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










Publication year range
1.
J Mol Endocrinol ; 46(1): 9-19, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20978111

ABSTRACT

The bicoid-like transcription factor PITX2 has been previously described to interact with the pituitary-specific POU homeodomain factor POU1F1 (human ortholog of PIT-1) to achieve cell-specific expression of prolactin (PRL) and GH in pituitary somatolactotroph cells. In this work, we have investigated the functional properties of three PITX2 mutants reported in Axenfeld-Rieger syndrome patients relative to the regulation of these genes, using reporter genes under the control of human PRL (hPRL), hGH, or POU1F1 promoters transfected in nonpituitary and pituitary cell lines. Among the three mutations studied, Y167X and E101X introduce a premature stop codon, and F104L leads to an amino acid substitution. While PITX2(E101X) is not expressed in the cells following transfection, and PITX2(F104L) is functionally inactive, the PITX2(Y167X) mutant keeps its DNA-binding capacity and displays a markedly enhanced activation of the hPRL and POU1F1 promoters, but not of the hGH promoter. Y167X is the first mutation of PITX2 described to result in a differential effect on the activation of its different physiological targets, hPRL and POU1F1 on one hand and hGH on the other hand. The differential effect of the Y167X mutation might be linked to an interaction of PITX2 with different transcription factors or cofactors when bound to the hPRL and POU1F1 or the hGH promoters. These results might form the basis for the identification of the PITX2 protein complex necessary for the differential GH or PRL expression.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Human Growth Hormone/genetics , Prolactin/genetics , Promoter Regions, Genetic , Transcription Factor Pit-1/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Anterior Eye Segment/abnormalities , Blotting, Western , Cell Line , Electrophoretic Mobility Shift Assay , Eye Abnormalities/genetics , Eye Diseases, Hereditary , Human Growth Hormone/metabolism , Humans , Mutation , Pituitary Gland/metabolism , Prolactin/metabolism , Transcription Factor Pit-1/metabolism , Transcriptional Activation , Homeobox Protein PITX2
2.
Mol Vis ; 15: 1139-52, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19503746

ABSTRACT

PURPOSE: The retina is highly exposed to oxidative stress due to the high level of oxygen consumption in this tissue and its exposure to light. The main DNA base lesion generated by oxygen free radicals is 8-oxoguanine (8-oxoG). However, its presence in retinal cells and the mechanisms underlying its repair remain undetermined. METHODS: 8-oxoguanine DNA glycosylase (Ogg1) gene expression and messenger localization in adult mouse ocular tissues was analyzed by RT-PCR and in situ hybridization. Using immunohistochemistry, we determined the localization of Ogg1 protein and three base excision repair (BER) enzymes: apurinic/apyrimidic endonuclease (APE1), DNA polymerase beta, and X-ray repair cross-complementation group 1 (XRCC1). Ogg1 and AP-lyase activities in the neuroretina were obtained using double-stranded oligonucleotides harboring either an 8-oxoG residue or a tetrahydrofuran. RESULTS: We report here that 8-oxoG is abundant in the retina. Ogg1, the enzyme responsible for the recognition and excision of the oxidized base, is present in its active form and found mainly in ganglion cells and photoreceptor inner segments. We show that APE1 and DNA polymerase beta, two BER proteins involved in 8-oxoG repair, are also present in these cells. The cellular distribution of these proteins was similar to that of Ogg1. XRRC1 is present in both inner nuclear and ganglion cells layers; however, this protein is absent from photoreceptor inner segments. CONCLUSIONS: This is the first study to demonstrate the presence of a functional 8-oxoG BER pathway in retinal neurons. The study of three BER proteins involved in 8-oxoG elimination demonstrates that XRCC1 localization differs from those of Ogg1, APE1, and DNA polymerase beta. This result suggests that the elimination of 8-oxoG is coordinated through two pathways, which differ slightly according to the cellular localization of the abnormally oxidized guanine.


Subject(s)
DNA Glycosylases/genetics , DNA Glycosylases/metabolism , Gene Expression , Retina/metabolism , Analysis of Variance , Animals , DNA Polymerase beta/genetics , DNA Polymerase beta/metabolism , DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eye/metabolism , Gene Expression Profiling/methods , Immunohistochemistry , In Situ Hybridization , Mice , Polymerase Chain Reaction , X-ray Repair Cross Complementing Protein 1
3.
Invest Ophthalmol Vis Sci ; 50(8): 3562-72, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19407027

ABSTRACT

PURPOSE: Sirtuin1 (Sirt1) is an NAD(+)-dependent deacetylase involved in development, cell survival, stress resistance, energy metabolism, and aging. It is expressed in the mammalian central nervous system (CNS) and is activated during processes associated with neuroprotection. The retinal degeneration 10 (rd10) mouse model of retinitis pigmentosa (RP) was used to investigate the possible role of Sirt1 in this type of retinal degeneration. METHODS: Eyes from control and rd10 mice were used. Sirt1 mRNA was detected by in situ hybridization, and its abundance was estimated by semiquantitative RT-PCR. The presence of Sirt1 protein was investigated by immunohistofluorescence and Western blot analysis. The apoptosis of photoreceptor cells was analyzed by terminal dUTP transferase nick-end labeling (TUNEL). Immunolabeling for Sirt1, apoptosis-inducing factor (Aif), and caspase-12 (Casp-12) was performed on retinal tissue sections. RESULTS: Sirt1 mRNA and immunoreactivity were observed in normal adult mouse eyes. In the control retina, Sirt1 was immunolocalized mostly to the nucleus. In rd10 mice with retinal degeneration, changes in Sirt1 immunolabeling were observed only in the retinal outer nuclear layer (ONL). The pathologic pattern of Sirt1 immunoreactivity correlated with the start of retinal degeneration in rd10 mice. CONCLUSIONS: The results suggest a link between Sirt1 production and retinal degeneration in rd10 mice. The anti-apoptotic, neuroprotective role of Sirt1 in the mouse retina is based on the involvement of Sirt1 in double DNA strand-break repair mechanisms and in maintaining energy homeostasis in photoreceptor cells. The results suggest that the neuroprotective properties of Sirt1 may gradually weaken in rd10 mouse photoreceptor cells.


Subject(s)
Retina/metabolism , Retinitis Pigmentosa/metabolism , Sirtuins/physiology , Animals , Apoptosis , Apoptosis Inducing Factor/metabolism , Blotting, Western , Caspase 12/metabolism , DNA Probes , Disease Models, Animal , Fluorescent Antibody Technique, Indirect , Gene Expression/physiology , In Situ Hybridization , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Confocal , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , RNA, Messenger/metabolism , Retina/pathology , Retinitis Pigmentosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1
4.
Invest Ophthalmol Vis Sci ; 50(8): 3931-42, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19420333

ABSTRACT

PURPOSE: To evaluate functional and ultrastructural changes in the retina of scavenger receptor B1 (SR-BI) knockout (KO) mice consuming a high fat cholate (HFC) diet. METHODS: Three-month-old male KO and wild-type (WT) mice were fed an HFC diet for 30 weeks. After diet supplementation, plasma cholesterol levels and electroretinograms were analyzed. Neutral lipids were detected with oil red O, and immunohistochemistry was performed on cryostat ocular tissue sections. The retina, Bruch's membrane (BM), retinal pigment epithelium (RPE), and choriocapillaris (CC) were analyzed by transmission electron microscopy. RESULTS: Using the WT for reference, ultrastructural changes were recorded in HFC-fed SR-BI KO mice, including lipid inclusions, a patchy disorganization of the photoreceptor outer segment (POS) and the outer nuclear layer (ONL), and BM thickening with sparse sub-RPE deposits. Within the CC, there was abnormal disorganization of collagen fibers localized in ectopic sites with sparse and large vacuolization associated with infiltration of macrophages in the subretinal space, reflecting local inflammation. These lesions were associated with electroretinographic abnormalities, particularly increasing implicit time in a- and b-wave scotopic responses. Abnormal vascular endothelial growth factor (VEGF) staining was detected in the outer nuclear layer. CONCLUSIONS: HFC-fed SR-BI KO mice thus presented sub-RPE lipid-rich deposits and functional and morphologic alterations similar to some features observed in dry AMD. The findings lend further support to the hypothesis that atherosclerosis causes retinal and subretinal damage that increases susceptibility to some forms of AMD.


Subject(s)
Coronary Artery Disease/pathology , Diet, Atherogenic , Hypercholesterolemia/pathology , Macular Degeneration/pathology , Retina/physiology , Retina/ultrastructure , Scavenger Receptors, Class B/physiology , Animals , Bruch Membrane/ultrastructure , Cholates/administration & dosage , Cholesterol/blood , Choroid/blood supply , Coronary Artery Disease/etiology , Coronary Artery Disease/metabolism , Electroretinography , Hypercholesterolemia/etiology , Hypercholesterolemia/metabolism , Macular Degeneration/etiology , Macular Degeneration/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Retinal Pigment Epithelium/ultrastructure , Vascular Endothelial Growth Factor A/metabolism
5.
Mol Vis ; 15: 393-416, 2009.
Article in English | MEDLINE | ID: mdl-19234633

ABSTRACT

PURPOSE: High levels of metabolism and oxygen consumption in most adult murine ocular compartments, combined with exposure to light and ultraviolet (UV) radiation, are major sources of oxidative stress, causing DNA damage in ocular cells. Of all mammalian body cells, photoreceptor cells consume the largest amount of oxygen and generate the highest levels of oxidative damage. The accumulation of such damage throughout life is a major factor of aging tissues. Several multiprotein complexes have recently been identified as the major sensors and mediators involved in the maintenance of DNA integrity. The activity of these complexes initially seemed to be restricted to dividing cells, given their ultimate role in major cell cycle checkpoints. However, it was later established that they are also active in post-mitotic cells. Recent findings demonstrate that the DNA damage response (DDR) is essential for the development, maintenance, and normal functioning of the adult central nervous system. One major molecular factor in the DDR is the protein, ataxia telangiectasia mutated (ATM). It is required for the rapid induction of cellular responses to DNA double-strand breaks. These cytotoxic DNA lesions may be caused by oxidative damage. To understand how ATM prevents oxidative stress and participates in the maintenance of genomic integrity and cell viability of the adult retina, we determined the ATM expression patterns and studied its localization in the adult mouse eye. METHODS: Atm gene expression was analyzed by RT-PCR experiments and its localization by in situ hybridization on adult mouse ocular and cerebellar tissue sections. ATM protein expression was determined by western blot analysis of proteins homogenates extracted from several mouse tissues and its localization by immunohistochemistry experiments performed on adult mouse ocular and cerebellar tissue sections. In addition, subcellular localization was realized by confocal microscopy imaging of ocular tissue sections, with a special focus on retinal cells. RESULTS: Using RT-PCR, we detected a band of the expected size, with its sequence matching the amplified Atm cDNA sequence. Atm mRNA was detected in most cell bodies of the adult mouse eye by in situ hybridization of ocular tissue sections with specific digoxigenin-labeled PCR-amplified cDNA probes. Western blotting with different specific antibodies revealed bands corresponding to the expected sizes of ATM and its active forms (ATMp). These bands were not observed in the analysis of protein homogenates from Atm-deficient mouse tissues. ATM immunoreactivity was detected in the nucleus of all adult mice retinal cells and in most non-neuronal ocular cell types. The active phosphorylated form of ATM was also present in the retina as well as in non-neuronal cells of the adult mouse eye. However, its subcellular localization differed as a function of the cell type examined. A major finding of this study was that ATMp immunostaining in photoreceptor cells was exclusively in the cytoplasm, whereas ATM immunostaining was only in the nucleus of these cells. Furthermore, the specific and distinct ATM and ATMp immunolabeling patterns in photoreceptor cells were identical to those observed in the adult mouse cerebellar granule cells. CONCLUSIONS: We report the expression profile of Atm gene and protein in the adult mouse eye. In particular, we observed a difference between the localization patterns of the active and inactive forms of ATM in photoreceptor cells. These localization patterns suggest that ATM and its phosphorylated activated form may be involved in both the protection of cells from oxidative damage and the maintenance of ocular cell structure and function. The protection mechanisms mediated by the two forms of ATM appear to be particularly important in maintaining photoreceptor integrity.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Eye/metabolism , Gene Expression , Protein Serine-Threonine Kinases/genetics , Retina/metabolism , Tumor Suppressor Proteins/genetics , 8-Hydroxy-2'-Deoxyguanosine , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cerebellum/cytology , Cerebellum/metabolism , Ciliary Body/cytology , Ciliary Body/metabolism , Cornea/cytology , Cornea/metabolism , Cytoplasm/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Eye/cytology , Histones/metabolism , Immunohistochemistry , In Situ Hybridization , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Oxidative Stress , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/cytology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/metabolism
6.
Stroke ; 38(12): 3237-44, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17975099

ABSTRACT

BACKGROUND AND PURPOSE: Retinal ischemia is a major cause of visual impairment and is associated with a high risk of subsequent ischemic stroke. The retina and its projections are easily accessible for experimental procedures and functional evaluation. We created and characterized a mouse model of global and transient retinal ischemia and provide a comprehensive chronologic profile of some genes that display altered expression during ischemia. METHODS: Ischemia and reperfusion were assessed by observing flat-mounted retinas after systemic fluorescein injection. The temporal pattern of gene expression modulation was evaluated by quantitative reverse transcription-polymerase chain reaction from the occurrence of unilateral 30-minute pterygopalatine artery occlusion until 4 weeks after reperfusion. Electroretinograms evaluated functional sequelae 4 weeks after the ischemic episode and were correlated with histologic lesions. RESULTS: This model is the first to reproduce the features of transient monocular amaurosis fugax resulting from ophthalmic artery occlusion. The histologic structure was roughly conserved, but functional lesions affected ganglion cells, inner nuclear layer cells, and photoreceptor cells. We observed an early and strong upregulation of c-fos, c-jun, Cox-2, Hsp70, and Gadd34 gene expression and a late decrease in Hsp70 transcript levels. CONCLUSIONS: A murine model of transient retinal ischemia was successfully developed that exhibited the characteristic upregulation of immediate-early genes and persistent functional deficits. The model should prove useful for investigating mechanisms of injury in genetically altered mice and for testing novel neuroprotective drugs.


Subject(s)
Amaurosis Fugax/diagnosis , Amaurosis Fugax/genetics , Gene Expression Regulation , Retinal Diseases/diagnosis , Animals , Antigens, Differentiation/physiology , Carotid Artery, Internal/pathology , Cell Cycle Proteins/physiology , Disease Models, Animal , Electroretinography/methods , Ischemia/pathology , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Protein Phosphatase 1 , Rats , Reperfusion Injury , Retina/pathology , Time Factors
8.
Mol Vis ; 13: 511-23, 2007 Apr 02.
Article in English | MEDLINE | ID: mdl-17417613

ABSTRACT

PURPOSE: The PAX6 gene was first described as a candidate for human aniridia. However, PAX6 expression is not restricted to the eye and it appears to be crucial for brain development. We studied PAX6 mutations in a large spectrum of patients who presented with aniridia phenotypes, Peters' anomaly, and anterior segment malformations associated or not with neurological anomalies. METHODS: Patients and related families were ophthalmologically phenotyped, and in some cases neurologically and endocrinologically examined. We screened the PAX6 gene by direct sequencing in three groups of patients: those affected by aniridia; those with diverse ocular manifestations; and those with Peters' anomaly. Two mutations were investigated by generating crystallographic representations of the amino acid changes. RESULTS: Three novel heterozygous mutations affecting three unrelated families were identified: the g.572T>C nucleotide change, located in exon 5, and corresponding to the Leucine 46 Proline amino-acid mutation (L46P); the g.655A>G nucleotide change, located in exon 6, and corresponding to the Serine 74 Glycine amino-acid mutation (S74G); and the nucleotide deletion 579delG del, located in exon 6, which induces a frameshift mutation leading to a stop codon (V48fsX53). The L46P mutation was identified in affected patients presenting bilateral microphthalmia, cataracts, and nystagmus. The S74G mutation was found in a large family that had congenital ocular abnormalities, diverse neurological manifestations, and variable cognitive impairments. The 579delG deletion (V48fsX53) caused in the affected members of the same family bilateral aniridia associated with congenital cataract, foveal hypolasia, and nystagmus. We also detected a novel intronic nucleotide change, IVS2+9G>A (very likely a mutation) in an apparently isolated patient affected by a complex ocular phenotype, characterized primarily by a bilateral microphthalmia. Whether this nucleotide change is indeed pathogenic remains to be demonstrated. Two previously known heterozygous mutations of the PAX6 gene sequence were also detected in patients affected by aniridia: a de novo previously known nucleotide change, g.972C>T (Q179X), in exon 8, leading to a stop codon and a heterozygous g.555C>A (C40X) recurrent nonsense mutation in exon 5. No mutations were found in patients with Peters' anomaly. CONCLUSIONS: We identified three mutations associated with aniridia phenotypes (Q179X, C40X, and V48fsX53). The three other mutations reported here cause non-aniridia ocular phenotypes associated in some cases with neurological anomalies. The IVS2+9G>A nucleotide change was detected in a patient with a microphthalmia phenotype. The L46P mutation was detected in a family with microphthalmia, cataract, and nystagmus. This mutation is located in the DNA-binding paired-domain and the crystallographic representations of this mutation show that this mutation may affect the helix-turn-helix motif, and as a consequence the DNA-binding properties of the resulting mutated protein. Ser74 is located in the PAX6 PD linker region, essential for DNA recognition and DNA binding, and the side chain of the Ser74 contributes to DNA recognition by the linker domain through direct contacts. Crystallographic representations show that the S74G mutation results in no side chain and therefore perturbs the DNA-binding properties of PAX6. This study highlights the severity and diversity of the consequences of PAX6 mutations that appeared to result from the complexity of the PAX6 gene structure, and the numerous possibilities for DNA binding. This study emphasizes the fact that neurodevelopmental abnormalities may be caused by PAX6 mutations. The neuro-developmental abnormalities caused by PAX6 mutations are probably still overlooked in the current clinical examinations performed throughout the world in patients affected by PAX6 mutations.


Subject(s)
Eye Abnormalities/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Mutation , Nervous System Malformations/genetics , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Abnormalities, Multiple/genetics , Adolescent , Adult , Aged , Aniridia/genetics , Anterior Eye Segment/abnormalities , Cataract/complications , Cataract/genetics , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Microphthalmos/complications , Microphthalmos/genetics , Middle Aged , Nystagmus, Congenital/complications , Nystagmus, Congenital/genetics , PAX6 Transcription Factor , Phenotype
9.
BMC Med Genet ; 7: 82, 2006 Nov 29.
Article in English | MEDLINE | ID: mdl-17134502

ABSTRACT

BACKGROUND: Axenfeld-Rieger syndrome (ARS) is characterized by bilateral congenital abnormalities of the anterior segment of the eye associated with abnormalities of the teeth, midface, and umbilicus. Most cases of ARS are caused by mutations in the genes encoding PITX2 or FOXC1. Here we describe a family affected by a severe form of ARS. CASE PRESENTATION: Two members of this family (father and daughter) presented with typical ARS and developed severe glaucoma. The ocular phenotype was much more severe in the daughter than in the father. Magnetic resonance imaging (MRI) detected an aggressive form of meningioma in the father. There was no mutation in the PITX2 gene, determined by exon screening. We identified an intragenic deletion by quantitative genomic PCR analysis and characterized this deletion in detail. CONCLUSION: Our findings implicate the first intragenic deletion of the PITX2 gene in the pathogenesis of a severe form of ARS in an affected family. This study stresses the importance of a systematic search for intragenic deletions in families affected by ARS and in sporadic cases for which no mutations in the exons or introns of PITX2 have been found. The molecular genetics of some ARS pedigrees should be re-examined with enzymes that can amplify medium and large genomic fragments.


Subject(s)
Abnormalities, Multiple/genetics , Eye Abnormalities , Gene Deletion , Homeodomain Proteins/genetics , Transcription Factors/genetics , Abnormalities, Multiple/pathology , Adult , Anterior Eye Segment/abnormalities , Base Sequence , DNA Mutational Analysis/methods , Electrophoresis, Agar Gel , Family Health , Female , Humans , Male , Middle Aged , Optic Nerve/abnormalities , Pedigree , Syndrome , Tooth Abnormalities , Homeobox Protein PITX2
10.
Mol Vis ; 12: 1448-60, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17167399

ABSTRACT

PURPOSE: Axenfeld Rieger syndrome (ARS) is an autosomal dominant inherited disorder affecting development of the ocular anterior chamber, abdomen, teeth and facial structures. The PITX2 gene is a major gene encoding a major transcription factor associated with ARS. METHODS: ARS patients were collected from six unrelated families. Patients and their families were ophthalmologically phenotyped and their blood was collected for DNA extraction. We screened the coding region of human PITX2 gene by direct sequencing. The consequences of the mutations described were investigated by generating crystallographic representations of the amino acid changes. In order to better understand the occurrence of glaucoma in ARS patients, we studied the PITX2 gene expression in human embryonic and fetal ocular tissue sections. RESULTS: We identified four novel PITX2 genetic alterations in four unrelated families with ARS. These mutations included two nonsense mutations (E55X and Y121X), an eight nucleotides insertion (1251 ins CGACTCCT) and a substitution (F58L), in familial and sporadic cases of ARS. We also showed for the first time that PITX2 is expressed at early stages of the human embryonic and fetal periocular mesenchyme, as well as at later stages of human development in the fetal ciliary body, ciliary processes, irido corneal angle and corneal endothelium. The human fetal eye PITX2 gene expression pattern reported here for the first time provides a strong basis for explaining the frequent occurrence of glaucoma in patients affected by PITX2 gene mutations. CONCLUSIONS: Two mutations identified affect the homeodomain (E55X and F58L). The E55X nonsense mutation is likely to alter dramatically the DNA-binding capabilities of the PITX2 homeodomain. Furthermore, there is a complete loss of the carboxy-terminal part of the PITX2 protein beyond the site of the mutation. The phenylalanine F58 is known to contribute to the hydrophobic network of the homeodomain. The crystallographic representations of the mutation F58L show that this mutation may change the conformation of the helical core. The F58L mutation is very likely to modify the homeodomain conformation and probably alters the DNA binding properties of PITX2. The other mutations (Y121X and the eight-nucleotide insertion (1251 ins CGA CTC CT) CGA CTC CT, at position 224 in PITX2A) result in partial loss of the C-terminal domain of PITX2. Pitx2 synergistically transactivates the prolactin promoter in the presence of the POU homeodomain protein Pit-1. Pitx2 activity is regulated by its own C-terminal tail. This region contains a highly conserved 14-amino-acid element involved in protein-protein interactions. The C-terminal 39-amino-acid tail represses DNA binding activity and is required for Pitx2 interactions with other transcription factors, for Pitx2-Pit-1 interaction and Pit-1synergism. Pit-1 interaction with the Pitx2 C terminus masks the inhibitory effect and promotes increased DNA binding activity. Thus, the partial or complete loss of the C terminus tail can lead to decreased or absent DNA binding activity and trigger severe ARS phenotypes. Our in situ hybridization results obtained on human embryonic and fetal ocular tissue sections constitute the first molecular histological data providing an explanation for the occurrence of precocious glaucoma in human patients affected by ARS caused by PITX2 mutations. Further structural and biochemical studies are needed for understanding the wide spectrum of clinical phenotypes caused by the increasing number of new PITX2 mutations found in ARS affected patients.


Subject(s)
Abdomen/abnormalities , Abnormalities, Multiple/genetics , Anterior Chamber/abnormalities , Face/abnormalities , Homeodomain Proteins/genetics , Mutation , Tooth Abnormalities/complications , Transcription Factors/genetics , Amino Acid Sequence , Codon, Nonsense , DNA Transposable Elements , Embryo, Mammalian/metabolism , Eye/embryology , Eye Abnormalities/complications , Female , Fetus/metabolism , Gene Expression , Glaucoma/etiology , Homeodomain Proteins/metabolism , Humans , Male , Pedigree , Syndrome , Transcription Factors/metabolism , Homeobox Protein PITX2
11.
Hum Mutat ; 27(6): 553-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16652336

ABSTRACT

Epithelial basement membrane corneal dystrophy (EBMD), also known as Cogan microcystic epithelial dystrophy or map-dot-fingerprint dystrophy, is a common bilateral epithelial dystrophy. Usually, this disease is not considered to be inherited although several families with autosomal dominant inheritance have been described. We report the analysis of two families with an autosomal dominant pattern of inheritance as well as the analysis of single affected individuals; we identified two different point mutations in the TGFBI/BIGH3 genes, genes known to be associated with other corneal dystrophies. This is the first report of a molecular mutation in individuals with EBMD and it increases the spectrum of mutations in the TGFBI/BIGH3 gene. Based on our screening, up to 10% of EBMD patients could have a mutation in this gene.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Epithelium, Corneal/pathology , Extracellular Matrix Proteins/genetics , Transforming Growth Factor beta/genetics , Adult , Basement Membrane/pathology , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/pathology , DNA Mutational Analysis , Genetic Testing , Humans , Middle Aged , Pedigree , Point Mutation
12.
BMC Med Genet ; 7: 41, 2006 Apr 28.
Article in English | MEDLINE | ID: mdl-16646960

ABSTRACT

BACKGROUND: Ocular albinism type 1 (OA1) is an X-linked ocular disorder characterized by a severe reduction in visual acuity, nystagmus, hypopigmentation of the retinal pigmented epithelium, foveal hypoplasia, macromelanosomes in pigmented skin and eye cells, and misrouting of the optical tracts. This disease is primarily caused by mutations in the OA1 gene. METHODS: The ophthalmologic phenotype of the patients and their family members was characterized. We screened for mutations in the OA1 gene by direct sequencing of the nine PCR-amplified exons, and for genomic deletions by PCR-amplification of large DNA fragments. RESULTS: We sequenced the nine exons of the OA1 gene in 72 individuals and found ten different mutations in seven unrelated families and three sporadic cases. The ten mutations include an amino acid substitution and a premature stop codon previously reported by our team, and eight previously unidentified mutations: three amino acid substitutions, a duplication, a deletion, an insertion and two splice-site mutations. The use of a novel Taq polymerase enabled us to amplify large genomic fragments covering the OA1 gene. and to detect very likely six distinct large deletions. Furthermore, we were able to confirm that there was no deletion in twenty one patients where no mutation had been found. CONCLUSION: The identified mutations affect highly conserved amino acids, cause frameshifts or alternative splicing, thus affecting folding of the OA1 G protein coupled receptor, interactions of OA1 with its G protein and/or binding with its ligand.


Subject(s)
Albinism, Ocular/genetics , Eye Proteins/genetics , Membrane Glycoproteins/genetics , Mutation , Amino Acid Sequence , DNA Mutational Analysis , Female , Humans , Male , Molecular Sequence Data , Pedigree , Point Mutation , RNA Splice Sites , Sequence Deletion
13.
Invest Ophthalmol Vis Sci ; 45(5): 1297-305, 2004 May.
Article in English | MEDLINE | ID: mdl-15111580

ABSTRACT

PURPOSE: To identify proteins secreted by the retinal pigment epithelium (RPE) and to analyze their cellular distribution in normal and pathologic rat retinas at various stages of eye development. METHODS: A cDNA library was constructed with RNA isolated from porcine RPE sheets and screened by using the yeast signal sequence trap system. In situ hybridization, immunohistochemistry, and semiquantitative RT-PCR analysis were performed on rat retinas. RESULTS: The cDNA encoding prosaposin was isolated. This is the first time this gene has been shown to be expressed in the retina. Prosaposin mRNA was detected in the rat RPE cell monolayer and in ganglion cells 14, 21, and 45 days after birth. The amount of prosaposin mRNA increased between days 14 and 45 after birth in normal retinas (rdy+), but not in the pathologic retinas (rdy-) of RCS rats. CONCLUSIONS: Several techniques were used to determine the localization of prosaposin in rat retinas. The increase in the amount of prosaposin mRNA in normal retinas coincided with the maturation of photoreceptor cells and the beginning of the phagocytosis process. In addition, the RCS rdy- RPE cells, characterized by the abrogation of the ingestion phase of the photoreceptor outer segments, are deficient in prosaposin expression.


Subject(s)
Gene Expression , Glycoproteins/genetics , Retina/metabolism , Retinal Degeneration/metabolism , Amino Acid Sequence , Animals , Base Sequence , Gene Library , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , RNA, Messenger/metabolism , Rats , Rats, Mutant Strains , Retinal Degeneration/pathology , Reverse Transcriptase Polymerase Chain Reaction , Saposins , Swine
14.
Invest Ophthalmol Vis Sci ; 45(1): 7-14, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14691147

ABSTRACT

PURPOSE: It is important to understand the development of the normal retinal vascular system, because it may provide clues for understanding the mechanisms underlying the neovascularization associated with several retinopathies of infancy and adulthood. However, little is known about normal human ocular vascularization. VEGF is a key growth factor during vascular development and one of its receptors, KDR, plays a pivotal role in endothelial cell proliferation and differentiation. The purpose of this study was to analyze VEGF and KDR gene expression patterns during the development of the human eye during the embryonic and fetal stages. METHODS: The gene expression of VEGF and KDR was analyzed by in situ hybridization in 7-week-old embryos and in 10- and 18-week-old fetuses. In addition, we performed VEGF and KDR immunohistochemistry experiments on 18-week-old fetus tissue sections. RESULTS: These results clearly demonstrated that the levels of VEGF and KDR transcripts are correlated during the normal development of the ocular vasculature in humans. The complementarity between the patterns of VEGF and KDR during the early stages of development suggests that VEGF-KDR interactions play a major role in the formation and regression of the hyaloid vascular system (HVS) and in the development of the choriocapillaris. In later stages (i.e., 18-weeks-old fetuses), the expression of KDR seems to be linked to the development of the retinal vascular system. VEGF and KDR transcripts were unexpectedly detected in some nonvascular tissues-that is, in the cornea and in the retina before the development of the retinal vascular system. CONCLUSIONS: The expression of VEGF and KDR correlates highly with the normal ocular vascularization in humans, but VEGF may also be necessary for nonvascular retinal developmental functions, especially for the coordination of neural retinal development and the preliminary steps of the establishment of the definitive stable retinal vasculature.


Subject(s)
Embryonic and Fetal Development/physiology , Eye/embryology , Gene Expression Regulation, Developmental/physiology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Antibodies, Monoclonal , DNA Primers/chemistry , DNA Probes , Eye/blood supply , Eye/metabolism , Humans , Immunoenzyme Techniques , In Situ Hybridization , Neovascularization, Physiologic/physiology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/metabolism
15.
Cell Commun Adhes ; 10(3): 129-40, 2003.
Article in English | MEDLINE | ID: mdl-14668060

ABSTRACT

Integrins are a family of cell surface molecules that mediate the attachment of cells to the extracellular matrix (ECM). These alphabeta heterodimers are involved in many biological processes. We used northern blotting and in situ hybridization to study the pattern of beta3 integrin gene expression during mouse embryogenesis. Northern blotting detected two species of beta3 mRNA from 7 to 17 days post coitum (dpc). These transcripts were abundant in the adult testis, kidney, liver, spleen, and heart. In situ hybridization experiments detected high levels of beta3 in the major haematopoietic and lymphoid organs: yolk sac, liver, and thymus. Moreover, beta3 transcripts were also detected in the vascular system, where beta3 integrin probably plays a key role in angiogenesis and vasculogenesis. We also detected a hybridization signal in the gut, the bronchioles of the lungs, and the bladder wall. beta3 transcripts were also present in the medullary regions of the adrenal glands and in the developing skeleton. Our study shows that beta3 gene expression is not restricted to the liver and gut during mouse development. We also detected beta3 integrin mRNA in the yolk sac, vessels, lung, bladder, and developing bones. Our data suggest that beta3 integrin plays a key role in many important physiological processes like haematopoiesis, angiogenesis, phagocytosis, and bone resorption.


Subject(s)
Bone and Bones/metabolism , Cardiovascular System/metabolism , Hematopoietic System/metabolism , Integrin beta3/genetics , Thymus Gland/embryology , Animals , Blotting, Northern , Bone and Bones/embryology , Cardiovascular System/embryology , Gene Expression Regulation, Developmental , Hematopoietic System/embryology , In Situ Hybridization , Integrin beta3/biosynthesis , Liver/embryology , Liver/metabolism , Lung/embryology , Lung/metabolism , Mice , Organ Specificity , RNA, Messenger/metabolism , Thymus Gland/metabolism , Yolk Sac/embryology , Yolk Sac/metabolism
16.
Neurobiol Dis ; 14(2): 166-80, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14572440

ABSTRACT

Shed photoreceptor outer segments (POS) are phagocytosed by RPE cells in a circadian manner. The homozygous deletion of the c-mer gene abolishes the ingestion phase of this phagocytosis in the Royal College of Surgeons (RCS) rat strain, which in turn leads to the death of photoreceptor cells. We identified RPE transcripts for which the expression is modulated by the abrogation of POS phagocytosis. A microarray approach and the differential display (DDRT-PCR) technique revealed 116 modulated known genes, 4 modulated unknown genes, and 15 expressed sequenced tags (ESTs) corresponding to unknown genes. The microarray and DDRT-PCR analyses detected alterations in signaling pathways such as the phosphatidylinositol 3-kinase-Akt-mTOR pathway and the DLK/JNK/SAPK pathway. The abrogation of POS phagocytosis caused a decrease in endomembrane biogenesis and altered endocytosis, exocytosis, transcytosis, and several metabolic and signaling pathways in RCS RPE cells. We also found differential levels of transcripts encoding proteins involved in phagocytosis, vesicle trafficking, the cytoskeleton, retinoic acid, and general metabolism.


Subject(s)
Pigment Epithelium of Eye/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Signal Transduction/genetics , Animals , Base Sequence , DNA, Complementary/genetics , Gene Expression Regulation/genetics , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis/methods , Phagocytosis/genetics , Pigment Epithelium of Eye/chemistry , Pigment Epithelium of Eye/metabolism , Rats , Rats, Mutant Strains , Retinal Degeneration/metabolism , Rod Cell Outer Segment/metabolism , Rod Cell Outer Segment/pathology
17.
Ophthalmic Genet ; 24(3): 167-73, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12868035

ABSTRACT

Ocular albinism type 1 (OA1) is an X-linked disorder, mainly characterized by a severe reduction in visual acuity, foveal hypoplasia, nystagmus, hypopigmentation of the retina, the presence of macromelanosomes in the skin and eyes, and the misrouting of optic pathways, resulting in the loss of stereoscopic vision. We screened the OA1 gene for mutations in three unrelated Canadian and French families and in two isolated patients with OA1. We found three different missense mutations and two different nonsense mutations, three of which were novel. To date, 41 mutations (including missense mutations, insertions, and deletions) have been reported in the OA1 gene. Mutation and polymorphism data for this gene are available from the international albinism center albinism database website: http://www.cbc.umn.edu/tad/oa1map.htm.


Subject(s)
Albinism, Ocular/genetics , Eye Proteins/genetics , Membrane Glycoproteins/genetics , Mutation/genetics , Polymorphism, Genetic/genetics , Albinism, Ocular/physiopathology , Amino Acid Sequence , DNA Mutational Analysis , Exons/genetics , Female , Humans , Male , Molecular Sequence Data , Pedigree , Phenotype , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL