Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 801: 31-41, 2014.
Article in English | MEDLINE | ID: mdl-24664678

ABSTRACT

Cone dystrophy with supernormal rod response (CDSRR) is an autosomal recessive disorder that leads to progressive retinal degeneration with a distinct electroretinogram (ERG) phenotype. CDSRR patients show reduced sensitivity to dim light, augmented response to suprathreshold light and reduced response to flicker. The disorder is caused by mutations in the KCNV2 gene, which encodes the Kv11.1 subunit of a voltage-gated potassium channel. Here, we studied the retina-specific expression and cis-regulatory activity of the murine Kcnv2 gene using electroporation of explanted retinas. Using qRT-PCR profiling of early postnatal retinas, we showed that Kcnv2 expression increased towards P14, which marks the beginning of visual activity in mice. In vivo electroporation of GFP-Kcnv2 expressing plasmids revealed that Kv11.1 localizes to the inner segment membranes of adult P21 photoreceptors. Using bioinformatic prediction and chromatin immunoprecipitation (ChIP), we identified two Crx binding sites (CBS) and one Nrl binding site (NBS) in the Kcnv2 promoter. Reporter electroporation of the wild type promoter region induced strong DsRed expression, indicating high regulatory activity, whereas shRNA-mediated knockdown of Crx and Nrl resulted in reduced Kcnv2 promoter activity and low endogenous Kcnv2 mRNA expression in the retina. Site-directed mutagenesis of the CBS and NBS demonstrated that CBS2 is crucial for Kcnv2 promoter activity. We conclude that nucleotide changes in evolutionary conserved CBS could impact retina-specific expression levels of Kcnv2.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Potassium Channels, Voltage-Gated/genetics , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/genetics , Trans-Activators/metabolism , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Eye Proteins/genetics , Genetic Complementation Test , Homeodomain Proteins/genetics , Leucine Zippers/genetics , Leucine Zippers/physiology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Potassium Channels, Voltage-Gated/physiology , Promoter Regions, Genetic/physiology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics
2.
PLoS One ; 8(4): e60633, 2013.
Article in English | MEDLINE | ID: mdl-23565263

ABSTRACT

Inherited retinal diseases are mainly caused by mutations in genes that are highly expressed in photoreceptors of the retina. The majority of these genes is under the control of the transcription factor Cone rod homeobox (Crx), that acts as a master transcription factor in photoreceptors. Using a genome-wide chromatin immunoprecipitation dataset that highlights all potential in vivo targets of Crx, we have identified a novel sterile alpha motif (SAM) domain containing protein, Samd7. mRNA Expression of Samd7 was confined to the late postnatal and adult mouse retina as well as the pineal gland. Using immunohistochemistry and Western blot, we could detect Samd7 protein in the outer nuclear layer of adult mouse retina. Ectopic over-expression in HEK293 cells demonstrated that Samd7 resides in the cytoplasm as well as the nucleus. In vitro electroporation of fluorescent reporters into living mouse retinal cultures revealed that transcription of the Samd7 gene depends on evolutionary conserved Crx motifs located in the first intron enhancer. Moreover, Crx knock-down with shRNA strongly reduced Samd7 reporter activity and endogenous Samd7 protein, indicating that Crx is required for retinal expression of Samd7. Finally, using co-transfections in luciferase reporter assays we found that Samd7 interferes with Crx-dependent transcription. Samd7 suppressed luciferase activity from a reporter plasmid with five Crx consensus repeats in a dose dependent manner and reduced Crx-mediated transactivation of regulatory sequences in the retinoschisin gene and the Samd7 gene itself. Taken together, we have identified a novel retinal SAM domain protein, Samd7, which could act as a transcriptional repressor involved in fine-tuning of Crx-regulated gene expression.


Subject(s)
Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Retina/metabolism , Trans-Activators/metabolism , Animals , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Pineal Gland/metabolism , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics
4.
J Neuroinflammation ; 8: 125, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21958395

ABSTRACT

BACKGROUND: Microglial cells are important effectors of the neuronal innate immune system with a major role in chronic neurodegenerative diseases. Curcumin, a major component of tumeric, alleviates pro-inflammatory activities of these cells by inhibiting nuclear factor kappa B (NFkB) signaling. To study the immuno-modulatory effects of curcumin on a transcriptomic level, DNA-microarray analyses were performed with resting and LPS-challenged microglial cells after short-term treatment with curcumin. METHODS: Resting and LPS-activated BV-2 cells were stimulated with curcumin and genome-wide mRNA expression patterns were determined using DNA-microarrays. Selected qRT-PCR analyses were performed to confirm newly identified curcumin-regulated genes. The migration potential of microglial cells was determined with wound healing assays and transwell migration assays. Microglial neurotoxicity was estimated by morphological analyses and quantification of caspase 3/7 levels in 661W photoreceptors cultured in the presence of microglia-conditioned medium. RESULTS: Curcumin treatment markedly changed the microglial transcriptome with 49 differentially expressed transcripts in a combined analysis of resting and activated microglial cells. Curcumin effectively triggered anti-inflammatory signals as shown by induced expression of Interleukin 4 and Peroxisome proliferator activated receptor α. Several novel curcumin-induced genes including Netrin G1, Delta-like 1, Platelet endothelial cell adhesion molecule 1, and Plasma cell endoplasmic reticulum protein 1, have been previously associated with adhesion and cell migration. Consequently, curcumin treatment significantly inhibited basal and activation-induced migration of BV-2 microglia. Curcumin also potently blocked gene expression related to pro-inflammatory activation of resting cells including Toll-like receptor 2 and Prostaglandin-endoperoxide synthase 2. Moreover, transcription of NO synthase 2 and Signal transducer and activator of transcription 1 was reduced in LPS-triggered microglia. These transcriptional changes in curcumin-treated LPS-primed microglia also lead to decreased neurotoxicity with reduced apoptosis of 661W photoreceptor cultures. CONCLUSIONS: Collectively, our results suggest that curcumin is a potent modulator of the microglial transcriptome. Curcumin attenuates microglial migration and triggers a phenotype with anti-inflammatory and neuroprotective properties. Thus, curcumin could be a nutraceutical compound to develop immuno-modulatory and neuroprotective therapies for the treatment of various neurodegenerative disorders.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Movement/drug effects , Curcumin/pharmacology , Gene Expression/drug effects , Microglia/drug effects , Microglia/physiology , Animals , Cell Line , Gene Expression Profiling , Lipopolysaccharides/pharmacology , Mice , Oligonucleotide Array Sequence Analysis , Transcriptome/drug effects
5.
Biochim Biophys Acta ; 1809(4-6): 245-54, 2011.
Article in English | MEDLINE | ID: mdl-21392589

ABSTRACT

X-linked juvenile retinoschisis (XLRS) is an orphan retinal disease in males caused by mutations in the RS1 gene. Previously we have characterized cone-rod homeobox (CRX)-responsive elements in the promoter region of RS1 driving selective gene expression in the retina. Here, we expanded our identification and functional analysis of cis-regulatory elements controlling quantitative expression of RS1 in vitro and in vivo. Sequence analysis identified a CpG island 3kb upstream of the transcription start site (TSS). In addition, chromatin immunoprecipitation coupled to microarrays (ChIP-Chip) targeting the retinal transcription factor CRX was performed. Thereby, we identified a second CRX-bound region (CBR2) in the first intron of RS1 which contains six evolutionarily conserved CRX binding motifs. In vitro luciferase reporter gene assays and dsRed reporter electroporation of mouse retinal organ cultures demonstrated a strong constitutive and orientation-independent enhancing effect of the upstream CpG island. The intronic CBR2 potently suppressed CBR1-driven RS1 promoter activity in vitro but failed to regulate a CBR1-reporter in short-term cultured mouse retinae. We conclude that a CpG island enhancer and two CBRs may act in a combinatorial fashion to fine-tune RS1 transcript levels in the retina.


Subject(s)
Cell Adhesion Molecules/genetics , CpG Islands/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Retina/metabolism , Trans-Activators/genetics , 5' Flanking Region/genetics , Animals , Base Sequence , Binding Sites/genetics , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Eye Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , Regulatory Sequences, Nucleic Acid/genetics , Sequence Homology, Nucleic Acid , Tissue Culture Techniques , Trans-Activators/metabolism
6.
J Immunol ; 185(6): 3379-90, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20709948

ABSTRACT

Microgliosis is a common phenomenon in neurodegenerative disorders, including retinal dystrophies. To identify candidate genes involved in microglial activation, we used DNA-microarray analysis of retinal microglia from wild-type and retinoschisin-deficient (Rs1h(-/Y)) mice, a prototypic model for inherited retinal degeneration. Thereby, we cloned a novel 76 aa protein encoding a microglia/macrophage-restricted whey acidic protein (WAP) termed activated microglia/macrophage WAP domain protein (AMWAP). The gene consists of three exons and is located on mouse chromosome 11 in proximity to a chemokine gene cluster. mRNA expression of AMWAP was detected in microglia from Rs1h(-/Y) retinas, brain microglia, and other tissue macrophages. AMWAP transcription was rapidly induced in BV-2 microglia upon stimulation with multiple TLR ligands and IFN-gamma. The TLR-dependent expression of AMWAP was dependent on NF-kappaB, whereas its microglia/macrophage-specific transcription was regulated by PU.1. Functional characterization showed that AMWAP overexpression reduced the proinflammatory cytokines IL-6 and IL-1beta and concomitantly increased expression of the alternative activation markers arginase 1 and Cd206. Conversely, small interfering RNA knockdown of AMWAP lead to higher IL-6, IL-1beta, and Ccl2 transcript levels, whereas diminishing arginase 1 and Cd206 expression. Moreover, AMWAP expressing cells had less migratory capacity and showed increased adhesion in a trypsin-protection assay indicating antiserine protease activity. In agreement with findings from other WAP proteins, micromolar concentrations of recombinant AMWAP exhibited significant growth inhibitory activity against Escherichia coli, Pseudomonas aeruginosa, and Bacillus subtilis. Taken together, we propose that AMWAP is a counter-regulator of proinflammatory microglia/macrophage activation and a potential modulator of innate immunity in neurodegeneration.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation Mediators/physiology , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Microglia/immunology , Microglia/metabolism , Milk Proteins/pharmacology , Nerve Tissue Proteins/pharmacology , Amino Acid Sequence , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Base Sequence , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Line , Cells, Cultured , Contraindications , Eye Proteins/genetics , Inflammation Mediators/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Milk Proteins/biosynthesis , Milk Proteins/genetics , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Structure, Tertiary , Retina/cytology , Retina/immunology , Retina/metabolism
7.
J Neuroinflammation ; 7: 3, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-20074346

ABSTRACT

BACKGROUND: Luteolin, a plant derived flavonoid, exerts a variety of pharmacological activities and anti-oxidant properties associated with its capacity to scavenge oxygen and nitrogen species. Luteolin also shows potent anti-inflammatory activities by inhibiting nuclear factor kappa B (NFkB) signaling in immune cells. To better understand the immuno-modulatory effects of this important flavonoid, we performed a genome-wide expression analysis in pro-inflammatory challenged microglia treated with luteolin and conducted a phenotypic and functional characterization. METHODS: Resting and LPS-activated BV-2 microglia were treated with luteolin in various concentrations and mRNA levels of pro-inflammatory markers were determined. DNA microarray experiments and bioinformatic data mining were performed to capture global transcriptomic changes following luteolin stimulation of microglia. Extensive qRT-PCR analyses were carried out for an independent confirmation of newly identified luteolin-regulated transcripts. The activation state of luteolin-treated microglia was assessed by morphological characterization. Microglia-mediated neurotoxicity was assessed by quantifying secreted nitric oxide levels and apoptosis of 661W photoreceptors cultured in microglia-conditioned medium. RESULTS: Luteolin dose-dependently suppressed pro-inflammatory marker expression in LPS-activated microglia and triggered global changes in the microglial transcriptome with more than 50 differentially expressed transcripts. Pro-inflammatory and pro-apoptotic gene expression was effectively blocked by luteolin. In contrast, mRNA levels of genes related to anti-oxidant metabolism, phagocytic uptake, ramification, and chemotaxis were significantly induced. Luteolin treatment had a major effect on microglial morphology leading to ramification of formerly amoeboid cells associated with the formation of long filopodia. When co-incubated with luteolin, LPS-activated microglia showed strongly reduced NO secretion and significantly decreased neurotoxicity on 661W photoreceptor cultures. CONCLUSIONS: Our findings confirm the inhibitory effects of luteolin on pro-inflammatory cytokine expression in microglia. Moreover, our transcriptomic data suggest that this flavonoid is a potent modulator of microglial activation and affects several signaling pathways leading to a unique phenotype with anti-inflammatory, anti-oxidative, and neuroprotective characteristics. With the identification of several novel luteolin-regulated genes, our findings provide a molecular basis to understand the versatile effects of luteolin on microglial homeostasis. The data also suggest that luteolin could be a promising candidate to develop immuno-modulatory and neuroprotective therapies for the treatment of neurodegenerative disorders.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Gene Expression Regulation/drug effects , Luteolin/pharmacology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cells, Cultured , Computational Biology/methods , Culture Media, Conditioned/pharmacology , Cytokines/genetics , Gene Expression Profiling/methods , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Microglia/chemistry , Nitric Oxide/metabolism , Oligonucleotide Array Sequence Analysis/methods , Signal Transduction/drug effects
8.
Methods Mol Biol ; 580: 187-201, 2009.
Article in English | MEDLINE | ID: mdl-19784600

ABSTRACT

Quantitative real-time reverse-transcription (RT)-PCR is a precise and sensitive method to measure mRNA levels over a broad dynamic range. This chapter describes the quantitative transcript analysis of 41 selected lipid-related transcripts in macrophages and microglia using a novel "Lipidomic" Taqman Array. The Taqman Array results show that (1) stimulation with the liver-X-receptor and retinoid-X-receptor ligands T0901317 and 9-cis retinoic acid induces several genes of lipid metabolism, (2) lipopolysaccharide (LPS) and interferon-g (Ifn-g) strongly repress lipid-related genes, and (3) coincubation with docosahexaenoic acid dampens the repressing effect of LPS. The method described in this chapter can be used to monitor the transcriptional response of 41 dynamic "lipid" genes simultaneously in any cell type.


Subject(s)
Macrophages/metabolism , Microglia/metabolism , RNA, Messenger/genetics , Animals , Cell Line , Hydrocarbons, Fluorinated/pharmacology , Interferon-gamma/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Lipopolysaccharides/pharmacology , Mice , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Tretinoin/pharmacology
9.
J Neurochem ; 110(6): 1863-75, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19627444

ABSTRACT

Microgliosis is a common phenomenon in neurodegenerative disorders including retinal dystrophies. We performed a detailed characterization of activated microglia in the retinoschisin (Rs1h)-deficient (Rs1h(-/Y)) mouse model of inherited retinal degeneration. To visualize and isolate microglia, we crossed Rs1h(-/Y) animals with transgenic MacGreen mice, which express green fluorescent protein under the control of the macrophage-specific csf1r promoter. Activated microglia were detected in retinal sections and whole-mounts of early postnatal MacGreen/Rs1h(-/Y) mice before the onset of overt neuronal cell death. These activated microglia contained prominent lipid droplets and analysis of the retinal lipid composition showed decreased docosahexaenoic acid (DHA) levels in Rs1h(-/Y) retinas. To establish a link between microglia activation, reduced DHA levels, and neurodegeneration, a dietary intervention study was performed. Female Rs1h(-/-) mice and their Rs1h(-/Y) litter were either subjected to a diet enriched with DHA, or a control chow lacking DHA. Supplementation with DHA enhanced photoreceptor survival and converted activated microglia to a quiescent phenotype. Furthermore, DHA, but not docosapentaenoic acid or adrenic acid reduced pro-inflammatory gene expression, migration, and lipid accumulation of cultured BV-2 microglia. We conclude that retinal DHA levels control the activity of microglia and thereby may affect the progression and extent of retinal degeneration.


Subject(s)
Docosahexaenoic Acids/pharmacology , Microglia/drug effects , Retina/drug effects , Retinal Degeneration/pathology , Age Factors , Animals , Animals, Newborn , Cell Adhesion Molecules/deficiency , Cell Death/drug effects , Cell Movement/drug effects , Cells, Cultured , Dietary Supplements , Disease Models, Animal , Dose-Response Relationship, Drug , Eye Proteins , Fatty Acids/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Green Fluorescent Proteins/genetics , In Situ Nick-End Labeling/methods , Lipopolysaccharides/pharmacology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis/methods , Phospholipids/metabolism , Retina/pathology , Retinal Degeneration/genetics , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
Neuromolecular Med ; 11(2): 87-96, 2009.
Article in English | MEDLINE | ID: mdl-19365618

ABSTRACT

We have previously identified activation of microglia and induction of the early growth response gene 1 (Egr1) in the retina of retinoschisin-deficient (Rs1h(-/Y)) mice. We hypothesized that microglial expression of Egr1 might support retinal microgliosis. To test this, Egr1 transcript levels were determined in RNAs isolated from early postnatal retinas and primary microglia from Rs1h(-/Y) mice and wild-type controls. Egr1 mRNA expression was strongly induced in retinoschisin-deficient retinas as well as in ex vivo isolated microglia. Increased microglial Egr1 protein expression was concordantly detected in retinal sections of Rs1h(-/Y) mice using immunohistochemistry. Prominent activation-dependent Egr1 mRNA and protein expression was also confirmed in murine BV-2 microglia. Using binding site prediction and chromatin immunoprecipitation, we identified that the Egr1 promoter itself and the microglial marker genes Clec7a and Caspase11 are direct transcriptional targets of Egr1. Over-expression of Egr1 in BV-2 cells by adenoviral infection promoted Clec7a and Caspase11 mRNA synthesis, whereas expression of the Egr1 repressor NAB2 blocked the transcription of these genes. To analyze whether Egr1 was absolutely required for microglial marker expression in vivo, transcript levels were quantified in Rs1h(-/Y)/Egr1(-/-) retinas. No significant differences in activation marker expression could be measured in retinal tissue from Rs1h(-/Y)/Egr1(-/-) mice compared to Rs1h(-/Y) mice, suggesting that lack of Egr1 does not impair transcription of microglia genes in vivo. Taken together, our findings suggest that increased Egr1 expression is present in activated retinal microglia and contributes to their activation. However, up-regulation of Egr1 is not absolutely required for retinal microglia activation in vivo.


Subject(s)
Cell Adhesion Molecules/metabolism , Early Growth Response Protein 1/metabolism , Eye Proteins/metabolism , Microglia/metabolism , Animals , Biomarkers/metabolism , Caspases/metabolism , Caspases, Initiator , Cell Adhesion Molecules/genetics , Cells, Cultured , Early Growth Response Protein 1/genetics , Eye Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retina/cytology , Retina/metabolism
11.
Biochem Biophys Res Commun ; 379(1): 121-6, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-19100238

ABSTRACT

Activated microglia contribute to neurodegenerative processes in the brain and the retina. Via DNA-microarray analysis, we have previously identified up-regulation of several immune-related genes in the dystrophic retina of retinoschisin-deficient (Rs1h(-/Y)) mice. Here we report a strong overexpression of transcripts for the signal-transducing adaptor protein-1 (STAP-1) in isolated Rs1h(-/Y) microglia. Furthermore, STAP-1 expression was induced in activated bone marrow-derived macrophages as well as LPS-, interferon-gamma-, and CpG-stimulated myeloid cell lines. Ectopic expression of STAP-1 in BV-2 microglia changed the morphology and cytoskeletal organization of the cells and transformed ramified cells to an activated state. STAP-1 overexpression also leads to an interaction with the M-CSF receptor/c-Fms diminishing its ligand-dependent phosphorylation. Finally, STAP-1 expressing cells showed strongly reduced migration with increased cytotoxicity against 661W photoreceptor like cells. Taken together, our study implicates a previously unknown role of STAP-1 in pro-inflammatory microglia activation potentially contributing to neuronal apoptosis and degeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Microglia/metabolism , Retinitis/metabolism , Animals , Apoptosis , Cell Adhesion Molecules/genetics , Chemotaxis , Eye Proteins/genetics , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages , Mice , Mice, Mutant Strains , Microglia/drug effects , Microglia/pathology , Nitric Oxide/metabolism , Phagocytosis , Retinitis/pathology
12.
J Leukoc Biol ; 84(3): 736-40, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18550791

ABSTRACT

A disaccharide degradation product of chondrotin sulfate proteoglycan-disaccharide (CSPG-DS) has been implicated previously in the inhibition of neurodegeneration by influencing microglia activation. In this study, genome-wide microarray analysis was used to identify specific gene expression profiles of CSPG-DS-stimulated BV-2 microglia-like cells. Gene products involved in phagocytosis, detoxification, migration, immune regulation, and antigen presentation were found to be altered significantly. These findings were replicated and compared with IFN-gamma-stimulated primary microglia using real-time quantitative RT-PCR validation. Importantly, a unique transcriptional phenotype with anti-inflammatory and IFN-gamma counter-regulatory properties partially related to alternatively activated macrophages was identified. Using functional cell assays, we found that CSPG-DS-stimulated microglia possess increased phagocytic capacity but lack direct cytotoxic effects such as secretion of NO. Furthermore, conditioned media from CSPG-DS-treated microglia did not diminish the viability or cause apoptosis of cultured photoreceptor cells and partially rescued these cells from IFN-gamma-induced apoptosis. Taken together, our data provide a unique transcript dataset and important in vitro findings about the functional properties of CSPG-DS-activated microglia. These might be starting points to explore the in vivo role of CSPG-DS as a bioactive microglia regulator and its potential, therapeutic application in immune-related, neurodegenerative disorders.


Subject(s)
Biomarkers/metabolism , Chondroitin Sulfate Proteoglycans/pharmacology , Microglia/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Gene Expression Profiling , Interferon-gamma/pharmacology , Mice , Microglia/metabolism , Nitric Oxide/metabolism , Oligonucleotide Array Sequence Analysis , Phagocytosis/physiology , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
13.
J Leukoc Biol ; 82(6): 1564-74, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17827340

ABSTRACT

Several alterations in the expression of immune-related transcripts were identified recently in the degenerating retina of the retinoschisin knockout (Rs1h(-/Y)) mouse, including the strong expression of the adaptor protein Dap12. As Dap12 is found in leukocytes, we hypothesized that its disease-related expression may be confined to activated retinal microglia cells. To test this hypothesis, we established a procedure for isolation and culture of retinal microglia cells and performed genome-wide expression profiling from Rs1h(-/Y) and control microglia. While retaining their activated state in culture, ex vivo microglia expressed high levels of Dap12 and the transcription factor PU.1. The activation-dependent induction of Dap12 was also confirmed in the microglia cell line BV-2 following in vitro stimulation. To examine the transcriptional regulation of Dap12 further, macrophage cell lines were transfected with several Dap12 reporter constructs. Promoter deletion assays and site-directed mutagenesis experiments demonstrated an essential role of evolutionarily conserved PU.1 consensus sites in the proximal -104/+118 Dap12 promoter. In vitro and in vivo binding of PU.1 to this promoter region was demonstrated using EMSA and chromatin immunoprecipitation. Knockdown of PU.1 by RNA interference caused a significant reduction of endogenous Dap12 expression and re-expression, and activation of PU.1 in PU.1(-/-) progenitor cells induced Dap12 transcription. Taken together, our results indicate that activated microglia from degenerating retinae express high levels of Dap12 and PU.1, and PU.1 controls the myeloid-specific regulation of Dap12 directly and may also play a general role in microglia gene expression during retinal degeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Adhesion Molecules/deficiency , Microglia/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Retina/metabolism , Trans-Activators/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Base Sequence , Binding Sites , Cell Separation , Conserved Sequence , Evolution, Molecular , Eye Proteins , Gene Expression Profiling , Gene Expression Regulation , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myeloid Cells/metabolism , Organ Specificity , Protein Binding , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/metabolism , Retina/pathology , Stem Cells/metabolism , Trans-Activators/deficiency , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...