Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Pathol ; 252(1): 41-52, 2020 09.
Article in English | MEDLINE | ID: mdl-32506441

ABSTRACT

The severity of sterile inflammation, as seen in acute pancreatitis, is determined by damage-sensing receptors, signalling cascades and cytokine production. Stat2 is a type I interferon signalling mediator that also has interferon-independent roles in murine lipopolysaccharide-induced NF-κB-mediated sepsis. However, its role in sterile inflammation is unknown. We hypothesised that Stat2 determines the severity of non-infective inflammation in the pancreas. Wild type (WT) and Stat2-/- mice were injected i.p. with caerulein or l-arginine. Specific cytokine-blocking antibodies were used in some experiments. Pancreata and blood were harvested 1 and 24 h after the final dose of caerulein and up to 96 h post l-arginine. Whole-tissue phosphoproteomic changes were assessed using label-free mass spectrometry. Tissue-specific Stat2 effects were studied in WT/Stat2-/- bone marrow chimera and using Cre-lox recombination to delete Stat2 in pancreatic and duodenal homeobox 1 (Pdx1)-expressing cells. Stat2-/- mice were protected from caerulein- and l-arginine-induced pancreatitis. Protection was independent of type I interferon signalling. Stat2-/- mice had lower cytokine levels, including TNF-α and IL-10, and reduced NF-κB nuclear localisation in pancreatic tissue compared with WT. Inhibition of TNF-α improved (inhibition of IL-10 worsened) caerulein-induced pancreatitis in WT but not Stat2-/- mice. Phosphoproteomics showed downregulation of MAPK mediators but accumulation of Ser412-phosphorylated Tak1. Stat2 deletion in Pdx1-expressing acinar cells (Stat2flox/Pdx1-cre ) reduced pancreatic TNF-α expression, but not histological injury or serum amylase. WT/Stat2-/- bone marrow chimera mice were protected from pancreatitis irrespective of host or recipient genotype. Stat2 loss results in disrupted signalling in pancreatitis, upstream of NF-κB in non-acinar and/or bone marrow-derived cells. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Inflammation/genetics , Pancreas/metabolism , Pancreatitis/genetics , STAT2 Transcription Factor/genetics , Acute Disease , Animals , Arginine , Ceruletide , Cytokines/blood , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/pathology , Phosphorylation , STAT2 Transcription Factor/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism
2.
Proc Natl Acad Sci U S A ; 110(21): 8656-61, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23653476

ABSTRACT

Deregulated Toll-like receptor (TLR)-triggered inflammatory responses that depend on NF-κB are detrimental to the host via excessive production of proinflammatory cytokines, including TNF-α. Stat2 is a critical component of type I IFN signaling, but it is not thought to participate in TLR signaling. Our study shows that LPS-induced lethality in Stat2(-/-) mice is accelerated as a result of increased cellular transmigration. Blocking intercellular adhesion molecule-1 prevents cellular egress and confers survival of Stat2(-/-) mice. The main determinant of cellular egress in Stat2(-/-) mice is the genotype of the host and not the circulating leukocyte. Surprisingly, lethality and cellular egress observed on Stat2(-/-) mice are not associated with excessive increases in classical sepsis cytokines or chemokines. Indeed, in the absence of Stat2, cytokine production in response to multiple TLR agonists is reduced. We find that Stat2 loss leads to reduced expression of NF-κB target genes by affecting nuclear translocation of NF-κB. Thus, our data reveal the existence of a different mechanism of LPS-induced lethality that is independent of NF-κB triggered cytokine storm but dependent on cellular egress.


Subject(s)
Cell Nucleus/metabolism , Cytokines/biosynthesis , Lipopolysaccharides/toxicity , NF-kappa B/metabolism , STAT2 Transcription Factor , Sepsis/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Animals , Cell Nucleus/genetics , Cytokines/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Knockout , NF-kappa B/genetics , Sepsis/chemically induced , Sepsis/genetics , Sepsis/pathology , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
3.
Epigenetics Chromatin ; 5(1): 8, 2012 Jun 18.
Article in English | MEDLINE | ID: mdl-22709888

ABSTRACT

BACKGROUND: CTCF is a highly conserved and essential zinc finger protein expressed in virtually all cell types. In conjunction with cohesin, it organizes chromatin into loops, thereby regulating gene expression and epigenetic events. The function of CTCFL or BORIS, the testis-specific paralog of CTCF, is less clear. RESULTS: Using immunohistochemistry on testis sections and fluorescence-based microscopy on intact live seminiferous tubules, we show that CTCFL is only transiently present during spermatogenesis, prior to the onset of meiosis, when the protein co-localizes in nuclei with ubiquitously expressed CTCF. CTCFL distribution overlaps completely with that of Stra8, a retinoic acid-inducible protein essential for the propagation of meiosis. We find that absence of CTCFL in mice causes sub-fertility because of a partially penetrant testicular atrophy. CTCFL deficiency affects the expression of a number of testis-specific genes, including Gal3st1 and Prss50. Combined, these data indicate that CTCFL has a unique role in spermatogenesis. Genome-wide RNA expression studies in ES cells expressing a V5- and GFP-tagged form of CTCFL show that genes that are downregulated in CTCFL-deficient testis are upregulated in ES cells. These data indicate that CTCFL is a male germ cell gene regulator. Furthermore, genome-wide DNA-binding analysis shows that CTCFL binds a consensus sequence that is very similar to that of CTCF. However, only ~3,700 out of the ~5,700 CTCFL- and ~31,000 CTCF-binding sites overlap. CTCFL binds promoters with loosely assembled nucleosomes, whereas CTCF favors consensus sites surrounded by phased nucleosomes. Finally, an ES cell-based rescue assay shows that CTCFL is functionally different from CTCF. CONCLUSIONS: Our data suggest that nucleosome composition specifies the genome-wide binding of CTCFL and CTCF. We propose that the transient expression of CTCFL in spermatogonia and preleptotene spermatocytes serves to occupy a subset of promoters and maintain the expression of male germ cell genes.

4.
Proc Natl Acad Sci U S A ; 107(50): 21836-41, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21106760

ABSTRACT

Cellular metabolism alters patterns of gene expression through a variety of mechanisms, including alterations in histone modifications and transcription factor activity. Nicotinamide adenine dinucleotide (NAD)-dependent proteins such as poly(ADP ribose) polymerases (PARPs) and sirtuin deacetylases play important roles in this regulation, thus NAD provides a crucial link between metabolism and these cellular signaling processes. Here, we found that lowering NAD levels in mouse primary cortical neurons led to decreased activity-dependent BDNF expression. The altered BDNF transcription occurred independently of Sirt or Parp activities; instead, low NAD levels promoted increased DNA methylation of the activity-dependent BDNF promoter. Increased methylation at this promoter triggered the dissociation of the insulator protein CTCF as well as the accompanying cohesin from the BDNF locus. The loss of these proteins resulted in histone acetylation and methylation changes at this locus consistent with chromatin compaction and gene silencing. Because BDNF is critical for neuronal function, these results suggest that age- or nutrition-associated declines in NAD levels as well as deficits in cohesin function associated with disease modulate BDNF expression and could contribute to cognitive impairment.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , NAD/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Animals , Brain-Derived Neurotrophic Factor/metabolism , CCCTC-Binding Factor , Cell Cycle Proteins/genetics , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , DNA Methylation , Gene Expression Regulation , Mice , Neurons/cytology , Neurons/physiology , Promoter Regions, Genetic , Repressor Proteins/genetics , Cohesins
5.
Epigenetics Chromatin ; 3(1): 19, 2010 Nov 08.
Article in English | MEDLINE | ID: mdl-21059229

ABSTRACT

BACKGROUND: CCCTC binding factor (CTCF) is a highly conserved zinc finger protein, which is involved in chromatin organization, local histone modifications, and RNA polymerase II-mediated gene transcription. CTCF may act by binding tightly to DNA and recruiting other proteins to mediate its various functions in the nucleus. To further explore the role of this essential factor, we used a mass spectrometry-based approach to screen for novel CTCF-interacting partners. RESULTS: Using biotinylated CTCF as bait, we identified upstream binding factor (UBF) and multiple other components of the RNA polymerase I complex as potential CTCF-interacting partners. Interestingly, CTCFL, the testis-specific paralog of CTCF, also binds UBF. The interaction between CTCF(L) and UBF is direct, and requires the zinc finger domain of CTCF(L) and the high mobility group (HMG)-box 1 and dimerization domain of UBF. Because UBF is involved in RNA polymerase I-mediated ribosomal (r)RNA transcription, we analyzed CTCF binding to the rDNA repeat. We found that CTCF bound to a site upstream of the rDNA spacer promoter and preferred non-methylated over methylated rDNA. DNA binding by CTCF in turn stimulated binding of UBF. Absence of CTCF in cultured cells resulted in decreased association of UBF with rDNA and in nucleolar fusion. Furthermore, lack of CTCF led to reduced binding of RNA polymerase I and variant histone H2A.Z near the rDNA spacer promoter, a loss of specific histone modifications, and diminished transcription of non-coding RNA from the spacer promoter. CONCLUSIONS: UBF is the first common interaction partner of CTCF and CTCFL, suggesting a role for these proteins in chromatin organization of the rDNA repeats. We propose that CTCF affects RNA polymerase I-mediated events globally by controlling nucleolar number, and locally by regulating chromatin at the rDNA spacer promoter, similar to RNA polymerase II promoters. CTCF may load UBF onto rDNA, thereby forming part of a network that maintains rDNA genes poised for transcription.

6.
J Immunol ; 182(2): 999-1010, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124743

ABSTRACT

Differentiation of naive CD4+ cells into Th2 cells is accompanied by chromatin remodeling at the Th2 cytokine locus allowing the expression of the IL-4, IL-5, and IL-13 genes. In this report, we investigated the role in Th2 differentiation of the transcription regulator CCCTC-binding factor (CTCF). Chromatin immunoprecipitation analysis revealed multiple CTCF binding sites in the Th2 cytokine locus. Conditional deletion of the Ctcf gene in double-positive thymocytes allowed development of peripheral T cells, but their activation and proliferation upon anti-CD3/anti-CD28 stimulation in vitro was severely impaired. Nevertheless, when TCR signaling was circumvented with phorbol ester and ionomycin, we observed proliferation of CTCF-deficient T cells, enabling the analysis of Th2 differentiation in vitro. We found that in CTCF-deficient Th2 polarization cultures, transcription of IL-4, IL-5, and IL-13 was strongly reduced. By contrast, CTCF deficiency had a moderate effect on IFN-gamma production in Th1 cultures and IL-17 production in Th17 cultures was unaffected. Consistent with a Th2 cytokine defect, CTCF-deficient mice had very low levels of IgG1 and IgE in their serum, but IgG2c was close to normal. In CTCF-deficient Th2 cultures, cells were polarized toward the Th2 lineage, as substantiated by induction of the key transcriptional regulators GATA3 and special AT-rich binding protein 1 (SATB1) and down-regulation of T-bet. Also, STAT4 expression was low, indicating that in the absence of CTCF, GATA3 still operated as a negative regulator of STAT4. Taken together, these findings show that CTCF is essential for GATA3- and SATB1-dependent regulation of Th2 cytokine gene expression.


Subject(s)
Cytokines/biosynthesis , Cytokines/genetics , DNA-Binding Proteins/physiology , Repressor Proteins/physiology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Binding Sites/genetics , CCCTC-Binding Factor , Cells, Cultured , Chromatin Immunoprecipitation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , GATA3 Transcription Factor/biosynthesis , GATA3 Transcription Factor/physiology , Gene Deletion , Matrix Attachment Region Binding Proteins/biosynthesis , Matrix Attachment Region Binding Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Th2 Cells/pathology , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/metabolism , Trans-Activators/physiology
7.
EMBO J ; 27(21): 2839-50, 2008 Nov 05.
Article in English | MEDLINE | ID: mdl-18923423

ABSTRACT

The 11-zinc finger protein CCCTC-binding factor (CTCF) is a highly conserved protein, involved in imprinting, long-range chromatin interactions and transcription. To investigate its function in vivo, we generated mice with a conditional Ctcf knockout allele. Consistent with a previous report, we find that ubiquitous ablation of the Ctcf gene results in early embryonic lethality. Tissue-specific inactivation of CTCF in thymocytes specifically hampers the differentiation of alphabeta T cells and causes accumulation of late double-negative and immature single-positive cells in the thymus of mice. These cells are normally large and actively cycling, and contain elevated amounts of CTCF. In Ctcf knockout animals, however, these cells are small and blocked in the cell cycle due to increased expression of the cyclin-CDK inhibitors p21 and p27. Taken together, our results show that CTCF is required in a dose-dependent manner and is involved in cell cycle progression of alphabeta T cells in the thymus. We propose that CTCF positively regulates cell growth in rapidly dividing thymocytes so that appropriate number of cells are generated before positive and negative selection in the thymus.


Subject(s)
Cell Cycle , DNA-Binding Proteins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , CCCTC-Binding Factor , Cell Lineage , Cell Proliferation , Cell Size , DNA-Binding Proteins/deficiency , Gene Deletion , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Gene Targeting , Genotype , Humans , Integrases/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , T-Lymphocytes/enzymology , Thymus Gland/enzymology
8.
Genes Dev ; 20(17): 2349-54, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951251

ABSTRACT

CTCF (CCCTC-binding factor) binds sites around the mouse beta-globin locus that spatially cluster in the erythroid cell nucleus. We show that both conditional deletion of CTCF and targeted disruption of a DNA-binding site destabilize these long-range interactions and cause local loss of histone acetylation and gain of histone methylation, apparently without affecting transcription at the locus. Our data demonstrate that CTCF is directly involved in chromatin architecture and regulates local balance between active and repressive chromatin marks. We postulate that throughout the genome, relative position and stability of CTCF-mediated loops determine their effect on enhancer-promoter interactions, with gene insulation as one possible outcome.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/physiology , Globins/genetics , Histones/metabolism , Repressor Proteins/physiology , Animals , CCCTC-Binding Factor , Cell Line , Cells, Cultured , Chromatin/chemistry , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Erythroid Cells/metabolism , Fetus , Genetic Markers , Globins/biosynthesis , Globins/metabolism , Mice , Mice, Knockout , Repressor Proteins/genetics
9.
Mol Cell Biol ; 26(3): 976-89, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16428451

ABSTRACT

Homologous recombination is a versatile DNA damage repair pathway requiring Rad51 and Rad54. Here we show that a mammalian Rad54 paralog, Rad54B, displays physical and functional interactions with Rad51 and DNA that are similar to those of Rad54. While ablation of Rad54 in mouse embryonic stem (ES) cells leads to a mild reduction in homologous recombination efficiency, the absence of Rad54B has little effect. However, the absence of both Rad54 and Rad54B dramatically reduces homologous recombination efficiency. Furthermore, we show that Rad54B protects ES cells from ionizing radiation and the interstrand DNA cross-linking agent mitomycin C. Interestingly, at the ES cell level the paralogs do not display an additive or synergic interaction with respect to mitomycin C sensitivity, yet animals lacking both Rad54 and Rad54B are dramatically sensitized to mitomycin C compared to either single mutant. This suggests that the paralogs possibly function in a tissue-specific manner. Finally, we show that Rad54, but not Rad54B, is needed for a normal distribution of Rad51 on meiotic chromosomes. Thus, even though the paralogs have similar biochemical properties, genetic analysis in mice uncovered their nonoverlapping roles.


Subject(s)
DNA Damage , DNA Helicases/physiology , DNA Repair , Nuclear Proteins/physiology , Recombination, Genetic , Animals , Antibiotics, Antineoplastic/pharmacology , Chromosome Aberrations , Chromosomes/chemistry , DNA Helicases/genetics , DNA-Binding Proteins , Drug Resistance, Neoplasm/drug effects , Humans , Meiosis , Mice , Mice, Mutant Strains , Mitomycin/pharmacology , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Rad51 Recombinase/analysis , Rad51 Recombinase/metabolism , Radiation Tolerance/genetics , Stem Cells/drug effects , Stem Cells/enzymology , Stem Cells/radiation effects
10.
Int J Nurs Stud ; 41(2): 141-50, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14725778

ABSTRACT

Novice qualitative researchers are often unsure regarding the analysis of their data and, where grounded theory is chosen, they may be uncertain regarding the differences that now exist between the approaches of Glaser and Strauss, who together first described the method. These two approaches are compared in relation to roots and divergences, role of induction, deduction and verification, ways in which data are coded and the format of generated theory. Personal experience of developing as a ground theorist is used to illustrate some of the key differences. A conclusion is drawn that, rather than debate relative merits of the two approaches, suggests that novice researchers need to select the method that best suits their cognitive style and develop analytic skills through doing research.


Subject(s)
Nursing Methodology Research/methods , Nursing Theory , Philosophy, Nursing , Qualitative Research , Research Design , Attitude of Health Personnel , Data Collection/methods , Data Interpretation, Statistical , Existentialism , Humans , Logic , Models, Nursing , Models, Psychological , Prejudice , Research Personnel/psychology , Sensitivity and Specificity , Symbolism
11.
Mol Cell ; 10(5): 1163-74, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12453423

ABSTRACT

The transcription/repair factor TFIIH operates as a DNA helix opener in RNA polymerase II (RNAP2) transcription and nucleotide excision repair. To study TFIIH in vivo, we generated cell lines expressing functional GFP-tagged TFIIH. TFIIH was homogeneously distributed throughout the nucleus with nucleolar accumulations. We provide in vivo evidence for involvement of TFIIH in RNA polymerase I (RNAP1) transcription. Photobleaching revealed that TFIIH moves freely and gets engaged in RNAP1 and RNAP2 transcription for approximately 25 and approximately 6 s, respectively. TFIIH readily switches between transcription and repair sites (where it is immobilized for approximately 4 min) without large-scale alterations in composition. Our findings support a model of diffusion and random collision of individual components that permits a quick and versatile response to changing conditions.


Subject(s)
DNA Repair , RNA Polymerase II/chemistry , RNA Polymerase I/chemistry , Transcription Factors, TFII/metabolism , Animals , CHO Cells , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , Cricetinae , Cytoplasm/metabolism , DNA/metabolism , DNA Damage , Dose-Response Relationship, Radiation , Green Fluorescent Proteins , Light , Luminescent Proteins/metabolism , Microscopy, Confocal , Models, Biological , RNA Polymerase I/metabolism , RNA Polymerase II/metabolism , Recombinant Fusion Proteins/metabolism , Time Factors , Transcription Factor TFIIH , Transcription Factors, TFII/chemistry , Transcription, Genetic , Transfection , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL