Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Clin Radiol ; 78(10): e773-e781, 2023 10.
Article in English | MEDLINE | ID: mdl-37550131

ABSTRACT

AIM: To gauge current final year medical students' exposure to interventional radiology (IR)and assess their perceptions of IR as a prospective career option. MATERIALS AND METHODS: An online questionnaire comprising of questions that gauge final-year medical students' understanding of and exposure to IR based on the recommendations set out by the British Society of Interventional Radiology (BSIR), was sent out to final-year students across 34 UK medical schools. RESULTS: Five hundred and ten responses were collected from 33 out of 34 eligible medical schools. Sixty-four per cent of respondents rated their own IR knowledge as inadequate. On average, only 50% of all subtopics proposed in the BSIR undergraduate curriculum was covered during medical school and 32.7% of respondents were not exposed to any fundamental IR principles and techniques recommended by the BSIR during medical school. Regarding careers, 2.7% of respondents reported a definite interest in pursuing a career in IR. Most respondents (89.8%) felt that there was insufficient undergraduate teaching on IR and that they lacked information to consider pursuing a career in IR (87.5%). CONCLUSION: Insufficient exposure and teaching on IR throughout medical schools have led to a lack of awareness and consideration of IR as a future career choice amongst UK medical students. The re-evaluation of IR teaching in the medical school curricula is needed. In the long-term, such recommendations could provide the much-needed solution to the workforce shortages seen in IR.


Subject(s)
Education, Medical, Undergraduate , Students, Medical , Humans , Prospective Studies , Radiology, Interventional/education , Education, Medical, Undergraduate/methods , Curriculum , Surveys and Questionnaires , Career Choice
3.
Clin Radiol ; 78(5): e458-e462, 2023 05.
Article in English | MEDLINE | ID: mdl-36842914

ABSTRACT

AIM: To evaluate percutaneous ultrasound-guided day-case mucin aspiration in advanced pseudomyxoma peritonei (PMP) using a wide-bore drain with regards to its safety and efficacy. MATERIALS AND METHODS: All patients who underwent percutaneous mucin aspiration for PMP between 2019-2021 at a single national peritoneal tumour service were included in this study. Under local anaesthesia, a suction-enabled 28-32 F catheter was used for drainage following wire-guided track dilatation. The volume drained and difference in abdominal girth pre- and post-procedure were measured. Patients graded difficulty in breathing and abdominal discomfort pre- and post-procedure. Histology reports were reviewed. RESULTS: Sixteen patients received 56 percutaneous mucin aspirations between 2019-2021. The aetiology was a low-grade appendiceal mucinous neoplasm (LAMN) in 50% of patients. The mean amount of mucin drained was 7,320 ± 3,000ml (range 300-13,500 ml). The mean reduction in abdominal girth post-procedure was 12.2 ± 5 cm (range 0-27 cm). Only grade 1 complications were observed. CONCLUSION: Percutaneous ultrasound-guided day-case aspiration of mucin for advanced and recurrent PMP using a wide-bore drain is a safe and effective procedure. It may be used in the palliative setting or as a bridge to surgery in the very symptomatic patient or if there is a reversible contraindication to surgery.


Subject(s)
Adenocarcinoma, Mucinous , Pseudomyxoma Peritonei , Humans , Pseudomyxoma Peritonei/diagnostic imaging , Pseudomyxoma Peritonei/surgery , Mucins , Suction , Adenocarcinoma, Mucinous/pathology , Ultrasonography, Interventional
5.
Br J Pharmacol ; 174(10): 1002-1014, 2017 05.
Article in English | MEDLINE | ID: mdl-27590257

ABSTRACT

The peptide hormone relaxin regulates the essential maternal haemodynamic adaptations in early pregnancy through direct actions on the renal and systemic vasculature. These vascular actions of relaxin occur mainly through endothelium-derived NO-mediated vasodilator pathways and improvements in arterial compliance in small resistance-size arteries. This work catalysed a plethora of studies which revealed quite heterogeneous responses across the different regions of the vasculature, and also uncovered NO-independent mechanisms of relaxin action. In this review, we first describe the role of endogenous relaxin in maintaining normal vascular function, largely referring to work in pregnant and male relaxin-deficient animals. We then discuss the diversity of mechanisms mediating relaxin action in different vascular beds, including the involvement of prostanoids, VEGF, endothelium-derived hyperpolarisation and antioxidant activity in addition to the classic NO-mediated vasodilatory pathway. We conclude the review with current perspectives on the vascular remodelling capabilities of relaxin. LINKED ARTICLES: This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , Relaxin/metabolism , Animals , Humans
6.
Br J Pharmacol ; 173(6): 1005-17, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26660642

ABSTRACT

BACKGROUND AND PURPOSE: In the RELAX-AHF trial, a 48 h i.v. serelaxin infusion reduced systemic vascular resistance in patients with acute heart failure. Consistent with preclinical studies, serelaxin augments endothelial vasodilator function in rat mesenteric arteries. Little is known about the contribution of endothelium-derived relaxing factors after a longer duration of continuous serelaxin treatment. Here we have assessed vascular reactivity and mechanistic pathways in mesenteric arteries and veins and the aorta after 48 or 72 h continuous i.v. infusion of serelaxin. EXPERIMENTAL APPROACH: Male rats were infused with either placebo or serelaxin (13.3 µg·kg(-1) ·h(-1) ) via the jugular vein using osmotic minipumps. Vascular function was assessed using wire myography. Changes in gene and protein expression and 6-keto PGF1α levels were determined by quantitative PCR, Western blot and ELISA respectively. KEY RESULTS: Continuous i.v. serelaxin infusion augmented endothelium-dependent relaxation in arteries (mesenteric and aorta) but not in mesenteric veins. In mesenteric arteries, 48 h i.v. serelaxin infusion increased basal NOS activity, associated with increased endothelial NOS (eNOS) expression. Interestingly, phosphorylated-eNOS(Ser1177) , eNOS and basal NOS activity were reduced in mesenteric arteries following 72 h serelaxin treatment. At 72 h, serelaxin treatment improved bradykinin-mediated relaxation through COX2-derived PGI2 production. CONCLUSIONS AND IMPLICATIONS: Continuous i.v. serelaxin infusion enhanced endothelial vasodilator function in arteries but not in veins. The underlying mediator at 48 h was NO but there was a transition to PGI2 by 72 h. Activation of the PGI2 -dependent pathway is key to the prolonged vascular response to serelaxin treatment.


Subject(s)
Aorta, Abdominal/drug effects , Epoprostenol/physiology , Mesenteric Arteries/drug effects , Nitric Oxide/physiology , Relaxin/pharmacology , Animals , Aorta, Abdominal/physiology , Infusions, Intravenous , Male , Mesenteric Arteries/physiology , Mesenteric Veins/drug effects , Mesenteric Veins/physiology , Rats, Wistar , Recombinant Proteins/blood , Recombinant Proteins/pharmacology , Relaxin/blood , Signal Transduction , Vasodilation/drug effects
7.
Exp Mol Pathol ; 92(3): 287-95, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22421441

ABSTRACT

Acute respiratory distress syndrome, a severe form of acute lung injury (ALI), is a major cause of death during influenza pneumonia. We have provided evidence for the involvement of recruited neutrophils, their toxic enzymes such as myeloperoxidase and matrix metalloproteinases (MMPs), and neutrophil extracellular traps in aggravating alveolar-capillary damage. In this study, we investigated the effects of doxycycline (DOX), an inhibitor of MMPs, on influenza-induced ALI. BALB/c mice were infected with a sublethal dose of mouse-adapted virulent influenza A/Aichi/2/68 (H3N2) virus, and administered daily with 20mg/kg or 60 mg/kg DOX orally. The effects of DOX on ALI were determined by measuring inflammation, capillary leakage, and MMP activities. Furthermore, levels of T1-α (a membrane protein of alveolar type I epithelium) and thrombomodulin (an endothelial protein) in the bronchoalveolar lavage fluid were evaluated by Western blot analysis. Our results demonstrate significantly decreased inflammation and protein leakage in the lungs after DOX treatment. Levels of MMP-2 and MMP-9 activity, T1-α and thrombomodulin were also diminished in the DOX-treated group. These findings were corroborated by histopathologic analyses, which demonstrated significant reduction in lung damage. Although DOX treatment reduced ALI, there were no effects on virus titers and body weights. Taken together, these results demonstrate that DOX may be useful in ameliorating ALI during influenza pneumonia. Further studies are warranted to determine whether DOX can be used in combination with anti-viral agents to alleviate severe influenza pneumonia.


Subject(s)
Acute Lung Injury/prevention & control , Doxycycline/pharmacology , Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections/complications , Acute Lung Injury/complications , Acute Lung Injury/enzymology , Animals , Anti-Bacterial Agents/pharmacology , Blotting, Western , Female , Humans , Lung/drug effects , Lung/metabolism , Lung/virology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Peroxidase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thrombomodulin/genetics , Thrombomodulin/metabolism
8.
Article in English | MEDLINE | ID: mdl-21047904

ABSTRACT

Pluripotency and self-renewal are the defining traits of embryonic stem cells (ESCs) and this status quo is maintained by the core transcription factors Oct4, Sox2, and Nanog. Genome-wide mapping of the binding sites of these pivotal factors and other ESC transcriptional regulators has unraveled the transcriptional network governing pluripotency. Strikingly, a sizeable fraction of the binding sites of Oct4 and Nanog are not conserved in mouse and human ESCs. Binding site turnover and the presence of species-specific transposable elements are some of the factors contributing to this disp arity. Hence, comparing human and mouse ESCs will shed new light on the design of transcriptional regulatory networks for pluripotency. Despite the significant differences among pluripotent mammalian stem cells, the same set of transcription factors (Oct4, Sox2, Klf4, and c-Myc) can be used to reprogram human and mouse somatic cells into induced pluripotent stem cells. Recent works also demonstrate that there are multiple ways of imparting pluripotency. For instance, the nuclear receptors Nr5a2 and Esrrb can, respectively, substitute for Oct4 and Klf4 in reprogramming. This chapter summarizes the different roles of transcription factors in the modulation of pluripotent states and in the induction of pluripotent phenotypes.


Subject(s)
Cellular Reprogramming/genetics , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Gene Expression Regulation, Developmental , Humans , Kruppel-Like Factor 4 , Species Specificity
9.
Am J Respir Cell Mol Biol ; 42(6): 732-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19617401

ABSTRACT

Recent studies have demonstrated an essential role of alveolar macrophages during influenza virus infection. Enhanced mortalities were observed in macrophage-depleted mice and pigs after influenza virus infection, but the basis for the enhanced pathogenesis is unclear. This study revealed that blocking macrophage recruitment into the lungs in a mouse model of influenza pneumonitis resulted in enhanced alveolar epithelial damage and apoptosis, as evaluated by histopathology, immunohistochemistry, Western blot, RT-PCR, and TUNEL assays. Abrogation of macrophage recruitment was achieved by treatment with monoclonal antibody against monocyte chemoattractant protein-1 (MCP-1) after sub-lethal challenge with mouse-adapted human influenza A/Aichi/2/68 virus. Interestingly, elevated levels of hepatocyte growth factor (HGF), a mitogen for alveolar epithelium, were detected in bronchoalveolar lavage samples and in lung homogenates of control untreated and nonimmune immunoglobulin (Ig)G-treated mice after infection compared with anti-MCP-1-treated infected mice. The lungs of control animals also displayed strongly positive HGF staining in alveolar macrophages as well as alveolar epithelial cell hyperplasia. Co-culture of influenza virus-infected alveolar epithelial cells with freshly isolated alveolar macrophages induced HGF production and phagocytic activity of macrophages. Recombinant HGF added to mouse lung explants after influenza virus infection resulted in enhanced BrdU labeling of alveolar type II epithelial cells, indicating their proliferation, in contrast with anti-HGF treatment showing significantly reduced epithelial regeneration. Our data indicate that inhibition of macrophage recruitment augmented alveolar epithelial damage and apoptosis during influenza pneumonitis, and that HGF produced by macrophages in response to influenza participates in the resolution of alveolar epithelium.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Chemokine CCL2/immunology , Influenza A virus/pathogenicity , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Viral/immunology , Pulmonary Alveoli/immunology , Respiratory Mucosa/immunology , Animals , Apoptosis , Blotting, Western , Body Weight , Bronchoalveolar Lavage Fluid/immunology , Cell Proliferation , Cells, Cultured , Chemokine CXCL1/blood , Chemotaxis, Leukocyte , Coculture Techniques , Disease Models, Animal , Female , Hepatocyte Growth Factor/metabolism , Humans , Hyperplasia , Immunohistochemistry , In Situ Nick-End Labeling , Injections, Intraperitoneal , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Macrophages, Alveolar/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Peroxidase/metabolism , Pneumonia, Viral/metabolism , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Recombinant Proteins/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Viral Load
10.
Microbes Infect ; 11(1): 2-11, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18983930

ABSTRACT

Most pandemic influenza virus strains undergo adaptation or reassortment before they acquire the ability to cause fatal infections in a new host species. The pathologic changes and tissue tropism during virus adaptation are not fully understood. Here we investigated pathologic changes and tissue tropism by serial lung-to-lung passaging of human influenza virus strain A/Aichi/2/68 (H3N2) in a BALB/c mouse model. Enhanced pulmonary lesions and systemic virus infection were observed during adaptation. Late passage 10 (P10) virus caused extra-pulmonary spread with necrotic and inflammatory lesions in the brain, heart, spleen and intestine of infected animals, in contrast to infection with earlier passage viruses which were restricted to lungs. Non-conservative mutations in the hemagglutinin (Gly218Glu) and non-structural 1 (Asp125Gly) proteins were identified in P10 virus which exhibited high virulence. Virus growth kinetics showed enhanced replication ability of P10 virus in different cell lines. P10 virus also exhibited the ability to bind to erythrocytes of different host species. These results demonstrate extra-pulmonary spread of influenza virus during adaptation with enhanced replication ability in a new host. This mouse adaptation model may provide a basis for understanding cross-species adaptability corresponding to increased virulence of the influenza A virus, a phenomenon of relevance to the emergence of future highly pathogenic strains.


Subject(s)
Adaptation, Physiological , Disease Models, Animal , Host-Pathogen Interactions , Influenza A Virus, H3N2 Subtype , Influenza, Human , Pneumonia , Adaptation, Physiological/genetics , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Dogs , Female , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/pathology , Influenza, Human/virology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Organ Specificity , Pneumonia/pathology , Pneumonia/virology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virulence
11.
Article in English | MEDLINE | ID: mdl-19022762

ABSTRACT

Embryonic stem (ES) cells are characterized by their ability to self-renew and remain pluripotent. Transcription factors have critical roles in the maintenance of ES cells through specifying an ES-cell-specific gene expression program. Deciphering the transcriptional regulatory network that describes the specific interactions of these transcription factors with the genomic template is crucial for understanding the design and key components of this network. Recent advances in genomic technologies have facilitated genome-wide disclosure of the repertoire of transcription-factor-binding sites. Transcription factor colocalization hot spots targeted by multiple transcription factors have been identified. These are sites that integrate the external signaling pathways to the transcriptional regulatory circuitry governed by Oct4, Sox2, and Nanog. In addition, these sites may serve as focal points for the assembly of nucleoprotein complexes known as enhanceosomes. Studying the properties of ES-cell-specific enhanceosomes in different pluripotent cells will shed light on the composition and regulation of their activity. Knowledge of the transcriptional regulatory networks in different pluripotent cells will also help to distinguish the core and peripheral parts of the networks. Collectively, these studies will facilitate the understanding of molecular mechanisms behind transcription-factor-mediated regulation of pluripotent stem cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Transcription, Genetic , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Regulatory Networks , Genomics , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Mice , Models, Biological , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/physiology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/physiology
12.
Trends Biochem Sci ; 25(3): 121-6, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10694882

ABSTRACT

Over the past few years, the long-standing idea that covalent modification of chromatin can play a role in determining states of gene activity has been confirmed. Eukaryotic genes can be silenced by deacetylation of acetyl-lysine moieties in the N-terminal tails of histones. Recent work links histone deacetylases with an increasing number of repressors, suggesting that deacetylation might be a rather pervasive feature of transcriptional repression systems.


Subject(s)
Gene Silencing , Histone Deacetylases/physiology , Saccharomyces cerevisiae Proteins , Animals , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Humans , Ikaros Transcription Factor , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Smad3 Protein , Trans-Activators/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
13.
Mol Cell Biol ; 20(4): 1394-406, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10648624

ABSTRACT

MBD1 belongs to a family of mammalian proteins that share a methyl-CpG binding domain. Previous work has shown that MBD1 binds to methylated sites in vivo and in vitro and can repress transcription from methylated templates in transcription extracts and in cultured cells. In the present study we established by several experimental criteria that, contrary to a previous report, MBD1 is not a component of the MeCP1 repressor complex. We identified a powerful transcriptional repression domain (TRD) at the C terminus of MBD1 that can actively repress transcription at a distance. Methylation-dependent repression in vivo depends on the presence of both the TRD and the methyl-CpG binding domain. The mechanism is likely to involve deacetylation, since the deacetylase inhibitor trichostatin A can overcome MBD1-mediated repression. Accordingly, we found that endogenous MBD1 is particularly concentrated at sites of centromeric heterochromatin, where acetylated histone H4 is deficient. Unlike MBD2 and MeCP2, MBD1 is not depleted by antibodies to the histone deacetylase HDAC1. Thus, the deacetylase-dependent pathway by which MBD1 actively silences methylated genes is likely to be different from that utilized by the methylation-dependent repressors MeCP1 and MeCP2.


Subject(s)
Repressor Proteins/metabolism , Acetylation , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Human/metabolism , CpG Islands , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Methylation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique, Indirect , HeLa Cells , Histone Deacetylase 1 , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , In Vitro Techniques , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/genetics , Sequence Homology, Amino Acid , Transcription Factors
15.
Nature ; 401(6750): 301-4, 1999 Sep 16.
Article in English | MEDLINE | ID: mdl-10499592

ABSTRACT

In addition to its well-documented effects on gene silencing, cytosine methylation is a prominent cause of mutations. In humans, the mutation rate from 5-methylcytosine (m5C) to thymine (T) is 10-50-fold higher than other transitions and the methylated sequence CpG is consequently under-represented. Over one-third of germline point mutations associated with human genetic disease and many somatic mutations leading to cancer involve loss of CpG. The primary cause of mutability appears to be hydrolytic deamination. Cytosine deamination produces mismatched uracil (U), which can be removed by uracil glycosylase, whereas m5C deamination generates a G x T mispair that cannot be processed by this enzyme. Correction of m5CpG x TpG mismatches may instead be initiated by the thymine DNA glycosylase, TDG. Here we show that MBD4, an unrelated mammalian protein that contains a methyl-CpG binding domain, can also efficiently remove thymine or uracil from a mismatches CpG site in vitro. Furthermore, the methyl-CpG binding domain of MBD4 binds preferentially to m5CpG x TpG mismatches-the primary product of deamination at methyl-CpG. The combined specificities of binding and catalysis indicate that this enzyme may function to minimize mutation at methyl-CpG.


Subject(s)
DNA Repair , DNA/metabolism , Dinucleoside Phosphates/metabolism , Endodeoxyribonucleases/metabolism , Amino Acid Sequence , Animals , DNA Methylation , Deoxyribonuclease (Pyrimidine Dimer) , Humans , Mice , Molecular Sequence Data , Nucleic Acid Heteroduplexes , Sequence Homology, Amino Acid , Substrate Specificity , Thymine/metabolism , Uracil/metabolism
16.
Nat Genet ; 23(1): 58-61, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10471499

ABSTRACT

Mammalian DNA is methylated at many CpG dinucleotides. The biological consequences of methylation are mediated by a family of methyl-CpG binding proteins. The best characterized family member is MeCP2, a transcriptional repressor that recruits histone deacetylases. Our report concerns MBD2, which can bind methylated DNA in vivo and in vitro and has been reported to actively demethylate DNA (ref. 8). As DNA methylation causes gene silencing, the MBD2 demethylase is a candidate transcriptional activator. Using specific antibodies, however, we find here that MBD2 in HeLa cells is associated with histone deacetylase (HDAC) in the MeCP1 repressor complex. An affinity-purified HDAC1 corepressor complex also contains MBD2, suggesting that MeCP1 corresponds to a fraction of this complex. Exogenous MBD2 represses transcription in a transient assay, and repression can be relieved by the deacetylase inhibitor trichostatin A (TSA; ref. 12). In our hands, MBD2 does not demethylate DNA. Our data suggest that HeLa cells, which lack the known methylation-dependent repressor MeCP2, use an alternative pathway involving MBD2 to silence methylated genes.


Subject(s)
DNA-Binding Proteins/physiology , Histone Deacetylases/physiology , Repressor Proteins/physiology , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , DNA Methylation , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Hydroxamic Acids/pharmacology , Mice , Models, Genetic , Molecular Sequence Data , Rats , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Transcriptional Activation , Transfection
17.
Genes Dev ; 13(15): 1924-35, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10444591

ABSTRACT

ATP-dependent nucleosome remodeling and core histone acetylation and deacetylation represent mechanisms to alter nucleosome structure. NuRD is a multisubunit complex containing nucleosome remodeling and histone deacetylase activities. The histone deacetylases HDAC1 and HDAC2 and the histone binding proteins RbAp48 and RbAp46 form a core complex shared between NuRD and Sin3-histone deacetylase complexes. The histone deacetylase activity of the core complex is severely compromised. A novel polypeptide highly related to the metastasis-associated protein 1, MTA2, and the methyl-CpG-binding domain-containing protein, MBD3, were found to be subunits of the NuRD complex. MTA2 modulates the enzymatic activity of the histone deacetylase core complex. MBD3 mediates the association of MTA2 with the core histone deacetylase complex. MBD3 does not directly bind methylated DNA but is highly related to MBD2, a polypeptide that binds to methylated DNA and has been reported to possess demethylase activity. MBD2 interacts with the NuRD complex and directs the complex to methylated DNA. NuRD may provide a means of gene silencing by DNA methylation.


Subject(s)
Adenosine Triphosphatases , Carrier Proteins/metabolism , DNA Helicases , DNA Methylation , DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Multienzyme Complexes/chemistry , Nucleosomes/metabolism , Repressor Proteins , Amino Acid Sequence , Autoantigens/isolation & purification , Autoantigens/metabolism , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Cloning, Molecular , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , HeLa Cells , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Histone Deacetylases/isolation & purification , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Molecular Sequence Data , Multienzyme Complexes/genetics , Multienzyme Complexes/isolation & purification , Multienzyme Complexes/metabolism , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Protein Binding , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Retinoblastoma-Binding Protein 4 , Retinoblastoma-Binding Protein 7 , Sequence Homology, Amino Acid , Transcription Factors
18.
Curr Opin Genet Dev ; 9(2): 158-63, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10322130

ABSTRACT

DNA methylation and chromatin modification are two global mechanisms that regulate gene expression. Recent studies provide insight into the mechanism of transcriptional silencing by a methyl-CpG binding protein, MeCP2. MeCP2 is shown to interact with the Sin3/histone deacetylase co-repressor complex. Thus, this interaction can provide a mechanistic explanation for the long-known relationship between DNA methylation and chromatin structure. Moreover, several studies have shown that inhibition of histone deacetylases by specific inhibitors can reactivate endogenous genes or reporter constructs previously silenced by DNA methylation. Taken together, the data strongly suggest that DNA methylation can pattern chromatin modification.


Subject(s)
Chromatin/metabolism , DNA Methylation , Acetylation , Animals , Down-Regulation , Histones/metabolism , Humans
19.
Nature ; 393(6683): 386-9, 1998 May 28.
Article in English | MEDLINE | ID: mdl-9620804

ABSTRACT

Cytosine residues in the sequence 5'CpG (cytosine-guanine) are often postsynthetically methylated in animal genomes. CpG methylation is involved in long-term silencing of certain genes during mammalian development and in repression of viral genomes. The methyl-CpG-binding proteins MeCP1 and MeCP2 interact specifically with methylated DNA and mediate transcriptional repression. Here we study the mechanism of repression by MeCP2, an abundant nuclear protein that is essential for mouse embryogenesis. MeCP2 binds tightly to chromosomes in a methylation-dependent manner. It contains a transcriptional-repression domain (TRD) that can function at a distance in vitro and in vivo. We show that a region of MeCP2 that localizes with the TRD associates with a corepressor complex containing the transcriptional repressor mSin3A and histone deacetylases. Transcriptional repression in vivo is relieved by the deacetylase inhibitor trichostatin A, indicating that deacetylation of histones (and/or of other proteins) is an essential component of this repression mechanism. The data suggest that two global mechanisms of gene regulation, DNA methylation and histone deacetylation, can be linked by MeCP2.


Subject(s)
Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Acetylation , Amino Acid Sequence , Animals , DNA Methylation , Dinucleoside Phosphates/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors , Histones/metabolism , Hydroxamic Acids/pharmacology , L Cells , Methyl-CpG-Binding Protein 2 , Mice , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Transcription, Genetic , Transfection
20.
Science ; 277(5334): 1996-2000, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9302295

ABSTRACT

DNA-(cytosine-5) methyltransferase (MCMT) methylates newly replicated mammalian DNA, but the factors regulating this activity are unknown. Here, MCMT is shown to bind proliferating cell nuclear antigen (PCNA), an auxiliary factor for DNA replication and repair. Binding of PCNA requires amino acids 163 to 174 of MCMT, occurs in intact cells at foci of newly replicated DNA, and does not alter MCMT activity. A peptide derived from the cell cycle regulator p21(WAF1) can disrupt the MCMT-PCNA interaction, which suggests that p21(WAF1) may regulate methylation by blocking access of MCMT to PCNA. MCMT and p21(WAF1) may be linked in a regulatory pathway, because the extents of their expression are inversely related in both SV40-transformed and nontransformed cells.


Subject(s)
Cyclins/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Proliferating Cell Nuclear Antigen/metabolism , Amino Acid Sequence , Cell Line, Transformed , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/chemistry , DNA Damage , DNA Repair , DNA Replication , Humans , Molecular Sequence Data , Peptides/pharmacology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...