Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Nat Immunol ; 17(3): 331-43, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26779602

ABSTRACT

The transcription factor Blimp-1 is necessary for the generation of plasma cells. Here we studied its functions in plasmablast differentiation by identifying regulated Blimp-1 target genes. Blimp-1 promoted the migration and adhesion of plasmablasts. It directly repressed genes encoding several transcription factors and Aicda (which encodes the cytidine deaminase AID) and thus silenced B cell-specific gene expression, antigen presentation and class-switch recombination in plasmablasts. It directly activated genes, which led to increased expression of the plasma cell regulator IRF4 and proteins involved in immunoglobulin secretion. Blimp-1 induced the transcription of immunoglobulin genes by controlling the 3' enhancers of the loci encoding the immunoglobulin heavy chain (Igh) and κ-light chain (Igk) and, furthermore, regulated the post-transcriptional expression switch from the membrane-bound form of the immunoglobulin heavy chain to its secreted form by activating Ell2 (which encodes the transcription-elongation factor ELL2). Notably, Blimp-1 recruited chromatin-remodeling and histone-modifying complexes to regulate its target genes. Hence, many essential functions of plasma cells are under the control of Blimp-1.


Subject(s)
Cell Differentiation/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin kappa-Chains/immunology , Interferon Regulatory Factors/immunology , Plasma Cells/immunology , Transcription Factors/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Differentiation/genetics , Cell Migration Assays, Leukocyte , Cell Movement/genetics , Cell Movement/immunology , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin kappa-Chains/genetics , Interferon Regulatory Factors/genetics , Mass Spectrometry , Mice , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Analysis, RNA , Transcription Factors/genetics
2.
BMC Public Health ; 18(1): 16, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28743246

ABSTRACT

BACKGROUND: As more countries worldwide develop national viral hepatitis strategies, it is important to ask whether context-specific factors affect their decision-making. This study aimed to determine whether country-level socioeconomic factors are associated with viral hepatitis programmes and policy responses across WHO Member States (MS). METHODS: WHO MS focal points completed a questionnaire on national viral hepatitis policies. This secondary analysis of data reported in the 2013 Global Policy Report on the Prevention and Control of Viral Hepatitis in WHO Member States used logistic regression to examine associations between four survey questions and four socioeconomic factors: country income level, Human Development Index (HDI), health expenditure and physician density. RESULTS: This analysis included 119 MS. MS were more likely to have routine viral hepatitis surveillance and to have a national strategy and/or policy/guidelines for preventing infection in healthcare settings if they were in the higher binary categories for income level, HDI, health expenditure and physician density. In multivariable analyses, the only significant finding was a positive association between having routine surveillance and being in the higher binary HDI category (adjusted odds ratio 26; 95% confidence interval 2.0-340). CONCLUSION: Countries with differing socioeconomic status indicators did not appear to differ greatly regarding the existence of key national policies and programmes. A more nuanced understanding of the multifaceted interactions of socioeconomic factors, health policy, service delivery and health outcomes is needed to support country-level efforts to eliminate viral hepatitis.


Subject(s)
Delivery of Health Care/organization & administration , Delivery of Health Care/statistics & numerical data , Health Policy , Hepatitis, Viral, Human/prevention & control , Hepatitis, Viral, Human/therapy , Socioeconomic Factors , Adult , Female , Humans , Odds Ratio , World Health Organization
3.
J Neurosci ; 28(26): 6703-13, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18579744

ABSTRACT

In the developing mouse brain, once the generation of neurons is mostly completed during the prenatal period, precisely coordinated signals act on competent neural precursors to direct their differentiation into astrocytes, which occurs mostly after birth. Among these signals, those provided by neurotrophic cytokines and bone morphogenetic proteins appear to have a key role in triggering the neurogenic to gliogenic switch and in regulating astrocyte numbers. In addition, we have reported previously that the neurotrophic peptide pituitary adenylate cyclase-activating polypeptide (PACAP) is able to promote astrocyte differentiation of cortical precursors via activation of a cAMP-dependent pathway. Signals acting on progenitor cells of the developing cortex to generate astrocytes activate glial fibrillary acidic protein (GFAP) gene expression, but the transcriptional mechanisms that regulate this activation are unclear. Here, we identify the previously known transcriptional repressor downstream regulatory element antagonist modulator (DREAM) as an activator of GFAP gene expression. We found that DREAM occupies specific sites on the GFAP promoter before and after differentiation is initiated by exposure of cortical progenitor cells to PACAP. PACAP raises intracellular calcium concentration via a mechanism that requires cAMP, and DREAM-mediated transactivation of the GFAP gene requires the integrity of calcium-binding domains. Cortical progenitor cells from dream(-/-) mice fail to express GFAP in response to PACAP. Moreover, the neonatal cortex of dream(-/-) mice exhibits a reduced number of astrocytes and increased number of neurons. These results identify the PACAP-cAMP-Ca(2+)-DREAM cascade as a new pathway to activate GFAP gene expression during astrocyte differentiation.


