pleiotropic cytokine strongly linked with autoimmunity and inflammation, regulates diverse immune responses. IL-21 can be potently induced in CD4(+) T cells by IL-6; however, very little is known about the mechanisms underlying the transcriptional regulation of the Il21 gene at the chromatin level. In this study, we demonstrated that a conserved noncoding sequence located 49 kb upstream of the Il21 gene contains an enhancer element that can upregulate Il21 gene expression in a STAT3- and NFAT-dependent manner. Additionally, we identified enhancer-blocking insulator elements in the Il21 locus, which constitutively bind CTCF and cohesin. In naive CD4(+) T cells, these upstream and downstream CTCF binding sites interact with each other to make a DNA loop; however, the Il21 promoter does not interact with any cis-elements in the Il21 locus. In contrast, stimulation of CD4(+) T cells with IL-6 leads to recruitment of STAT3 to the promoter and novel distal enhancer region. This induces dynamic changes in chromatin configuration, bringing the promoter and the regulatory elements in close spatial proximity. The long-range interaction between the promoter and distal enhancer region was dependent on IL-6/STAT3 signaling pathway but was disrupted in regulatory T cells, where IL-21 expression was repressed. Thus, our work uncovers a novel topological chromatin framework underlying proper transcriptional regulation of the Il21 gene.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Chromatin/genetics , DNA-Binding Proteins/genetics , Interleukins/genetics , Animals , Base Sequence , CCCTC-Binding Factor , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Conserved Sequence , Enhancer Elements, Genetic , Gene Expression Regulation , Humans , Interleukin-6/pharmacology , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Cohesins
9.
Korean J Spine ; 12(2): 95-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26217391

ABSTRACT

Fibrous dysplasia of the spine is very rarely observed. We reported a case of a 57-year-old woman, who presented with neck and bilateral shoulder pain with histologically confirmed fibrous dysplasia, involving the first and second thoracic vertebrae. Clinical and radiological findings were not specific for fibrous dysplasia. The histological biopsy was required for a confirmed diagnosis. Endocrine and metabolic evaluations are required to rule out diseases such as hyperthyroidism, Cushing syndrome and osteomalacia. Fibrous dyplasia can be managed by appropriate medical and surgical treatments based on the patient's neurological status and symptoms. Our patient was given intravenous pamidronate 60mg/day for 3 days. After 9 months, her initial symptoms were improved, but computed tomography scan of the thoracic spine showed no change of the lesions.

10.
J Allergy Clin Immunol ; 136(3): 713-24, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25936568

ABSTRACT

BACKGROUND: Langerhans cells (LCs) are skin-resident dendritic cells (DCs) that orchestrate skin immunity. CCCTC-binding factor (CTCF) is a highly conserved DNA-binding protein that regulates higher-order chromatin organization and is involved in various gene regulation processes. OBJECTIVE: We sought to clarify a possible role for CTCF in LC homeostasis and function in vivo. METHODS: We used a conditional gene deletion mouse system to generate DC- and LC-specific CTCF-ablated mice. Short hairpin RNA-mediated RNA interference was used to silence CTCF expression in human monocyte-derived Langerhans cells. DC populations were assessed by using flow cytometry and immunofluorescence. Gene expression arrays were performed to identify genes regulated by CTCF in LCs. Contact hypersensitivity and epicutaneous sensitization responses were measured to examine the functional significance of CTCF ablation. RESULTS: DC-specific CTCF deletion led to a reduced pool of systemic DCs, with LCs most severely affected. Decreases in epidermal LC numbers were specifically associated with self-turnover defects. Interestingly, CTCF-deficient LCs demonstrated impaired migration out of the epidermis. Whole-transcriptome analyses revealed that genes that promoted cell adhesion were highly expressed, but CCR7 was downregulated in CTCF-depleted LCs. Hapten-induced contact hypersensitivity responses were more sustained in LC-specific CTCF-deficient mice, whereas epicutaneous sensitization to protein antigen was attenuated, indicating that CTCF-dependent LC homeostasis is required for optimal immune function of LCs in a context-dependent manner. CONCLUSION: Our results show that CTCF positively regulates the homeostatic pool and the efficient emigration of LCs, which are required for modulating the functional immune network of the skin.


Subject(s)
Dermatitis, Contact/genetics , Homeostasis/genetics , Langerhans Cells/metabolism , Repressor Proteins/genetics , Animals , CCCTC-Binding Factor , Cell Adhesion , Cell Movement/genetics , Cell Movement/immunology , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Epidermis/immunology , Epidermis/metabolism , Epidermis/pathology , Gene Expression Profiling , Gene Expression Regulation , Haptens , Homeostasis/immunology , Humans , Langerhans Cells/immunology , Langerhans Cells/pathology , Mice , Mice, Knockout , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/deficiency , Repressor Proteins/immunology , Signal Transduction
11.
BMB Rep ; 48(2): 103-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24965578

ABSTRACT

Trichomoniasis caused by the parasitic protozoan Trichomonas vaginalis is the most common sexually transmitted disease in the world. Dendritic cells are antigen presenting cells that initiate immune responses by directing the activation and differentiation of naïve T cells. In this study, we analyzed the effect of Trichomonas vaginalis-derived Secretory Products on the differentiation and function of dendritic cells. Differentiation of bone marrow-derived dendritic cells in the presence of T. vaginalis-derived Secretory Products resulted in inhibition of lipopolysaccharide-induced maturation of dendritic cells, down-regulation of IL-12, and up-regulation of IL-10. The protein components of T. vaginalis-derived Secretory Products were shown to be responsible for altered function of bone marrow- derived dendritic cells. Chromatin immunoprecipitation assay demonstrated that IL-12 expression was regulated at the chromatin level in T. vaginalis-derived Secretory Productstreated dendritic cells. Our results demonstrated that T. vaginalis- derived Secretory Products modulate the maturation and cytokine production of dendritic cells leading to immune tolerance.


Subject(s)
Dendritic Cells/cytology , Trichomonas vaginalis/metabolism , Animals , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Antigens, Protozoan/pharmacology , Bone Marrow Cells/cytology , Cell Differentiation , Chromatin Immunoprecipitation , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Down-Regulation/drug effects , Endopeptidase K/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-10 , Interleukin-12 , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Up-Regulation/drug effects
12.
Mol Cells ; 36(4): 368-75, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23996530

ABSTRACT

ABCG2 is a member of the ATP binding cassette (ABC) transmembrane proteins that plays an important role in stem cell biology and drug resistance of cancer cells. In this study, we investigated how expression of human ABCG2 gene is regulated in lung cancer A549 cells. Binding of Sp1 and Sp3 transcription factors to the ABCG2 promoter in vitro and in vivo was elucidated by electrophoretic mobility shift assay and chromatin immunoprecipitation assay. The ABCG2 promoter activity was impaired when Sp1 sites were mutated but was enhanced by overexpression of Sp1 or Sp3 proteins. Knockdown of Sp1 or Sp3 expression by short interfering RNA significantly decreased the expression of ABCG2 mRNA and protein, resulting in attenuated formation of the side population in A549 cells. In addition, Sp1 inhibition in vivo by mithramycin A suppressed the percentage of the side population fraction and sphere forming activities of A549 cells. Moreover, inhibiting Sp1- or Sp3-dependent ABCG2 expression caused chemosensitization to the anticancer drug cisplatin. Collectively, our results demonstrate that Sp1 and Sp3 transcription factors are the primary determinants for activating basal transcription of the ABCG2 gene and play an important role in maintaining the side population phenotype of lung cancer cells.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Binding Sites , Cell Line , Cisplatin/pharmacology , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Promoter Regions, Genetic , Side-Population Cells/physiology , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics
13.
Acta Otolaryngol ; 133(8): 866-73, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23647464

ABSTRACT

CONCLUSION: The oncological and functional outcomes of hypopharyngeal cancer after conservative laryngeal surgery are fairly acceptable, making this a reasonable initial treatment option for selected patients. OBJECTIVE: The purpose of this study was to assess the clinical outcomes of patients with hypopharyngeal squamous cell carcinoma (SCC) treated with conservative laryngeal surgery with partial pharyngectomy. METHODS: Fifty-eight patients with hypopharyngeal SCC who underwent laryngeal preservation surgery were enrolled. The tumors were classified as cT1 in 5 (8.6%) patients, cT2 in 35 (60.3%), cT3 in 14 (24.1%), and cT4a in 4 (6.9%) patients. RESULTS: Surgical outcomes: 5-year overall and disease-specific survival rates were 78% and 77.6%. Recurrent disease developed in 13 patients (22.4%). Multivariate analysis revealed that level VI metastasis confirmed by histopathological analysis, close (< 5 mm) histologic margin, advanced N stage, and posterior pharyngeal wall tumor were independent factors associated with poor disease-specific survival. Functional outcomes: 50 patients (86.2%) could obtain all their nutritional needs orally. Eight patients needed the assistance of a percutaneous endoscopic gastrostomy tube. Oral re-alimentation was achieved within a mean of 26.1 days after surgery. Fifty-one patients (87.9%) could be decannulated after a mean of 43.8 days postoperatively.


Subject(s)
Carcinoma, Squamous Cell/surgery , Hypopharyngeal Neoplasms/surgery , Pharyngectomy , Adult , Aged , Carcinoma, Squamous Cell/pathology , Female , Humans , Hypopharyngeal Neoplasms/pathology , Lymphatic Metastasis , Male , Middle Aged , Pharynx/pathology , Retrospective Studies , Treatment Outcome
14.
Cell ; 149(6): 1233-44, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682246

ABSTRACT

Chromatin loops juxtapose distal enhancers with active promoters, but their molecular architecture and relationship with transcription remain unclear. In erythroid cells, the locus control region (LCR) and ß-globin promoter form a chromatin loop that requires transcription factor GATA1 and the associated molecule Ldb1. We employed artificial zinc fingers (ZF) to tether Ldb1 to the ß-globin promoter in GATA1 null erythroblasts, in which the ß-globin locus is relaxed and inactive. Remarkably, targeting Ldb1 or only its self-association domain to the ß-globin promoter substantially activated ß-globin transcription in the absence of GATA1. Promoter-tethered Ldb1 interacted with endogenous Ldb1 complexes at the LCR to form a chromatin loop, causing recruitment and phosphorylation of RNA polymerase II. ZF-Ldb1 proteins were inactive at alleles lacking the LCR, demonstrating that their activities depend on long-range interactions. Our findings establish Ldb1 as a critical effector of GATA1-mediated loop formation and indicate that chromatin looping causally underlies gene regulation.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , LIM Domain Proteins/metabolism , Transcription, Genetic , beta-Globins/genetics , Animals , Cell Line , Cell Separation , DNA-Binding Proteins/chemistry , Embryo, Mammalian/cytology , Erythroblasts/metabolism , Female , GATA1 Transcription Factor/metabolism , Gene Expression Regulation , LIM Domain Proteins/chemistry , Male , Mice , Promoter Regions, Genetic , Protein Structure, Tertiary , Zinc Fingers
15.
Toxicology ; 296(1-3): 73-82, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22484357

ABSTRACT

Bisphenol A (BPA) is used as a monomer during the manufacture of polycarbonate plastics and epoxy resins. However, BPA adversely affects reproductive organ growth and development, and it has been proposed that the detrimental effects of BPA could extend to future generations. The present study was conducted to evaluate the transgenerational effects of BPA on hippocampal neurogenesis and neurocognitive function. Pregnant female C57BL/6 mice (F0) were exposed to BPA (0.1-10 mg/kg) from gestation day 6 to 17, and female offspring (F2) from F1 generation mice were prepared. It was found that exposure of F0 mice to BPA at 10 mg/kg decreased the number of newly generated cells in the hippocampi of F2 female mice. Passive avoidance testing revealed that high-doses BPA (1 mg/kg and 10 mg/kg) decreased cross-over latency time in F2 mice, suggesting a BPA-mediated neurocognitive deficit in terms of memory retention. Furthermore, it was found that levels of phospho-ERK, brain-derived neurotrophic factor (BDNF), and phospho-CREB in hippocampi were significantly lower in F2 mice. Interestingly, the effects of BPA on hippocampal neurogenesis were found to be correlated with altered DNA methylation. In particular, high-dose BPA exposure increased DNA methylation of the CREB regulated transcription coactivator 1 (Crtc1) generated in F2 mice. These findings suggest that BPA exposure of pregnant mothers could adversely affect hippocampal neurogenesis and cognitive function in future generations by modulating the ERK and BDNF-CREB signaling cascades.


Subject(s)
Endocrine Disruptors/toxicity , Estrogens, Non-Steroidal/toxicity , Hippocampus/drug effects , Neurogenesis/drug effects , Phenols/toxicity , Animals , Benzhydryl Compounds , Brain-Derived Neurotrophic Factor/metabolism , Cognition/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Hippocampus/cytology , Hippocampus/physiology , Learning/drug effects , Maternal-Fetal Exchange , Mice , Mice, Inbred C57BL , Pregnancy
16.
Blood ; 119(16): 3820-7, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22378846

ABSTRACT

The ß-globin locus control region (LCR) is necessary for high-level ß-globin gene transcription and differentiation-dependent relocation of the ß-globin locus from the nuclear periphery to the central nucleoplasm and to foci of hyperphosphorylated Pol II "transcription factories" (TFys). To determine the contribution of individual LCR DNaseI hypersensitive sites (HSs) to transcription and nuclear location, in the present study, we compared ß-globin gene activity and location in erythroid cells derived from mice with deletions of individual HSs, deletions of 2 HSs, and deletion of the whole LCR and found all of the HSs had a similar spectrum of activities, albeit to different degrees. Each HS acts as an independent module to activate expression in an additive manner, and this is correlated with relocation away from the nuclear periphery. In contrast, HSs have redundant activities with respect to association with TFys and the probability that an allele is actively transcribed, as measured by primary RNA transcript FISH. The limiting effect on RNA levels occurs after ß-globin genes associate with TFys, at which time HSs contribute to the amount of RNA arising from each burst of transcription by stimulating transcriptional elongation.


Subject(s)
Cell Nucleus/metabolism , Locus Control Region/genetics , Nucleoplasmins/metabolism , Transcription, Genetic/physiology , beta-Globins/genetics , Animals , Erythroid Cells/metabolism , Gene Deletion , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Transgenic , RNA, Messenger/genetics , beta-Globins/metabolism
17.
J Leukoc Biol ; 91(2): 245-57, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22045870

ABSTRACT

IL-31, a newly identified member of the IL-6 cytokine family, is involved in many pathological conditions, including atopic dermatitis and pruritis. In this study, we investigated how expression of IL-31 is regulated in T cells and mast cells. We observed that expression of IL-31 required a calcium signal and was dependent on the calcineurin-NFAT signaling pathway. Moreover, we found that IL-31 promoter contains a positive regulatory region that mediates calcium- and IL-4-dependent induction of the IL-31 gene and demonstrated that a change into an open chromatin conformation occurs in this region after stimulation with calcium and IL-4. Whereas IL-4 responsiveness required STAT6 binding sites, calcium responsiveness of IL-31 promoter required NFAT binding sites that bind NFATc1 and NFATc2 in vitro and in vivo. The induction of IL-31 promoter activity was impaired when these sites were mutated but was enhanced by CA-NFATc1 or STAT6 proteins and further increased synergistically by combinations of both proteins. Furthermore, the importance of STAT6 proteins was indicated by impaired, IL-4-mediated induction of IL-31 in STAT6-diminished Jurkat cells. Thus, our data demonstrate that calcium and IL-4 signals are required to mediate induction of IL-31 in Th2 cells and mast cells and that this induction appears to result from specific binding of NFAT and STAT6 proteins.


Subject(s)
Interleukins/genetics , NFATC Transcription Factors/physiology , STAT6 Transcription Factor/physiology , Animals , Base Sequence , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Calcium/physiology , Dinitrophenols/pharmacology , HEK293 Cells/drug effects , HEK293 Cells/metabolism , Humans , Interleukin-4/pharmacology , Interleukins/biosynthesis , Ionomycin/pharmacology , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Promoter Regions, Genetic , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Regulatory Sequences, Nucleic Acid , Sequence Alignment , Sequence Homology, Nucleic Acid , Serum Albumin/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic
18.
Blood ; 118(23): 6200-8, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22010104

ABSTRACT

The Ldb1/GATA-1/TAL1/LMO2 complex mediates long-range interaction between the ß-globin locus control region (LCR) and gene in adult mouse erythroid cells, but whether this complex mediates chromatin interactions at other developmental stages or in human cells is unknown. We investigated NLI (Ldb1 homolog) complex occupancy and chromatin conformation of the ß-globin locus in human erythroid cells. In addition to the LCR, we found robust NLI complex occupancy at a site downstream of the (A)γ-globin gene within sequences of BGL3, an intergenic RNA transcript. In cells primarily transcribing ß-globin, BGL3 is not transcribed and BGL3 sequences are occupied by NLI core complex members, together with corepressor ETO2 and by γ-globin repressor BCL11A. The LCR and ß-globin gene establish proximity in these cells. In contrast, when γ-globin transcription is reactivated in these cells, ETO2 participation in the NLI complex at BGL3 is diminished, as is BCL11A occupancy, and both BGL3 and γ-globin are transcribed. In these cells, proximity between the BGL3/γ-globin region and the LCR is established. We conclude that alternative NLI complexes mediate γ-globin transcription or silencing through long-range LCR interactions involving an intergenic site of noncoding RNA transcription and that ETO2 is critical to this process.


Subject(s)
DNA-Binding Proteins/genetics , Erythroid Cells/metabolism , LIM Domain Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , gamma-Globins/genetics , 3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Erythroid Cells/cytology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Humans , K562 Cells , LIM Domain Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Untranslated/genetics , Repressor Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/metabolism , Transcription, Genetic/physiology , Tumor Suppressor Proteins/metabolism , gamma-Globins/metabolism
19.
Biochem Biophys Res Commun ; 409(2): 222-8, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21554857

ABSTRACT

Transforming growth factor beta 1-induced (TGFBI) protein is an extracellular matrix (ECM) protein that is associated with other ECM proteins and functions as a ligand for various types of integrins. In this study, we investigated how human TGFBI expression is regulated in lung and breast cancer cells. We observed that the TGFBI promoter in A549 and MBA-MD-231 cells, which constitutively express TGFBI, existed in an open chromatin conformation associated with transcriptionally permissive histone modifications. Moreover, we found that TGFBI expression required Sp1 transcription elements that can bind transcription factors Sp1 and Sp3 in vitro. Occupancy of the TGFBI promoter by Sp1 and Sp3 in vivo was only observed in TGFBI-expressing cells, indicating that open chromatin conformation might facilitate the binding of Sp1 and Sp3 to the TGFBI promoter region. TGFBI promoter activity was impaired when Sp1 elements were mutated, but was increased when Sp1 or Sp3 factors was overexpressed. Furthermore, Sp1 inhibition in vivo by mithramycin A, as well as knockdown of Sp1 and/or Sp3 expression by short interfering RNA, significantly reduced TGFBI mRNA and protein levels. Thus, our data demonstrated that the expression of TGFBI is well correlated with chromatin conformation at the TGFBI promoter, and that factors Sp1 and Sp3 are the primary determinants for the control of constitutive expression of TGFBI gene.


Subject(s)
Chromatin/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Transforming Growth Factor beta/genetics , Cell Line , Cell Line, Tumor , Chromatin/chemistry , Gene Knockdown Techniques , Histones/metabolism , Humans , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor/genetics
20.
Biomed Chromatogr ; 25(12): 1389-94, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21387355

ABSTRACT

A liquid chromatography-tandem mass spectrometric (LC/MS/MS) method was developed for the determination of an atypical antipsychotic drug, lurasidone, in rat plasma. The method involves the addition of acetonitrile and ziprasidone (internal standard) solution to plasma samples, followed by centrifugation. An aliquot of the supernatant was diluted with water and directly injected into the LC/MS/MS system. The separations were performed on a column packed with octadecylsilica (5 µm, 2.0 × 50 mm) with 0.1% formic acid and 0.1% formic acid in acetonitrile as mobile phase and the detection was performed using tandem mass spectrometry by multiple-reaction monitoring via an electrospray ionization source. The standard curve was linear (r = 0.9982) over the concentration range 0.002-1 µg/mL. The intra- and inter-assay precisions were 1.7 and 8.6%, respectively. The accuracy range was from 90.3 to 101.8%. The lower limit of quantification was 2.0 ng/mL using 50 µL of rat plasma sample. The developed analytical method was successfully applied to the pharmacokinetic study of lurasidone in rats.


Subject(s)
Antipsychotic Agents/blood , Chromatography, High Pressure Liquid/methods , Isoindoles/blood , Tandem Mass Spectrometry/methods , Thiazoles/blood , Acetonitriles/blood , Animals , Antipsychotic Agents/pharmacokinetics , Isoindoles/pharmacokinetics , Linear Models , Lurasidone Hydrochloride , Male , Piperazines/blood , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Thiazoles/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...