Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 18(9): 1793-1810, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37541258

ABSTRACT

CRB1 gene mutations can cause early- or late-onset retinitis pigmentosa, Leber congenital amaurosis, or maculopathy. Recapitulating human CRB1 phenotypes in animal models has proven challenging, necessitating the development of alternatives. We generated human induced pluripotent stem cell (iPSC)-derived retinal organoids of patients with retinitis pigmentosa caused by biallelic CRB1 mutations and evaluated them against autologous gene-corrected hiPSCs and hiPSCs from healthy individuals. Patient organoids show decreased levels of CRB1 and NOTCH1 expression at the retinal outer limiting membrane. Proximity ligation assays show that human CRB1 and NOTCH1 can interact via their extracellular domains. CRB1 patient organoids feature increased levels of WDFY1+ vesicles, fewer RAB11A+ recycling endosomes, decreased VPS35 retromer complex components, and more degradative endolysosomal compartments relative to isogenic control organoids. Taken together, our data demonstrate that patient-derived retinal organoids enable modeling of retinal degeneration and highlight the importance of CRB1 in early endosome maturation receptor recycling in the retina.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Degeneration , Retinitis Pigmentosa , Animals , Humans , Induced Pluripotent Stem Cells/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Mutation , Organoids/metabolism , Eye Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
2.
Int J Mol Sci ; 22(7)2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33808129

ABSTRACT

Mutations in the Crumbs homologue 1 (CRB1) gene cause inherited retinal dystrophies, such as early-onset retinitis pigmentosa and Leber congenital amaurosis. A Brown Norway rat strain was reported with a spontaneous insertion-deletion (indel) mutation in exon 6 of Crb1. It has been reported that these Crb1 mutant rats show vascular abnormalities associated with retinal telangiectasia and possess an early-onset retinal degenerative phenotype with outer limiting membrane breaks and focal loss of retinal lamination at 2 months of age. Here, we further characterized the morphological phenotype of new-born and adult Crb1 mutant rats in comparison with age-matched Brown Norway rats without a mutation in Crb1. A significantly decreased retinal function and visual acuity was observed in Crb1 mutant rats at 1 and 3 months of age, respectively. Moreover, in control rats, the subcellular localization of canonical CRB1 was observed at the subapical region in Müller glial cells while CRB2 was observed at the subapical region in both photoreceptors and Müller glial cells by immuno-electron microscopy. CRB1 localization was lost in the Crb1 mutant rats, whereas CRB2 was still observed. In addition, we determined the tropism of subretinal or intravitreally administered AAV5-, AAV9- or AAV6-variant ShH10Y445F vectors in new-born control and Crb1 mutant rat retinas. We showed that subretinal injection of AAV5 and AAV9 at postnatal days 5 (P5) or 8 (P8) predominantly infected the retinal pigment epithelium (RPE) and photoreceptor cells; while intravitreal injection of ShH10Y445F at P5 or P8 resulted in efficient infection of mainly Müller glial cells. Using knowledge of the subcellular localization of CRB1 and the ability of ShH10Y445F to infect Müller glial cells, canonical hCRB1 and hCRB2 AAV-mediated gene therapy were explored in new-born Crb1 mutant rats. Enhanced retinal function after gene therapy delivery in the Crb1 rat was not observed. No timely rescue of the retinal phenotype was observed using retinal function and visual acuity, suggesting the need for earlier onset of expression of recombinant hCRB proteins in Müller glial cells to rescue the severe retinal phenotype in Crb1 mutant rats.


Subject(s)
Calcium-Binding Proteins/genetics , Dependovirus/physiology , Genetic Therapy/methods , Nerve Tissue Proteins/genetics , Retinal Dystrophies/genetics , Animals , Animals, Newborn , Calcium-Binding Proteins/metabolism , Carrier Proteins/genetics , Dependovirus/genetics , Ependymoglial Cells/metabolism , Eye Proteins/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacology , Intravitreal Injections , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/metabolism , Phenotype , Rats , Rats, Mutant Strains , Retina/physiopathology , Retinal Dystrophies/etiology , Retinal Dystrophies/therapy , Retinal Pigment Epithelium/metabolism , Viral Tropism
3.
Oncotarget ; 12(5): 422-439, 2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33747358

ABSTRACT

Compelling evidence demonstrates that miR-193a-3p is a tumor suppressor microRNA in many cancer types, and its reduced expression is linked to cancer initiation and progression, metastasis, and therapy resistance. However, its mechanism of action is not consistently described between studies, and often contradicts the pleiotropic role of a microRNA in manipulating several different mRNA targets. We therefore comprehensively investigated miRNA-193a-3p's mode of action in a panel of human cancer cell lines, with a variety of genetic backgrounds, using 1B3, a synthetic microRNA mimic. Interestingly, the exact mechanism through which 1B3 reduced cell proliferation varied between cell lines. 1B3 efficiently reduced target gene expression, leading to reduced cell proliferation/survival, cell cycle arrest, induction of apoptosis, increased cell senescence, DNA damage, and inhibition of migration. SiRNA silencing of 1B3 target mRNAs further highlighted the advantage of the pleiotropic mechanism of 1B3 action, as repression of individual targets did not achieve the same robust effect on cell proliferation in all cell lines. Importantly, a novel lipid nanoparticle-based formulation of 1B3, INT-1B3, demonstrated marked anti-tumor activity as a single agent following systemic administration in tumor-bearing mice. Together, these data strongly support the development of 1B3 as a novel therapeutic agent for treatment of human cancer.

4.
Mol Ther Nucleic Acids ; 23: 1161-1171, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33664995

ABSTRACT

Emerging data show that microRNA 193a-3p (miR-193a-3p) has a suppressive role in many cancers and is often downregulated in tumors, as compared to surrounding normal tissues. Therefore, mimics of miR-193a-3p could be used as an attractive therapeutic approach in oncology. To better understand and document the molecular mechanism of action of 1B3, a novel synthetic miRNA-193a-3p mimic, RNA sequencing was performed after transfection of 1B3 in six different human tumor cell lines. Genes differentially expressed (DE) in at least three cell lines were mapped by Ingenuity Pathway Analysis (IPA), and interestingly, these results strongly indicated upregulation of the tumor-suppressive phosphatase and tensin homolog (PTEN) pathway, as well as downregulation of many oncogenic growth factor signaling pathways. Importantly, although unsurprisingly, IPA identified miR-193a-3p as a strong upstream regulator of DE genes in an unbiased manner. Furthermore, biological function analysis pointed to an extensive link of 1B3 with cancer, via expected effects on tumor cell survival, proliferation, migration, and cell death. Our data strongly suggest that miR-193a-3p/1B3 is a potent tumor suppressor agent that targets various key oncogenic pathways across cancer types. Therefore, the introduction of 1B3 into tumor cells may represent a promising strategy for cancer treatment.

5.
Mol Ther Methods Clin Dev ; 20: 423-441, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33575434

ABSTRACT

Loss of Crumbs homolog 1 (CRB1) or CRB2 proteins in Müller cells or photoreceptors in the mouse retina results in a CRB dose-dependent retinal phenotype. In this study, we present a novel Müller cell-specific Crb1 KO Crb2 LowMGC retinitis pigmentosa mouse model (complete loss of CRB1 and reduced levels of CRB2 specifically in Müller cells). The Crb double mutant mice showed deficits in electroretinography, optokinetic head tracking, and retinal morphology. Exposure of retinas to low levels of dl-α-aminoadipate acid induced gliosis and retinal disorganization in Crb1 KO Crb2 LowMGC retinas but not in wild-type or Crb1-deficient retinas. Crb1 KO Crb2 LowMGC mice showed a substantial decrease in inner/outer photoreceptor segment length and optokinetic head-tracking response. Intravitreal application of rAAV vectors expressing human CRB2 (hCRB2) in Müller cells of Crb1 KO Crb2 LowMGC mice subsequently exposed to low levels of dl-α-aminoadipate acid prevented loss of vision, whereas recombinant adeno-associated viral (rAAV) vectors expressing human CRB1 (hCRB1) did not. Both rAAV vectors partially protected the morphology of the retina. The results suggest that hCRB expression in Müller cells is vital for control of retinal cell adhesion at the outer limiting membrane, and that the rAAV-cytomegalovirus (CMV)-hCRB2 vector is more potent than rAAV-minimal CMV (CMVmin)-hCRB1 in protection against loss of vision.

6.
Stem Cell Reports ; 12(5): 906-919, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30956116

ABSTRACT

Human retinal organoids from induced pluripotent stem cells (hiPSCs) can be used to confirm the localization of proteins in retinal cell types and to test transduction and expression patterns of gene therapy vectors. Here, we compared the onset of CRB protein expression in human fetal retina with human iPSC-derived retinal organoids. We show that CRB2 protein precedes the expression of CRB1 in the developing human retina. Our data suggest the presence of CRB1 and CRB2 in human photoreceptors and Müller glial cells. Thus the fetal CRB complex formation is replicated in hiPSC-derived retina. CRB1 patient iPSC retinal organoids showed disruptions at the outer limiting membrane as found in Crb1 mutant mice. Furthermore, AAV serotype 5 (AAV5) is potent in infecting human Müller glial cells and photoreceptors in hiPSC-derived retinas and retinal explants. Our data suggest that human photoreceptors can be efficiently transduced by AAVs in the presence of photoreceptor segments.


Subject(s)
Carrier Proteins/metabolism , Ependymoglial Cells/metabolism , Eye Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Organoids/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Adult , Carrier Proteins/genetics , Cells, Cultured , Dependovirus/genetics , Ependymoglial Cells/cytology , Ependymoglial Cells/ultrastructure , Eye Proteins/genetics , Female , Fetus , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/cytology , Membrane Proteins/genetics , Microscopy, Immunoelectron , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/genetics , Organoids/cytology , Photoreceptor Cells, Vertebrate/ultrastructure , Pregnancy , Retina/cytology , Retina/embryology
7.
Methods Mol Biol ; 1715: 275-288, 2018.
Article in English | MEDLINE | ID: mdl-29188521

ABSTRACT

This protocol details on a screening method for infectivity and tropism of different serotypes of adeno-associated viruses (AAVs) on human retinal explants with cell-type specific or ubiquitous green fluorescent protein (GFP) expression vectors. Eyes from deceased adult human donors are enucleated and the retinas are isolated. Each retina is punched into eight to ten 6-mm equal pieces. Whatman™ paper punches are placed on the retinas and the stack is transferred onto 24-well culture inserts with the photoreceptors facing the membrane. AAVs are applied on the retinal explant punches to allow transduction for 48 h. Retinas are nourished by a serum-free Neurobasal®-A based medium composition that allows extended culturing of explants containing photoreceptor inner and outer segments. The protocols include quality control measurements and histological staining for retina cells. The cost and time effective procedure permits AAV transgene expression assays, RNAi knockdown, and pharmacological intervention on human retinas for 21 days ex vivo.


Subject(s)
Dependovirus/genetics , Genetic Vectors , Green Fluorescent Proteins/metabolism , Injections, Intraocular/methods , Organ Culture Techniques/methods , Retina/metabolism , Transduction, Genetic , Dependovirus/immunology , Humans , Photoreceptor Cells/metabolism , Serogroup , Transgenes
8.
Hum Mol Genet ; 24(11): 3104-18, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25701872

ABSTRACT

Mutations in the Crumbs-homologue-1 (CRB1) gene lead to severe recessive inherited retinal dystrophies. Gene transfer therapy is the most promising cure for retinal dystrophies and has primarily been applied for recessive null conditions via a viral gene expression vector transferring a cDNA encoding an enzyme or channel protein, and targeting expression to one cell type. Therapy for the human CRB1 disease will be more complex, as CRB1 is a structural and signaling transmembrane protein present in three cell classes: Müller glia, cone and rod photoreceptors. In this study, we applied CRB1 and CRB2 gene therapy vectors in Crb1-retinitis pigmentosa mouse models at mid-stage disease. We tested if CRB expression restricted to Müller glial cells or photoreceptors or co-expression in both is required to recover retinal function. We show that targeting both Müller glial cells and photoreceptors with CRB2 ameliorated retinal function and structure in Crb1 mouse models. Surprisingly, targeting a single cell type or all cell types with CRB1 reduced retinal function. We show here the first pre-clinical studies for CRB1-related eye disorders using CRB2 vectors and initial elucidation of the cellular mechanisms underlying CRB1 function.


Subject(s)
Ependymoglial Cells/physiology , Nerve Tissue Proteins/genetics , Retinitis Pigmentosa/genetics , Animals , Carrier Proteins/genetics , Disease Models, Animal , Genetic Therapy , HEK293 Cells , Humans , Intravitreal Injections , Membrane Proteins/genetics , Mice, Inbred C57BL , Retina/pathology , Retina/physiopathology , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/therapy
9.
Hum Mol Genet ; 23(14): 3759-71, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24565864

ABSTRACT

Mutations in the CRB1 gene lead to retinal dystrophies ranging from Leber congenital amaurosis (LCA) to early-onset retinitis pigmentosa (RP), due to developmental defects or loss of adhesion between photoreceptors and Müller glia cells, respectively. Whereas over 150 mutations have been found, no clear genotype-phenotype correlation has been established. Mouse Crb1 knockout retinas show a mild phenotype limited to the inferior quadrant, whereas Crb2 knockout retinas display a severe degeneration throughout the retina mimicking the phenotype observed in RP patients associated with CRB1 mutations. Crb1Crb2 double mutant retinas have severe developmental defects similar to the phenotype observed in LCA patients associated with CRB1 mutations. Therefore, CRB2 is a candidate modifying gene of human CRB1-related retinal dystrophy. In this study, we studied the cellular localization of CRB1 and CRB2 in human retina and tested the influence of the Crb2 gene allele on Crb1-retinal dystrophies in mice. We found that in contrast to mice, in the human retina CRB1 protein was expressed at the subapical region in photoreceptors and Müller glia cells, and CRB2 only in Müller glia cells. Genetic ablation of one allele of Crb2 in heterozygote Crb1(+/-) retinas induced a mild retinal phenotype, but in homozygote Crb1 knockout mice lead to an early and severe phenotype limited to the entire inferior retina. Our data provide mechanistic insight for CRB1-related LCA and RP.


Subject(s)
Carrier Proteins/metabolism , Ependymoglial Cells/metabolism , Eye Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Retinal Dystrophies/metabolism , Adult , Aged , Aged, 80 and over , Animals , Carrier Proteins/genetics , Disease Models, Animal , Eye Proteins/genetics , Gene Knockout Techniques , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Middle Aged , Nerve Tissue Proteins/genetics , Photoreceptor Cells/metabolism
10.
Hum Mol Genet ; 23(13): 3384-401, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24493795

ABSTRACT

In humans, the Crumbs homolog-1 (CRB1) gene is mutated in autosomal recessive Leber congenital amaurosis and early-onset retinitis pigmentosa. In mammals, the Crumbs family is composed of: CRB1, CRB2, CRB3A and CRB3B. Recently, we showed that removal of mouse Crb2 from retinal progenitor cells, and consequent removal from Müller glial and photoreceptor cells, results in severe and progressive retinal degeneration with concomitant loss of retinal function that mimics retinitis pigmentosa due to mutations in the CRB1 gene. Here, we studied the effects of cell-type-specific loss of CRB2 from the developing mouse retina using targeted conditional deletion of Crb2 in photoreceptors or Müller cells. We analyzed the consequences of targeted loss of CRB2 in the adult mouse retina using adeno-associated viral vectors encoding Cre recombinase and short hairpin RNA against Crb2. In vivo retinal imaging by means of optical coherence tomography on retinas lacking CRB2 in photoreceptors showed progressive thinning of the photoreceptor layer and cellular mislocalization. Electroretinogram recordings under scotopic conditions showed severe attenuation of the a-wave, confirming the degeneration of photoreceptors. Retinas lacking CRB2 in developing photoreceptors showed early onset of abnormal lamination, whereas retinas lacking CRB2 in developing Müller cells showed late onset retinal disorganization. Our data suggest that in the developing retina, CRB2 has redundant functions in Müller glial cells, while CRB2 has essential functions in photoreceptors. Our data suggest that short-term loss of CRB2 in adult mouse photoreceptors, but not in Müller glial cells, causes sporadic loss of adhesion between photoreceptors and Müller cells.


Subject(s)
Membrane Proteins/metabolism , Photoreceptor Cells/metabolism , Retinitis Pigmentosa/etiology , Retinitis Pigmentosa/metabolism , Animals , Ependymoglial Cells/metabolism , Female , Immunohistochemistry , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Retinitis Pigmentosa/genetics
11.
Mol Ther Methods Clin Dev ; 1: 14009, 2014.
Article in English | MEDLINE | ID: mdl-26015954

ABSTRACT

Despite their physiological roles, Müller glial cells are involved directly or indirectly in retinal disease pathogenesis and are an interesting target for therapeutic approaches for retinal diseases and regeneration such as CRB1 inherited retinal dystrophies. In this study, we characterized the efficiency of adeno-associated virus (AAV) capsid variants and different promoters to drive protein expression in Müller glial cells. ShH10Y and AAV9 were the most powerful capsids to infect mouse Müller glial cells. Retinaldehyde-binding protein 1 (RLBP1) promoter was the most powerful promoter to transduce Müller glial cells. ShH10Y capsids and RLBP1 promoter targeted human Müller glial cells in vitro. We also developed and tested smaller promoters to express the large CRB1 gene via AAV vectors. Minimal cytomegalovirus (CMV) promoter allowed expression of full-length CRB1 protein in Müller glial cells. In summary, ShH10Y and AAV9 capsids, and RLBP1 or minimal CMV promoters are of interest as specific tools to target and express in mouse or human Müller glial cells.

12.
PLoS Genet ; 9(12): e1003976, 2013.
Article in English | MEDLINE | ID: mdl-24339791

ABSTRACT

Development in the central nervous system is highly dependent on the regulation of the switch from progenitor cell proliferation to differentiation, but the molecular and cellular events controlling this process remain poorly understood. Here, we report that ablation of Crb1 and Crb2 genes results in severe impairment of retinal function, abnormal lamination and thickening of the retina mimicking human Leber congenital amaurosis due to loss of CRB1 function. We show that the levels of CRB1 and CRB2 proteins are crucial for mouse retinal development, as they restrain the proliferation of retinal progenitor cells. The lack of these apical proteins results in altered cell cycle progression and increased number of mitotic cells leading to an increased number of late-born cell types such as rod photoreceptors, bipolar and Müller glia cells in postmitotic retinas. Loss of CRB1 and CRB2 in the retina results in dysregulation of target genes for the Notch1 and YAP/Hippo signaling pathways and increased levels of P120-catenin. Loss of CRB1 and CRB2 result in altered progenitor cell cycle distribution with a decrease in number of late progenitors in G1 and an increase in S and G2/M phase. These findings suggest that CRB1 and CRB2 suppress late progenitor pool expansion by regulating multiple proliferative signaling pathways.


Subject(s)
Central Nervous System/metabolism , Leber Congenital Amaurosis/genetics , Membrane Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Retina/growth & development , Animals , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Proliferation , Central Nervous System/growth & development , Central Nervous System/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Humans , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mitosis/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Retina/cytology , Retina/metabolism , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Stem Cells/metabolism
13.
PLoS One ; 8(12): e82532, 2013.
Article in English | MEDLINE | ID: mdl-24324803

ABSTRACT

In humans, the Crumbs homologue-1 (CRB1) gene is mutated in progressive types of autosomal recessive retinitis pigmentosa and Leber congenital amaurosis. The severity of the phenotype due to human CRB1 or mouse Crb1 mutations is dependent on the genetic background. Mice on C57BL/6J background with Crb1 mutations show late onset of retinal spotting phenotype or no phenotype. Recently, we showed that conditional deletion of mouse Crb2 in the retina results in early retinal disorganization leading to severe and progressive retinal degeneration with concomitant visual loss that mimics retinitis pigmentosa due to mutations in the CRB1 gene. Recent studies in the fruit fly and zebrafish suggest roles of the Crumbs (CRB) complex members in the regulation of cellular signalling pathways including the Notch1, mechanistic target of rapamycin complex 1 (mTORC1) and the Hippo pathway. Here, we demonstrate that mice backcrossed to C57BL/6J background with loss of CRB2 in the retina show a progressive disorganization and degeneration phenotype during late retinal development. We used microarray gene profiling to study the transcriptome of retinas lacking CRB2 during late retinal development. Unexpectedly, the retinas of newborn mice lacking CRB2 showed no changes in the transcriptome during retinal development. These findings suggest that loss of CRB2 in the developing retina results in retinal disorganization and subsequent degeneration without major changes in the transcriptome of the retina. These mice might be an interesting model to study the onset of retinal degeneration upon loss of CRB proteins.


Subject(s)
Membrane Proteins/genetics , Mutation , Retina/metabolism , Retina/pathology , Animals , Animals, Newborn , Cluster Analysis , Gene Expression Profiling , Gene Order , Gene Targeting , Gliosis/genetics , Gliosis/metabolism , Gliosis/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Multigene Family , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Signal Transduction
14.
PLoS One ; 5(8): e12387, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20808778

ABSTRACT

BACKGROUND: Müller cell gliosis occurs in various retinal pathologies regardless of the underlying cellular defect. Because activated Müller glial cells span the entire retina and align areas of injury, they are ideal targets for therapeutic strategies, including gene therapy. METHODOLOGY/PRINCIPAL FINDINGS: We used adeno-associated viral AAV2/6 vectors to transduce mouse retinas. The transduction pattern of AAV2/6 was investigated by studying expression of the green fluorescent protein (GFP) transgene using scanning-laser ophthalmoscopy and immuno-histochemistry. AAV2/6 vectors transduced mouse Müller glial cells aligning the retinal blood vessels. However, the transduction capacity was hindered by the inner limiting membrane (ILM) and besides Müller glial cells, several other inner retinal cell types were transduced. To obtain Müller glial cell-specific transgene expression, the cytomegalovirus (CMV) promoter was replaced by the glial fibrillary acidic protein (GFAP) promoter. Specificity and activation of the GFAP promoter was tested in a mouse model for retinal gliosis. Mice deficient for Crumbs homologue 1 (CRB1) develop gliosis after light exposure. Light exposure of Crb1(-/-) retinas transduced with AAV2/6-GFAP-GFP induced GFP expression restricted to activated Müller glial cells aligning retinal blood vessels. CONCLUSIONS/SIGNIFICANCE: Our experiments indicate that AAV2 vectors carrying the GFAP promoter are a promising tool for specific expression of transgenes in activated glial cells.


Subject(s)
Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Neuroglia/metabolism , Retinal Vessels/cytology , Transduction, Genetic/methods , Adenoviridae/genetics , Animals , Gene Expression , Humans , Injections , Mice , Promoter Regions, Genetic/genetics , Transgenes/genetics
15.
Mol Cell Neurosci ; 41(2): 156-65, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19233278

ABSTRACT

At the presynaptic plasma membrane of the photoreceptor the correct localization of the calcium extruder, plasma membrane Ca2+-ATPase (PMCA), is determined by a unique protein complex. Here, the role of two proteins within the complex; membrane palmitoylated protein 4 (MPP4) and postsynaptic density protein 95 (PSD95) is investigated in more details, using Mpp4 and Psd95 mutant mice. MPP4 deficiency results in the loss of both PMCA and PSD95 from the photoreceptor synapse. Truncation of the C-terminal part of MPP4 leads to a loss of PSD95 and mislocalization of PMCA, while truncation of the C-terminal part of PSD95 did not affect the localization of the complex members. Lentivirus-mediated molecular replacement strategy was used to selectively express either PSD95alpha or PSD95beta in wild type or Mpp4 mutant primary retinal explants. Silencing of the Psd95 gene resulted in the loss of presynaptic MPP4 and PMCA1. The plasma membrane localization of MPP4 and PMCA1 could be restored by the expression of PSD95beta. We conclude that both scaffold proteins PSD95beta and MPP4 are essential for the modulation of PMCA levels at the presynaptic plasma membrane and thereby influence the photoreceptor synaptic calcium handling.


Subject(s)
Cell Membrane/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Isoenzymes/metabolism , Membrane Proteins/metabolism , Photoreceptor Cells , Plasma Membrane Calcium-Transporting ATPases/metabolism , Synapses/metabolism , Animals , Disks Large Homolog 4 Protein , Guanylate Kinases , Intracellular Signaling Peptides and Proteins/genetics , Isoenzymes/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , Plasma Membrane Calcium-Transporting ATPases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retina/cytology , Retina/metabolism , Synapses/ultrastructure
16.
Glia ; 55(14): 1486-97, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17705196

ABSTRACT

Mutations in the human Crumbs homologue-1 (CRB1) gene cause retinal blinding diseases, such as Leber congenital amaurosis and retinitis pigmentosa. In the previous studies we have shown that Crb1 resides in retinal Müller glia cells and that loss of Crb1 results in retinal degeneration (particularly in the inferior temporal quadrant of the mouse eye). Degeneration is increased by exposure to white light. Here, we studied the role of light and aging to gain a better understanding of the factors involved in the progress of retinal disease. Our data reveal that light is neither sufficient nor required to induce retinal disorganization and degeneration in young Crb1(-/-) mutant mice, suggesting that it rather modulates the retinal phenotype. Gene expression profiling showed that expression of five genes is altered in light-exposed Crb1(-/-) mutant retinas. Three of the five genes are involved in chromosome stabilization (Pituitary tumor transforming gene 1 or Pttg1, Establishment of cohesion 1 homolog 1 or Esco1, and a gene similar to histone H2B). In aged retinas, degeneration of photoreceptors, inner retinal neurons, and retinal pigment epithelium was practically limited to the inferior temporal quadrant. Loss of Crb1 in Müller glia cells resulted in an irregular number and size of their apical villi. We propose that Crb1 is required to regulate number and size of these Müller glia cell villi. The subsequent loss of retinal integrity resulted in neovascularization, in which blood vessels of the choroid protruded into the neural retina.


Subject(s)
Aging/metabolism , Neovascularization, Pathologic/genetics , Nerve Tissue Proteins/genetics , Neuroglia/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Aging/genetics , Aging/pathology , Animals , Gene Expression Profiling , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Light/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Microvilli/metabolism , Microvilli/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Neuroglia/pathology , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/metabolism , Optic Atrophy, Hereditary, Leber/physiopathology , Photic Stimulation/adverse effects , Photoreceptor Cells/metabolism , Photoreceptor Cells/pathology , Photoreceptor Cells/physiopathology , Retina/pathology , Retina/physiopathology , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/physiopathology
17.
Atherosclerosis ; 192(2): 283-90, 2007 Jun.
Article in English | MEDLINE | ID: mdl-16938301

ABSTRACT

Accumulation of lipid-laden macrophages is a hallmark of atherosclerosis. The relevance of the key transcription factor nuclear factor kappaB (NF-kappaB) for macrophage-derived foam-cell formation has not been unequivocally resolved. Transgenic mice lines were generated in which NF-kappaB activation is specifically inhibited in macrophages by overexpressing a trans-dominant, non-degradable form of IkappaBalpha (IkappaBalpha (32A/36A)) under control of the macrophage-specific SR-A promoter. Alanine substitution of serines 32 and 36 prevents degradation and retains the inactive NF-kappaB/IkappaBalpha (32A/36A) complex in the cytoplasm. Similarly, stable human THP1 monocytic cell lines were generated with integrated copies of IkappaBalpha (32A/36A) cDNA. Upon treatment with oxidized low-density lipoprotein (ox-LDL), murine peritoneal macrophages from transgenic IkappaBalpha (32A/36A) mice, as well as THP1/IkappaBalpha (32A/36A) clones, display decreased lipid loading after differentiation into macrophages. This is accompanied by increased expression of the transcription factors PPARgamma and LXRalpha as well as of the major cholesterol-efflux transporter ABCA1. Paradoxically, mRNA expression of the 'lipid-uptake' receptor CD36 is also increased. Since the net result of these changes is reduction of foam-cell formation, it is proposed that under specific inhibition of NF-kappaB activation, ABCA1-mediated cholesterol efflux prevails over CD36-mediated lipid influx.


Subject(s)
Foam Cells/physiology , Macrophages/drug effects , NF-kappa B/antagonists & inhibitors , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/biosynthesis , Animals , CD36 Antigens/biosynthesis , Cell Line , DNA-Binding Proteins/biosynthesis , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/physiology , Lipoproteins, LDL/metabolism , Liver X Receptors , Macrophages/physiology , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , Orphan Nuclear Receptors , PPAR gamma/biosynthesis , Receptors, Cytoplasmic and Nuclear/biosynthesis , Scavenger Receptors, Class A/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...