Subject(s)
Astrocytes/metabolism , Calcium Signaling/genetics , Cerebral Cortex/embryology , Cyclic AMP/metabolism , Glial Fibrillary Acidic Protein/genetics , Kv Channel-Interacting Proteins/genetics , Repressor Proteins/genetics , Animals , Astrocytes/cytology , Astrocytes/drug effects , Calcium/metabolism , Calcium/pharmacology , Calcium Signaling/drug effects , Cell Count , Cell Differentiation/genetics , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Promoter Regions, Genetic/genetics , Rats , Rats, Wistar , Signal Transduction/genetics
4.
Mol Cell Neurosci ; 39(3): 291-301, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18707003

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) acts on cortical precursor cells to trigger glial fibrillary acidic protein (GFAP) gene expression and astrocyte differentiation by stimulation of intracellular cAMP production. Here, we show that as expected, PACAP activates cAMP-dependent protein kinase A. However, inhibition of protein kinase A does not prevent PACAP-induced GFAP gene expression or astrocytogenesis. PACAP also activates the small GTPases Rap1 and Ras, but either activation of Rap1 alone by selective stimulation of the guanine nucleotide exchange factor Epac, or expression of a constitutively active form of Ras, do not induce GFAP gene expression. Ras is activated by PACAP in a cAMP-dependent manner, and inhibition of Ras and/or Rap1 decreases PACAP-induced GFAP promoter stimulation. Thus, cAMP-dependent PACAP-induced GFAP expression during astrocytogenesis involves the coordinated activation of both Ras and Rap1, but activation of either one of them in isolation is not sufficient to trigger this response.


Subject(s)
Cerebral Cortex/cytology , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Stem Cells/physiology , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/metabolism , Enzyme Activation , Glial Fibrillary Acidic Protein/genetics , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Stem Cells/cytology , rap1 GTP-Binding Proteins/genetics , ras Proteins/genetics
5.
J Neurochem ; 97(4): 1057-70, 2006 May.
Article in English | MEDLINE | ID: mdl-16606365

ABSTRACT

The elucidation of the transcriptional mechanisms that regulate glial fibrillary acidic protein (GFAP) gene expression is important for the understanding of the molecular mechanisms that control astrocyte differentiation during brain development. We investigated regulatory elements located in a proximal region of the GFAP promoter, important for expression in cortical precursor cells differentiating into astrocytes. One of these elements recognizes transcription factors of the nuclear factor-I family (NFI). We found that, in primary cultures of cortical cells, NFI occupies the GFAP promoter prior to the induction of astrocyte differentiation. In the developing cerebral cortex, the onset of expression of NFI coincides chronologically with the beginning of astrocytogenesis. Mutational analysis of the GFAP gene and transfections in primary cortical precursors show that inhibition of binding of NFI to the GFAP promoter results in decreased levels of transcriptional activity and is required for the synergistic stimulation of the GFAP promoter by the astrogenic agents, pituitary adenylate cyclase-activating polypeptide and ciliary neurotrophic factor, which in combination enhance astrocyte differentiation to generate astrocytes with longer processes. Thus, NFI appears to be an important factor for the integration of astrogenic stimuli in the developing central nervous system.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Glial Fibrillary Acidic Protein/genetics , NFI Transcription Factors/metabolism , Stem Cells/metabolism , Animals , Astrocytes/cytology , Binding Sites/drug effects , Binding Sites/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/biosynthesis , NFI Transcription Factors/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , Rats , Rats, Wistar , Regulatory Elements, Transcriptional/genetics , Stem Cells/cytology , Stem Cells/drug effects , Transcriptional Activation/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL