Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1244765, 2023.
Article in English | MEDLINE | ID: mdl-38016059

ABSTRACT

Sirtuin 6 (SIRT6) is a member of the mammalian sirtuin family of NAD+-dependent protein deacylases, homologues of the yeast silent information regulator 2 (Sir2). SIRT6 has remarkably diverse functions and plays a key role in a variety of biological processes for maintaining cellular and organismal homeostasis. In this review, our primary aim is to summarize recent progress in understanding SIRT6's functions in the retina and retinal pigment epithelium (RPE), with the hope of further drawing interests in SIRT6 to increase efforts in exploring the therapeutic potential of this unique protein in the vision field. Before describing SIRT6's role in the eye, we first discuss SIRT6's general functions in a wide range of biological contexts. SIRT6 plays an important role in gene silencing, metabolism, DNA repair, antioxidant defense, inflammation, aging and longevity, early development, and stress response. In addition, recent studies have revealed SIRT6's role in macrophage polarization and mitochondrial homeostasis. Despite being initially understudied in the context of the eye, recent efforts have begun to elucidate the critical functions of SIRT6 in the retina and RPE. In the retina, SIRT6 is essential for adult retinal function, regulates energy metabolism by suppressing glycolysis that affects photoreceptor cell survival, protects retinal ganglion cells from oxidative stress, and plays a role in Müller cells during early neurodegenerative events in diabetic retinopathy. In the RPE, SIRT6 activates autophagy in culture and protects against oxidative stress in mice. Taken together, this review demonstrates that better understanding of SIRT6's functions and their mechanisms, both in and out of the context of the eye, holds great promise for the development of SIRT6-targeted strategies for prevention and treatment of blinding eye diseases.

2.
Cells ; 12(8)2023 04 13.
Article in English | MEDLINE | ID: mdl-37190063

ABSTRACT

Epithelial-mesenchymal transition (EMT), which is well known for its role in embryonic development, malignant transformation, and tumor progression, has also been implicated in a variety of retinal diseases, including proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), and diabetic retinopathy. EMT of the retinal pigment epithelium (RPE), although important in the pathogenesis of these retinal conditions, is not well understood at the molecular level. We and others have shown that a variety of molecules, including the co-treatment of human stem cell-derived RPE monolayer cultures with transforming growth factor beta (TGF-ß) and the inflammatory cytokine tumor necrosis factor alpha (TNF-α), can induce RPE-EMT; however, small molecule inhibitors of RPE-EMT have been less well studied. Here, we demonstrate that BAY651942, a small molecule inhibitor of nuclear factor kapa-B kinase subunit beta (IKKß) that selectively targets NF-κB signaling, can modulate TGF-ß/TNF-α-induced RPE-EMT. Next, we performed RNA-seq studies on BAY651942 treated hRPE monolayers to dissect altered biological pathways and signaling events. Further, we validated the effect of IKKß inhibition on RPE-EMT-associated factors using a second IKKß inhibitor, BMS345541, with RPE monolayers derived from an independent stem cell line. Our data highlights the fact that pharmacological inhibition of RPE-EMT restores RPE identity and may provide a promising approach for treating retinal diseases that involve RPE dedifferentiation and EMT.


Subject(s)
Retinal Pigment Epithelium , Vitreoretinopathy, Proliferative , Humans , Retinal Pigment Epithelium/metabolism , Epithelial-Mesenchymal Transition , I-kappa B Kinase/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Vitreoretinopathy, Proliferative/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/metabolism , Protein Serine-Threonine Kinases/metabolism , Stem Cells/metabolism
3.
Life Sci Alliance ; 6(7)2023 07.
Article in English | MEDLINE | ID: mdl-37185874

ABSTRACT

Retinal pigment epithelium (RPE) is essential for the survival of retinal photoreceptors. To study retinal degeneration, sodium iodate (NaIO3) has been used to cause oxidative stress-induced RPE death followed by photoreceptor degeneration. However, analyses of RPE damage itself are still limited. Here, we characterized NaIO3-induced RPE damage, which was divided into three regions: periphery with normal-shaped RPE, transitional zone with elongated cells, and center with severely damaged or lost RPE. Elongated cells in the transitional zone exhibited molecular characteristics of epithelial-mesenchymal transition. Central RPE was more susceptible to stresses than peripheral RPE. Under stresses, SIRT6, an NAD+-dependent protein deacylase, rapidly translocated from the nucleus to the cytoplasm and colocalized with stress granule factor G3BP1, leading to nuclear SIRT6 depletion. To overcome this SIRT6 depletion, SIRT6 overexpression was induced in the nucleus in transgenic mice, which protected RPE from NaIO3 and partially preserved catalase expression. These results demonstrate topological differences of mouse RPE and warrant further exploring SIRT6 as a potential target for protecting RPE from oxidative stress-induced damage.


Subject(s)
Retinal Degeneration , Sirtuins , Mice , Animals , Retinal Pigment Epithelium/metabolism , DNA Helicases/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Retinal Degeneration/metabolism , Oxidative Stress , Sirtuins/genetics , Sirtuins/adverse effects , Sirtuins/metabolism
4.
Cell Rep ; 37(3): 109866, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34686321

ABSTRACT

Epithelial-mesenchymal transition (EMT) of the retinal pigment epithelium (RPE) is associated with several blinding retinal diseases. Using proteomics and phosphoproteomics studies of human induced pluripotent stem cell-derived RPE monolayers with induced EMT, we capture kinase/phosphatase signaling cascades 1 h and 12 h after induction to better understand the pathways mediating RPE EMT. Induction by co-treatment with transforming growth factor ß and tumor necrosis factor alpha (TGNF) or enzymatic dissociation perturbs signaling in many of the same pathways, with striking similarity in the respective phosphoproteomes at 1 h. Liver hyperplasia and hepatocyte growth factor (HGF)-MET signaling exhibit the highest overall enrichment. We also observe that HGF and epidermal growth factor signaling, two cooperative pathways inhibited by EMT induction, regulate the RPE transcriptional profile.


Subject(s)
Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Induced Pluripotent Stem Cells/metabolism , Liver/metabolism , Proteome , Proteomics , Retinal Pigment Epithelium/metabolism , Cell Line , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Hyperplasia , Induced Pluripotent Stem Cells/drug effects , Liver/pathology , Phosphorylation , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Retinal Pigment Epithelium/drug effects , Signal Transduction , Transcriptome , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
5.
Antioxidants (Basel) ; 11(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35052607

ABSTRACT

Oxidative stress of the retinal pigment epithelium (RPE) is a major risk factor for age-related macular degeneration (AMD). As a dry AMD model via oxidative stress, sodium iodate (NaIO3), which is primarily toxic to the RPE, has often been used at a high dose to cause RPE death for studying photoreceptor degeneration. Thus, characterization of RPE damage by a low dose of NaIO3 is still limited. To quantify RPE damage caused by NaIO3 in mice, we recently developed a morphometric method using RPE flat-mounts. Here, we report that NaIO3 has a narrow range of dose-effect correlation at 11-18 mg/kg body weight in male C57BL/6J mice. We evaluated the usefulness of our quantification method in two experimental settings. First, we tested the effect of NF-κB inhibition on NaIO3-induced RPE damage in male C57BL/6J mice. IKKß inhibitor BAY 651942 suppressed upregulation of NF-κB targets and protected the RPE from oxidative stress. Second, we tested sex-specific differences in NaIO3-induced RPE damage in C57BL/6J mice using a low dose near the threshold. NaIO3 caused more severe RPE damage in female mice than in male mice. These results demonstrate the usefulness of the quantification method and the importance of fine-tuning of the NaIO3 dose. The results also show the therapeutic potential of IKKß inhibition for oxidative stress-related RPE diseases, and reveal previously-unrecognized sex-specific differences in RPE susceptibility to oxidative stress.

6.
PLoS One ; 13(1): e0191279, 2018.
Article in English | MEDLINE | ID: mdl-29338041

ABSTRACT

The retinal pigment epithelium (RPE) supports the health and function of retinal photoreceptors and is essential for normal vision. RPE cells are post-mitotic, terminally differentiated, and polarized epithelial cells. In pathological conditions, however, they lose their epithelial integrity, become dysfunctional, even dedifferentiate, and ultimately die. The integrity of epithelial cells is maintained, in part, by adherens junctions, which are composed of cadherin homodimers and p120-, ß-, and α-catenins linking to actin filaments. While E-cadherin is the major cadherin for forming the epithelial phenotype in most epithelial cell types, it has been reported that cadherin expression in RPE cells is different from other epithelial cells based on results with cultured RPE cells. In this study, we revisited the expression of cadherins in the RPE to clarify their relative contribution by measuring the absolute quantity of cDNAs produced from mRNAs of three classical cadherins (E-, N-, and P-cadherins) in the RPE in vivo. We found that P-cadherin (CDH3) is highly dominant in both mouse and human RPE in situ. The degree of dominance of P-cadherin is surprisingly large, with mouse Cdh3 and human CDH3 accounting for 82-85% and 92-93% of the total of the three cadherin mRNAs, respectively. We confirmed the expression of P-cadherin protein at the cell-cell border of mouse RPE in situ by immunofluorescence. Furthermore, we found that oxidative stress induces dissociation of P-cadherin and ß-catenin from the cell membrane and subsequent translocation of ß-catenin into the nucleus, resulting in activation of the canonical Wnt/ß-catenin pathway. This is the first report of absolute comparison of the expression of three cadherins in the RPE, and the results suggest that the physiological role of P-cadherin in the RPE needs to be reevaluated.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Retinal Pigment Epithelium/metabolism , Adherens Junctions/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cdh1 Proteins/genetics , Cdh1 Proteins/metabolism , Cells, Cultured , Choroid/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Epithelial-Mesenchymal Transition , Gene Expression , Humans , Mice , Oxidative Stress , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Pigment Epithelium/cytology , beta Catenin/metabolism
7.
J Biol Chem ; 289(18): 12908-21, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24634209

ABSTRACT

The retinal pigment epithelium (RPE) performs specialized functions to support retinal photoreceptors, including regeneration of the visual chromophore. Enzymes and carrier proteins in the visual cycle function sequentially to regenerate and continuously supply 11-cis-retinal to retinal photoreceptor cells. However, it is unknown how the expression of the visual cycle genes is coordinated at the transcriptional level. Here, we show that the proximal upstream regions of six visual cycle genes contain chromatin-accessible sex-determining region Y box (SOX) binding sites, that SOX9 and LIM homeobox 2 (LHX2) are coexpressed in the nuclei of mature RPE cells, and that SOX9 acts synergistically with orthodenticle homeobox 2 (OTX2) to activate the RPE65 and retinaldehyde binding protein 1 (RLBP1) promoters and acts synergistically with LHX2 to activate the retinal G protein-coupled receptor (RGR) promoter. ChIP reveals that SOX9 and OTX2 bind to the promoter regions of RPE65, RLBP1, and RGR and that LHX2 binds to those of RPE65 and RGR in bovine RPE. ChIP with human fetal RPE cells shows that SOX9 and OTX2 also bind to the human RPE65, RLBP1, and RGR promoters. Conditional inactivation of Sox9 in mouse RPE results in reduced expression of several visual cycle genes, most dramatically Rpe65 and Rgr. Furthermore, bioinformatic analysis predicts that multiple common microRNAs (miRNAs) regulate visual cycle genes, and cotransfection of miRNA mimics with luciferase reporter constructs validated some of the predicted miRNAs. These results implicate SOX9 as a key regulator of visual cycle genes, reveal for the first time the functional role of LHX2 in the RPE, and suggest the possible regulation of visual cycle genes by common miRNAs.


Subject(s)
Eye Proteins/genetics , Gene Expression Regulation , Retinal Pigment Epithelium/metabolism , SOX9 Transcription Factor/physiology , Animals , Binding Sites/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Eye Proteins/metabolism , Gene Regulatory Networks , HEK293 Cells , Humans , Immunohistochemistry , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , Models, Genetic , Otx Transcription Factors/genetics , Otx Transcription Factors/metabolism , Retinal Pigment Epithelium/cytology , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , cis-trans-Isomerases/genetics , cis-trans-Isomerases/metabolism
8.
Mol Vis ; 19: 1371-86, 2013.
Article in English | MEDLINE | ID: mdl-23805044

ABSTRACT

PURPOSE: Ras-like without CAAX 2 (RIT2), a member of the Ras superfamily of small guanosine triphosphatases, is involved in regulating neuronal function. RIT2 is a unique member of the Ras family in that RIT2 is preferentially expressed in various neurons, including retinal neurons. The mechanisms that regulate RIT2 expression in neurons were studied. METHODS: Reverse transcription-quantitative PCR (RT-qPCR), immunohistochemistry, western blotting, bioinformatic prediction, electrophoretic mobility shift assay (EMSA), and cell transfection methods were used. RESULTS: With immunohistochemistry of the mouse retina, RIT2 protein was detected in the ganglion cell layer (GCL), inner plexiform layer, inner nuclear layer, and outer plexiform layer, with the strongest staining in the GCL and the inner plexiform layer. RT-qPCR combined with laser capture microdissection detected Rit2 messenger RNA in the GCL and the inner nuclear layer. Western blot analysis showed a large increase in the RIT2 protein in the retina during maturation from newborn to adult. Transient transfection identified the 1.3 kb upstream region of human RIT2 as capable of driving expression in neuronal cell lines. Based on the known expression pattern and biological activity, we hypothesized that POU4 family factors might modulate RIT2 expression in retinal ganglion cells (RGCs). Bioinformatic analyses predicted six POU4 factor-binding sites within the 1.3 kb human RIT2 promoter region. EMSA analyses showed binding of POU4 proteins to three of the six predicted sites. Cotransfection with expression vectors demonstrated that POU4 proteins can indeed modulate the human RIT2 promoter, and that ISL1, a LIM homeodomain factor, can further modulate the activity of the POU4 factors. CONCLUSIONS: These studies confirm the expression of RIT2 in retinal neuronal cells, including RGCs, begin to reveal the mechanisms responsible for neuronal expression of RIT2, and suggest a role for the POU4 family factors in modulating RIT2 expression in RGCs.


Subject(s)
Glycoproteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , POU Domain Factors/metabolism , Promoter Regions, Genetic/genetics , Retinal Neurons/enzymology , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Computational Biology , Gene Expression Regulation , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Monomeric GTP-Binding Proteins/genetics , Organ Specificity , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Curr Gene Ther ; 13(4): 275-81, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23773177

ABSTRACT

Erythropoietin (EPO) can protect the retina from acute damage, but long-term systemic treatment induces polycythemia. Intraocular gene delivery of EPO is not protective despite producing high levels of EPO likely due to its bellshaped dose curve. The goal of this study was to identify a therapeutic dose of continuously produced EPO in the eye. We packaged a mutated form of EPO (EPOR76E) that has equivalent neuroprotective activity as wild-type EPO and attenuated erythropoietic activity into a recombinant adeno-associated viral vector under the control of the tetracycline inducible promoter. This vector was injected into the subretinal space of homozygous postnatal 5-7 day retinal degeneration slow mice, that express the tetracycline transactivators from a retinal pigment epithelium specific promoter. At weaning, mice received a single intraperitoneal injection of doxycycline and were then maintained on water with or without doxycycline until postnatal day 60. Intraocular EPO levels and outer nuclear layer thickness were quantified and correlated. Control eyes contained 6.1 ± 0.1 (SEM) mU/ml EPO. The eyes of mice that received an intraperitoneal injection of doxycycline contained 11.8 ± 2.0 (SEM) mU/ml EPO-R76E. Treatment with doxycycline water induced production of 35.9 ± 2.4 (SEM) mU/ml EPO-R76E in the eye. The outer nuclear layer was approximately 8 µm thicker in eyes of mice that received doxycycline water as compared to the control groups. Our data indicates that drug delivery systems should be optimized to deliver at least 36 mU/ml EPO into the eye since this dose was effective for the treatment of a progressive retinal degeneration.


Subject(s)
Drug Delivery Systems , Erythropoietin/administration & dosage , Gene Transfer Techniques , Retinal Degeneration , Animals , Erythropoietin/genetics , Genetic Therapy , Genetic Vectors/therapeutic use , Humans , Mice , Promoter Regions, Genetic/genetics , Retina/drug effects , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/therapy
11.
Invest Ophthalmol Vis Sci ; 52(3): 1378-83, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21212186

ABSTRACT

PURPOSE: To generate and characterize a constitutively active, RPE-specific, cre-expressing transgenic mouse line. This line can be used to create RPE-specific knockouts by crossing with mice harboring loxP-flanked (floxed) genes. METHODS: A transgene construct was assembled with the BEST1 promoter driving cre expression. Transgenic mice were generated on a C57BL/6 background. Cre expression was assessed by immunofluorescence and Western blot analysis. Cre enzymatic activity was tested by crossing to three lines with floxed DNA regions and detecting deletion of the intervening sequences or through histochemical detection of lacZ activity. Potential cre-mediated toxicity was assessed by retinal histology up to 24 months of age and by electroretinography. RESULTS: The BEST1-cre line with expression in the highest percentage of RPE cells displayed a patchy mosaic expression pattern, with 50% to 90% of RPE cells expressing cre. In mice outcrossed to a mixed B6/129 background, expression was consistently found in 90% of RPE cells. Within the eye, only the RPE cells were immunoreactive with an anti-cre antibody. Maximum cre expression quantified by Western blot analysis occurred at P28. Crosses with three lines containing floxed sequences revealed RPE-specific cre activity in the eye and extraocular expression limited to the testes. Histology and electroretinography showed no cre-mediated RPE toxicity. CONCLUSIONS: This BEST1-cre transgenic line enables generation of RPE-specific knockout mice. The mosaic expression pattern provides an internal control; the non-cre-expressing RPE cells continue to express the floxed genes. These mice should facilitate study of the multifunctional RPE and the generation of mouse models of human retinal disease.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Integrases/genetics , Retinal Pigment Epithelium/enzymology , Animals , Bestrophins , Blotting, Western , Electroretinography , Eye Proteins/genetics , Female , Fluorescent Antibody Technique, Indirect , Ion Channels/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics
12.
J Biol Chem ; 285(35): 26933-26944, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20530484

ABSTRACT

BEST1 is highly and preferentially expressed in the retinal pigment epithelium (RPE) and causes Best macular dystrophy when mutated. We previously demonstrated that the human BEST1 upstream region -154 to +38 bp is sufficient to direct expression in the RPE of transgenic mice, and microphthalmia-associated transcription factor (MITF) and OTX2 regulate this BEST1 promoter. However, a number of questions remained. Here, we show that yeast one-hybrid screen with bait corresponding to BEST1 -120 to -88 bp identified the SOX-E factors, SOX8, SOX9, and SOX10. A paired SOX site was found in this bait, and mutation of either of the paired sites significantly decreased BEST1 promoter activity in RPE primary cultures. Among the SOX-E genes, SOX9 is highly and preferentially expressed in the RPE, and chromatin immunoprecipitation with fresh RPE cells revealed binding of SOX9, but not SOX10, to the BEST1 region where the paired SOX site is located. BEST1 promoter activity was increased by SOX9 overexpression and decreased by siRNA-mediated SOX9 knockdown. Importantly, SOX9 physically interacted with MITF and OTX2 and orchestrated synergistic activation of the BEST1 promoter with the paired SOX site playing essential roles. A combination of the expression patterns of SOX9, MITF, and OTX2 yielded tissue distribution remarkably similar to that of BEST1. Lastly, the BEST1 promoter was also active in Sertoli cells of the testis in transgenic mice where SOX9 is highly expressed. These results define SOX9 as a key regulator of BEST1 expression and demonstrate for the first time its functional role in the RPE.


Subject(s)
Chloride Channels/biosynthesis , Eye Proteins/biosynthesis , Gene Expression Regulation/physiology , Microphthalmia-Associated Transcription Factor/metabolism , Otx Transcription Factors/metabolism , Response Elements/physiology , Retinal Pigment Epithelium/metabolism , SOX9 Transcription Factor/metabolism , Animals , Bestrophins , Cell Line, Tumor , Chloride Channels/genetics , Eye Proteins/genetics , Humans , Ion Channels , Male , Mice , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/genetics , Otx Transcription Factors/genetics , Retinal Pigment Epithelium/cytology , SOX9 Transcription Factor/genetics , Sertoli Cells/cytology , Sertoli Cells/metabolism , Swine
13.
Hum Mol Genet ; 18(1): 128-41, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18849347

ABSTRACT

A number of genes preferentially expressed in the retinal pigment epithelium (RPE) are associated with retinal degenerative disease. One of these, BEST1, encodes bestrophin-1, a protein that when mutated causes Best macular dystrophy. As a model for RPE gene regulation, we have been studying the mechanisms that control BEST1 expression, and recently demonstrated that members of the MITF-TFE family modulate BEST1 transcription. The human BEST1 upstream region from -154 to +38 bp is sufficient to direct expression in the RPE, and positive-regulatory elements exist between -154 and -104 bp. Here, we show that the -154 to -104 bp region is necessary for RPE expression in transgenic mice and contains a predicted OTX-binding site (Site 1). Since another non-canonical OTX site (Site 2) is located nearby, we tested the function of these sites using BEST1 promoter/luciferase constructs by in vivo electroporation and found that mutation of both sites reduces promoter activity. Three OTX family proteins - OTX1, OTX2 and CRX - bound to both Sites 1 and 2 in vitro, and all of them increased BEST1 promoter activity. Surprisingly, we found that human and bovine RPE expressed not only OTX2 but also CRX, the CRX genomic region in bovine RPE was hypersensitive to DNase I, consistent with active transcription, and that both OTX2 and CRX bound to the BEST1 proximal promoter in vivo. These results demonstrate for the first time CRX expression in the RPE, and suggest that OTX2 and CRX may act as positive modulators of the BEST1 promoter in the RPE.


Subject(s)
Chloride Channels/genetics , Eye Proteins/genetics , Gene Expression Regulation , Homeodomain Proteins/metabolism , Otx Transcription Factors/metabolism , Retinal Pigment Epithelium/metabolism , Trans-Activators/metabolism , Animals , Bestrophins , Binding Sites , Cattle , Cell Line , Chloride Channels/metabolism , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Eye Proteins/metabolism , Genes, Reporter , Homeodomain Proteins/genetics , Humans , Mice , Mice, Transgenic , Multigene Family , Otx Transcription Factors/genetics , Promoter Regions, Genetic , Trans-Activators/genetics
14.
Invest Ophthalmol Vis Sci ; 49(3): 1248-53, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18326755

ABSTRACT

PURPOSE: The retinal pigmented epithelium (RPE) expresses many genes that play important roles in the support and maintenance of photoreceptors. The present study was conducted to develop a system amenable to the dissection of the temporal function of these genes, specifically within RPE cells. Transgenic mice were generated and characterized in which the expression of Cre recombinase could be specifically induced within the RPE. METHODS: Transgenic mice carrying the human vitelliform macular dystrophy-2 (VMD2) promoter (P(VMD2))-directed reverse tetracycline-dependent transactivator (rtTA) and the tetracycline-responsive element (TRE)-directed cre were generated. Inducible Cre expression was achieved by feeding doxycycline to these mice and was characterized by using a Cre-activatable lacZ reporter mouse strain (R26R). RESULTS: A beta-galactosidase assay of rtTA/Cre-R26R mice demonstrated that the basal level of Cre expression without doxycycline induction was negligible. Addition of doxycycline led to induction of RPE-specific Cre expression/function at least from embryonic day 9 to postnatal day 60. The highest induction occurred at approximately postnatal day 4. As measured by ERG and histology, retinal function and morphology were normal in 10-month-old rtTA/Cre mice that were treated with doxycycline at weaning age. CONCLUSIONS: Transgenic mice were generated that express Cre recombinase in the RPE in an inducible fashion. These mice will be useful for studies of the RPE-specific role of genes that are expressed in the RPE as well as other cells, particularly for avoiding embryonic lethality and dissecting the function of genes that play dual roles in development and adulthood.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Integrases/genetics , Pigment Epithelium of Eye/enzymology , Animals , Bestrophins , Chloride Channels/genetics , Doxycycline/administration & dosage , Electroretinography , Eye Proteins/genetics , Mice , Mice, Transgenic , Pigment Epithelium of Eye/drug effects , Plasmids , Promoter Regions, Genetic/genetics , beta-Galactosidase/metabolism
15.
J Biol Chem ; 282(3): 1838-50, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17085443

ABSTRACT

The retinal pigment epithelium (RPE) is crucial for the function and survival of retinal photoreceptors. VMD2 encodes bestrophin, an oligomeric chloride channel that is preferentially expressed in the RPE and, when mutated, causes Best macular dystrophy. Previously, we defined the VMD2 upstream region from -253 to +38 bp as being sufficient to direct RPE-specific expression in the eye, and we suggested microphthalmia-associated transcription factor (MITF) as a possible positive regulator. Here we show that in transgenic mice the -154 to +38 bp region is sufficient for RPE expression, and mutation of two E-boxes, 1 and 2, within this region leads to loss of promoter activity. A yeast one-hybrid screen using bait containing E-box 1 identified clones encoding MITF, TFE3, and TFEB, and chromatin immunoprecipitation with antibodies against these proteins enriched the VMD2 proximal promoter. Analysis using in vivo electroporation with constructs containing mutation of each E-box indicated that expression in native RPE requires both E-boxes, yet in vitro DNA binding studies suggested that MITF binds well to E-box 1 but only minimally to E-box 2. MITF knockdown by small interfering RNA (siRNA) in cell culture revealed a strong correlation between MITF and VMD2 mRNA levels. Sequential transfection of a luciferase construct with expression vectors following MITF siRNA revealed that TFE3 and TFEB can also transactivate the VMD2 promoter. Taken together, we suggest that VMD2 is regulated by the MITF-TFE family through two E-boxes, with E-box 1 required for a direct interaction of MITF-TFE factors and E-box 2 for binding of the as yet unidentified factor(s).


Subject(s)
Eye Proteins/genetics , Eye Proteins/physiology , Gene Expression Regulation , Microphthalmia-Associated Transcription Factor/physiology , Promoter Regions, Genetic , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Bestrophins , DNA-Binding Proteins/chemistry , Ion Channels , Mice , Mice, Transgenic , Mutation , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Transcriptional Activation
16.
Nucleic Acids Res ; 34(3): 917-27, 2006.
Article in English | MEDLINE | ID: mdl-16464824

ABSTRACT

Combinatorial regulation by transcription factor complexes is an important feature of eukaryotic gene regulation. Here, we propose a new method for identification of interactions between transcription factors (TFs) that relies on the relationship of their binding sites, and we test it using Saccharomyces cerevisiae as a model system. The algorithm predicts interacting TF pairs based on the co-occurrence of their binding motifs and the distance between the motifs in promoter sequences. This allows investigation of interactions between TFs without known binding motifs or expression data. With this approach, 300 significant interactions involving 77 TFs were identified. These included more than 70% of the known protein-protein interactions. Approximately half of the detected interacting motif pairs showed strong preferences for particular distances and orientations in the promoter sequences. These one dimensional features may reflect constraints on allowable spatial arrangements for protein-protein interactions. Evidence for biological relevance of the observed characteristic distances is provided by the finding that target genes with the same characteristic distances show significantly higher co-expression than those without preferred distances. Furthermore, the observed interactions were dynamic: most of the TF pairs were not constitutively active, but rather showed variable activity depending on the physiological condition of the cells. Interestingly, some TF pairs active in multiple conditions showed preferences for different distances and orientations depending on the condition. Our prediction and characterization of TF interactions may help to understand the transcriptional regulatory networks in eukaryotic systems.


Subject(s)
Genomics/methods , Protein Interaction Mapping/methods , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transcription Factors/metabolism , Algorithms , Binding Sites , Genome, Fungal , Promoter Regions, Genetic , Saccharomyces cerevisiae/metabolism
17.
Nucleic Acids Res ; 33(11): 3479-91, 2005.
Article in English | MEDLINE | ID: mdl-15967807

ABSTRACT

Identification of tissue-specific gene regulatory networks can yield insights into the molecular basis of a tissue's development, function and pathology. Here, we present a computational approach designed to identify potential regulatory target genes of photoreceptor cell-specific transcription factors (TFs). The approach is based on the hypothesis that genes related to the retina in terms of expression, disease and/or function are more likely to be the targets of retina-specific TFs than other genes. A list of genes that are preferentially expressed in retina was obtained by integrating expressed sequence tag, SAGE and microarray datasets. The regulatory targets of retina-specific TFs are enriched in this set of retina-related genes. A Bayesian approach was employed to integrate information about binding site location relative to a gene's transcription start site. Our method was applied to three retina-specific TFs, CRX, NRL and NR2E3, and a number of potential targets were predicted. To experimentally assess the validity of the bioinformatic predictions, mobility shift, transient transfection and chromatin immunoprecipitation assays were performed with five predicted CRX targets, and the results were suggestive of CRX regulation in 5/5, 3/5 and 4/5 cases, respectively. Together, these experiments strongly suggest that RP1, GUCY2D, ABCA4 are novel targets of CRX.


Subject(s)
Computational Biology/methods , Eye Proteins/metabolism , Gene Expression Regulation , Retina/metabolism , Transcription Factors/metabolism , Animals , Basic-Leucine Zipper Transcription Factors , Bayes Theorem , Binding Sites , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Humans , Mice , Orphan Nuclear Receptors , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Analysis, DNA , Trans-Activators/metabolism
18.
J Cell Physiol ; 201(3): 393-400, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15389527

ABSTRACT

Increased expression of vascular endothelial cell growth factor (VEGF) in the retina is sufficient to stimulate sprouting of neovascularization from the deep capillary bed of the retina, but not the superficial retinal capillaries or the choriocapillaris. Coexpression of VEGF and angiopoietin 2 (Ang2) results in sprouting of neovascularization from superficial and deep retinal capillaries, but not the choriocapillaris. However, retina-derived VEGF and Ang2 may not reach the choriocapillaris, because of tight junctions between retinal pigmented epithelial (RPE) cells. To eliminate this possible confounding factor, we used the human vitelliform macular dystrophy 2 (VMD2) promoter, an RPE-specific promoter, combined with the tetracycline-inducible promoter system, to generate double transgenic mice with inducible expression of VEGF in RPE cells. Adult mice with increased expression of VEGF in RPE cells had normal retinas and choroids with no choroidal neovascularization (CNV), but when increased expression of VEGF in RPE cells was combined with subretinal injection of a gutless adenoviral vector containing an expression construct for Ang2 (AGVAng2), CNV consistently occurred. In contrast, triple transgenic mice with induced expression of Ang2 and VEGF in RPE cells, did not develop CNV. These data suggest that increased expression of VEGF and/or Ang2 in RPE cells is not sufficient to cause CNV unless it is combined with a subretinal injection of a gutless adenoviral vector, which is likely to perturb RPE cells. These data also suggest that the effects of angiogenic proteins may vary among vascular beds, even those that are closely related, and, therefore, generalizations should be avoided.


Subject(s)
Choroidal Neovascularization/metabolism , Pigment Epithelium of Eye/metabolism , Retina/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Angiopoietin-2/pharmacology , Animals , Bestrophins , Chloride Channels , Choroidal Neovascularization/genetics , Doxycycline/pharmacology , Eye Proteins/genetics , Gene Expression/drug effects , Humans , Mice , Mice, Transgenic , Pigment Epithelium of Eye/drug effects , Promoter Regions, Genetic/genetics , Retina/drug effects , Tetracycline/pharmacology , Vascular Endothelial Growth Factor A/genetics
19.
Hum Mol Genet ; 13(10): 1025-40, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15028672

ABSTRACT

A novel paired-like homeobox gene, designated as Qrx, was identified by a yeast one-hybrid screen using the bovine Rhodopsin promoter Ret-1 DNA regulatory element as bait. Qrx is preferentially expressed in both the outer and inner nuclear layers of the retina. Its homeodomain is nearly identical to that of Rx/Rax, a transcription factor that is essential for eye development, but it shares only limited homology elsewhere. Although Qrx and Rx/Rax show similar DNA binding properties in vitro, the two proteins demonstrate distinct target selectivity and functional behavior in promoter activity assays. QRX synergistically increases the transactivating function of the photoreceptor transcription factors Crx and NRL and it physically interacts with CRX. Qrx is present in the bovine and human genomes, but appears to be absent from the mouse genome. Nonetheless, a 5.8 kb upstream region of human QRX is capable of directing expression in presumptive photoreceptor precursor cells in transgenic mice. These results indicate that Qrx may be involved in modulating photoreceptor gene expression. In addition, the finding of rare heterozygous QRX sequence changes in three individuals with retinal degeneration raises the possibility that QRX may be involved in disease pathogenesis.


Subject(s)
Gene Expression Regulation/genetics , Genes, Homeobox/genetics , Rhodopsin/genetics , Amino Acid Sequence , Animals , Basic-Leucine Zipper Transcription Factors , Cattle , Chromosome Mapping , Chromosomes, Human, Pair 9/genetics , Coatomer Protein/genetics , DNA Footprinting , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Molecular Sequence Data , Point Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Response Elements/genetics , Retinal Diseases/genetics , Sequence Alignment , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
20.
J Biol Chem ; 279(18): 19064-73, 2004 Apr 30.
Article in English | MEDLINE | ID: mdl-14982938

ABSTRACT

The retinal pigment epithelium (RPE) is crucial for the normal development and function of retinal photo-receptors, and mutations in several genes that are preferentially expressed in the RPE have been shown to cause retinal degeneration. We analyzed the 5'-up-stream region of human VMD2, a gene that is preferentially expressed in the RPE and, when mutated, causes Best macular dystrophy. Transgenic mouse studies with VMD2 promoter/lacZ constructs demonstrated that a-253 to +38 bp fragment is sufficient to direct RPE-specific expression in the eye. Transient transfection assays using the D407 human RPE cell line with VMD2 promoter/luciferase reporter constructs identified two positive regulatory regions, -585 to -541 bp for high level expression and -56 to -42 bp for low level expression. Mutation of a canonical E-box located in the -56 to -42 bp region greatly diminished luciferase expression in D407 cells and abolished the bands shifted with bovine RPE nuclear extract in electrophoretic mobility shift assays. Independently a candidate approach was used to select microphthalmia-associated transcription factor (MITF) for testing because it is expressed in the RPE and associated with RPE abnormalities when mutated. MITF-M significantly increased luciferase expression in D407 cells in an E-box-dependent manner. These studies define the VMD2 promoter region sufficient to drive RPE-specific expression in the eye, identify positive regulatory regions in vitro, and suggest that MITF as well as other E-box binding factors may act as positive regulators of VMD2 expression.


Subject(s)
E-Box Elements/physiology , Eye Proteins/genetics , Gene Expression Regulation , Promoter Regions, Genetic , 5' Flanking Region , Animals , Base Sequence , Bestrophins , Cattle , Chloride Channels , DNA-Binding Proteins/physiology , Eye/cytology , Humans , Ion Channels , Mice , Mice, Transgenic , Microphthalmia-Associated Transcription Factor , Molecular Sequence Data , Pigment Epithelium of Eye/metabolism , RNA/isolation & purification , Transcription Factors/physiology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...