Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Mol Ther Methods Clin Dev ; 32(1): 101187, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38327809

ABSTRACT

Inherited retinal diseases are a leading and untreatable cause of blindness and are therefore candidate diseases for gene therapy. Recombinant vectors derived from adeno-associated virus (rAAV) are currently the most promising vehicles for in vivo therapeutic gene delivery to the retina. However, there is a need for novel AAV-based vectors with greater efficacy for ophthalmic applications, as underscored by recent reports of dose-related inflammatory responses in clinical trials of rAAV-based ocular gene therapies. Improved therapeutic efficacy of vectors would allow for decreases in the dose delivered, with consequent reductions in inflammatory reactions. Here, we describe the development of new rAAV vectors using bioconjugation chemistry to modify the rAAV capsid, thereby improving the therapeutic index. Covalent coupling of a mannose ligand, via the formation of a thiourea bond, to the amino groups of the rAAV capsid significantly increases vector transduction efficiency of both rat and nonhuman primate retinas. These optimized rAAV vectors have important implications for the treatment of a wide range of retinal diseases.

2.
Mol Ther Methods Clin Dev ; 28: 387-393, 2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36874242

ABSTRACT

The subretinal injection protocol for the only approved retinal gene therapy (voretigene neparvovec-rzyl) includes air tamponade at the end of the procedure, but its effects on the subretinal bleb have not been described. In the present study, we evaluated the distribution of enhanced green fluorescent protein (EGFP) after subretinal injection of AAV2 in non-human primates (NHP) without (group A = 3 eyes) or with (group B = 3 eyes) air tamponade. The retinal expression of EGFP was assessed 1 month after subretinal injection with in vivo fundus photographs and fundus autofluorescence. In group A (without air), EGFP expression was limited to the area of the initial subretinal bleb. In group B (with air), EGFP was expressed in a much wider area. These data show that the buoyant force of air on the retina causes a wide subretinal diffusion of vector, away from the injection site. In the present paper, we discuss the beneficial and deleterious clinical effects of this finding. Whereas subretinal injection is likely to become more common with the coming of new gene therapies, the effects of air tamponade should be explored further to improve efficacy, reproducibility, and safety of the protocol.

3.
Proc Natl Acad Sci U S A ; 117(49): 31278-31289, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229591

ABSTRACT

Presbycusis, or age-related hearing loss (ARHL), is a major public health issue. About half the phenotypic variance has been attributed to genetic factors. Here, we assessed the contribution to presbycusis of ultrarare pathogenic variants, considered indicative of Mendelian forms. We focused on severe presbycusis without environmental or comorbidity risk factors and studied multiplex family age-related hearing loss (mARHL) and simplex/sporadic age-related hearing loss (sARHL) cases and controls with normal hearing by whole-exome sequencing. Ultrarare variants (allele frequency [AF] < 0.0001) of 35 genes responsible for autosomal dominant early-onset forms of deafness, predicted to be pathogenic, were detected in 25.7% of mARHL and 22.7% of sARHL cases vs. 7.5% of controls (P = 0.001); half were previously unknown (AF < 0.000002). MYO6, MYO7A, PTPRQ, and TECTA variants were present in 8.9% of ARHL cases but less than 1% of controls. Evidence for a causal role of variants in presbycusis was provided by pathogenicity prediction programs, documented haploinsufficiency, three-dimensional structure/function analyses, cell biology experiments, and reported early effects. We also established Tmc1N321I/+ mice, carrying the TMC1:p.(Asn327Ile) variant detected in an mARHL case, as a mouse model for a monogenic form of presbycusis. Deafness gene variants can thus result in a continuum of auditory phenotypes. Our findings demonstrate that the genetics of presbycusis is shaped by not only well-studied polygenic risk factors of small effect size revealed by common variants but also, ultrarare variants likely resulting in monogenic forms, thereby paving the way for treatment with emerging inner ear gene therapy.


Subject(s)
Deafness/genetics , Genes, Dominant , Mutation/genetics , Presbycusis/genetics , Age Factors , Age of Onset , Animals , Case-Control Studies , Cohort Studies , Heterozygote , Humans , Membrane Proteins/genetics , Mice , MicroRNAs/genetics , Mitochondria/genetics , Exome Sequencing
4.
Eur J Hum Genet ; 24(12): 1730-1738, 2016 12.
Article in English | MEDLINE | ID: mdl-27460420

ABSTRACT

Usher syndrome (USH), the most prevalent cause of hereditary deafness-blindness, is an autosomal recessive and genetically heterogeneous disorder. Three clinical subtypes (USH1-3) are distinguishable based on the severity of the sensorineural hearing impairment, the presence or absence of vestibular dysfunction, and the age of onset of the retinitis pigmentosa. A total of 10 causal genes, 6 for USH1, 3 for USH2, and 1 for USH3, and an USH2 modifier gene, have been identified. A robust molecular diagnosis is required not only to improve genetic counseling, but also to advance gene therapy in USH patients. Here, we present an improved diagnostic strategy that is both cost- and time-effective. It relies on the sequential use of three different techniques to analyze selected genomic regions: targeted exome sequencing, comparative genome hybridization, and quantitative exon amplification. We screened a large cohort of 427 patients (139 USH1, 282 USH2, and six of undefined clinical subtype) from various European medical centers for mutations in all USH genes and the modifier gene. We identified a total of 421 different sequence variants predicted to be pathogenic, about half of which had not been previously reported. Remarkably, we detected large genomic rearrangements, most of which were novel and unique, in 9% of the patients. Thus, our strategy led to the identification of biallelic and monoallelic mutations in 92.7% and 5.8% of the USH patients, respectively. With an overall 98.5% mutation characterization rate, the diagnosis efficiency was substantially improved compared with previously reported methods.


Subject(s)
Genetic Testing/methods , Mutation , Usher Syndromes/genetics , Alleles , Comparative Genomic Hybridization/methods , Europe , Exome , Extracellular Matrix Proteins/genetics , Genes, Modifier , Humans , Sensitivity and Specificity , Sequence Analysis, DNA/methods , Usher Syndromes/diagnosis
5.
J Biol Chem ; 291(34): 17919-28, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27288410

ABSTRACT

The primary role of the RNAi machinery is to promote mRNA degradation within the cytoplasm in a microRNA-dependent manner. However, both Dicer and the Argonaute protein family have expanded roles in gene regulation within the nucleus. To further our understanding of this role, we have identified chromatin binding sites for AGO2 throughout the 45S region of the human rRNA gene. The location of these sites was mirrored by the positions of AGO2 cross-linking sites identified via PAR-CLIP-seq. AGO2 binding to the rRNA within the nucleus was confirmed by RNA immunoprecipitation and quantitative-PCR. To explore a possible mechanism by which AGO2 could be recruited to the rRNA, we identified 1174 regions within the 45S rRNA transcript that have the ability to form a perfect duplex with position 2-6 (seed sequence) of each microRNA expressed in HEK293T cells. Of these potential AGO2 binding sites, 479 occurred within experimentally verified AGO2-rRNA cross-linking sites. The ability of AGO2 to cross-link to rRNA was almost completely lost in a DICER knock-out cell line. The transfection of miR-92a-2-3p into the noDICE cell line facilitated AGO2 cross-linking at a region of the rRNA that has a perfect seed match at positions 3-8, including a single G-U base pair. Knockdown of AGO2 within HEK293T cells causes a slight, but statistically significant increase in the overall rRNA synthesis rate but did not impact the ratio of processing intermediates or the recruitment of the Pol I transcription factor UBTF.


Subject(s)
Argonaute Proteins/metabolism , MicroRNAs/metabolism , Pol1 Transcription Initiation Complex Proteins/metabolism , RNA Polymerase I/metabolism , RNA, Ribosomal/biosynthesis , Argonaute Proteins/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Knockdown Techniques , Humans , K562 Cells , MicroRNAs/genetics , Pol1 Transcription Initiation Complex Proteins/genetics , RNA Polymerase I/genetics , RNA, Ribosomal/genetics , Ribonuclease III/genetics , Ribonuclease III/metabolism
6.
Orphanet J Rare Dis ; 10: 96, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26282398

ABSTRACT

BACKGROUND: More than 70 % of the cases of congenital deafness are of genetic origin, of which approximately 80 % are non-syndromic and show autosomal recessive transmission (DFNB forms). To date, 60 DFNB genes have been identified, most of which cause congenital, severe to profound deafness, whereas a few cause delayed progressive deafness in childhood. We report the study of two Algerian siblings born to consanguineous parents, and affected by progressive hearing loss. METHOD: After exclusion of GJB2 (the gene most frequently involved in non-syndromic deafness in Mediterranean countries), we performed whole-exome sequencing in one sibling. RESULTS: A frame-shift variant (c.1014delC; p.Ser339Alafs*15) was identified in EPS8L2, encoding Epidermal growth factor receptor Pathway Substrate 8 L2, a protein of hair cells' stereocilia previously implicated in progressive deafness in the mouse. This variant predicts a truncated, inactive protein, or no protein at all owing to nonsense-mediated mRNA decay. It was detected at the homozygous state in the two clinically affected siblings, and at the heterozygous state in the unaffected parents and one unaffected sibling, whereas it was never found in a control population of 150 Algerians with normal hearing or in the Exome Variant Server database. CONCLUSION: Whole-exome sequencing allowed us to identify a new gene responsible for childhood progressive hearing loss transmitted on the autosomal recessive mode.


Subject(s)
Genes, Recessive , Hearing Loss/genetics , Membrane Proteins/genetics , Child , Connexin 26 , Connexins , Consanguinity , Disease Progression , Female , Frameshift Mutation , Humans , Male , Microfilament Proteins , Pedigree
7.
Mol Genet Genomic Med ; 3(3): 189-96, 2015 May.
Article in English | MEDLINE | ID: mdl-26029705

ABSTRACT

The genetic heterogeneity of congenital hearing disorders makes molecular diagnosis expensive and time-consuming using conventional techniques such as Sanger sequencing of DNA. In order to design an appropriate strategy of molecular diagnosis in the Algerian population, we explored the diversity of the involved mutations by studying 65 families affected by autosomal recessive forms of nonsyndromic hearing impairment (DFNB forms), which are the most prevalent early onset forms. We first carried out a systematic screening for mutations in GJB2 and the recurrent p.(Arg34*) mutation in TMC1, which were found in 31 (47.7%) families and 1 (1.5%) family, respectively. We then performed whole exome sequencing in nine of the remaining families, and identified the causative mutations in all the patients analyzed, either in the homozygous state (eight families) or in the compound heterozygous state (one family): (c.709C>T: p.(Arg237*)) and (c.2122C>T: p.(Arg708*)) in OTOF, (c.1334T>G: p.(Leu445Trp)) in SLC26A4, (c.764T>A: p.(Met255Lys)) in GIPC3, (c.518T>A: p.(Cys173Ser)) in LHFPL5, (c.5336T>C: p.(Leu1779Pro)) in MYO15A, (c.1807G>T: p.(Val603Phe)) in OTOA, (c.6080dup: p.(Asn2027Lys*9)) in PTPRQ, and (c.6017del: p.(Gly2006Alafs*13); c.7188_7189ins14: p.(Val2397Leufs*2)) in GPR98. Notably, 7 of these 10 mutations affecting 8 different genes had not been reported previously. These results highlight for the first time the genetic heterogeneity of the early onset forms of nonsyndromic deafness in Algerian families.

8.
Proc Natl Acad Sci U S A ; 111(47): 16760-5, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385647

ABSTRACT

We used circular chromatin conformation capture (4C) to identify a physical contact in human pancreatic islets between the region near the insulin (INS) promoter and the ANO1 gene, lying 68 Mb away on human chromosome 11, which encodes a Ca(2+)-dependent chloride ion channel. In response to glucose, this contact was strengthened and ANO1 expression increased, whereas inhibition of INS gene transcription by INS promoter targeting siRNA decreased ANO1 expression, revealing a regulatory effect of INS promoter on ANO1 expression. Knockdown of ANO1 expression caused decreased insulin secretion in human islets, establishing a physical proximity-dependent feedback loop involving INS transcription, ANO1 expression, and insulin secretion. To explore a possible role of ANO1 in insulin metabolism, we carried out experiments in Ano1(+/-) mice. We observed reduced serum insulin levels and insulin-to-glucose ratios in high-fat diet-fed Ano1(+/-) mice relative to Ano1(+/+) mice fed the same diet. Our results show that determination of long-range contacts within the nucleus can be used to detect novel and physiologically relevant mechanisms. They also show that networks of long-range physical contacts are important to the regulation of insulin metabolism.


Subject(s)
Chloride Channels/physiology , Insulin/genetics , Neoplasm Proteins/physiology , Promoter Regions, Genetic , Animals , Anoctamin-1 , Chloride Channels/genetics , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Polymerase Chain Reaction
9.
PLoS Genet ; 6(10): e1001142, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21060806

ABSTRACT

Methylation of specific lysine residues in core histone proteins is essential for embryonic development and can impart active and inactive epigenetic marks on chromatin domains. The ubiquitous nuclear protein PTIP is encoded by the Paxip1 gene and is an essential component of a histone H3 lysine 4 (H3K4) methyltransferase complex conserved in metazoans. In order to determine if PTIP and its associated complexes are necessary for maintaining stable gene expression patterns in a terminally differentiated, non-dividing cell, we conditionally deleted PTIP in glomerular podocytes in mice. Renal development and function were not impaired in young mice. However, older animals progressively exhibited proteinuria and podocyte ultra structural defects similar to chronic glomerular disease. Loss of PTIP resulted in subtle changes in gene expression patterns prior to the onset of a renal disease phenotype. Chromatin immunoprecipitation showed a loss of PTIP binding and lower H3K4 methylation at the Ntrk3 (neurotrophic tyrosine kinase receptor, type 3) locus, whose expression was significantly reduced and whose function may be essential for podocyte foot process patterning. These data demonstrate that alterations or mutations in an epigenetic regulatory pathway can alter the phenotypes of differentiated cells and lead to a chronic disease state.


Subject(s)
Histones/metabolism , Kidney Diseases/metabolism , Podocytes/metabolism , Signal Transduction , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chronic Disease , DNA-Binding Proteins , Female , Gene Expression Profiling , Kidney/metabolism , Kidney/pathology , Kidney/ultrastructure , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Lysine/metabolism , Male , Methylation , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Podocytes/cytology , Podocytes/ultrastructure , Receptor, trkC/genetics , Receptor, trkC/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Pflugers Arch ; 459(1): 115-30, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19756723

ABSTRACT

We assessed the involvement of harmonin-b, a submembranous protein containing PDZ domains, in the mechanoelectrical transduction machinery of inner ear hair cells. Harmonin-b is located in the region of the upper insertion point of the tip link that joins adjacent stereocilia from different rows and that is believed to gate transducer channel(s) located in the region of the tip link's lower insertion point. In Ush1c (dfcr-2J/dfcr-2J) mutant mice defective for harmonin-b, step deflections of the hair bundle evoked transduction currents with altered speed and extent of adaptation. In utricular hair cells, hair bundle morphology and maximal transduction currents were similar to those observed in wild-type mice, but adaptation was faster and more complete. Cochlear outer hair cells displayed reduced maximal transduction currents, which may be the consequence of moderate structural anomalies of their hair bundles. Their adaptation was slower and displayed a variable extent. The latter was positively correlated with the magnitude of the maximal transduction current, but the cells that showed the largest currents could be either hyperadaptive or hypoadaptive. To interpret our observations, we used a theoretical description of mechanoelectrical transduction based on the gating spring theory and a motor model of adaptation. Simulations could account for the characteristics of transduction currents in wild-type and mutant hair cells, both vestibular and cochlear. They led us to conclude that harmonin-b operates as an intracellular link that limits adaptation and engages adaptation motors, a dual role consistent with the scaffolding property of the protein and its binding to both actin filaments and the tip link component cadherin-23.


Subject(s)
Adaptation, Physiological , Carrier Proteins/metabolism , Hair Cells, Auditory, Inner/metabolism , Mechanotransduction, Cellular/physiology , Action Potentials/physiology , Animals , Cell Cycle Proteins , Cytoskeletal Proteins , Fluorescent Antibody Technique , Mice , Mice, Mutant Strains , Microscopy, Electron, Scanning , Patch-Clamp Techniques
11.
Invest Ophthalmol Vis Sci ; 50(3): 1047-57, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19029025

ABSTRACT

PURPOSE: Constitutive activation of ERK1/2 controls proliferation of uveal melanoma cells. Because an autocrine fibroblast growth factor (FGF) activation loop controls ERK1/2 activation in many cancers, this study was conducted to examine the role of the FGF/FGF receptor autocrine loop in the ERK1/2-dependent proliferation and survival of uveal melanoma cells. METHODS: Primary tumors and cell lines (OCM-1, MKT-BR, SP6.5, Mel270 and 92.1) were used to define the role of the FGF/FGFR system in human uveal melanoma. Cell proliferation was assessed by MTT-staining, and apoptosis was quantified by flow cytometry. Specific pharmacologic inhibitors of ERK1/2 and FGFR1, an anti-FGF2 neutralizing antibody and an antisense oligonucleotide directed against FGF2 were used to analyze signaling in the FGF/FGFR autocrine loop. RESULTS: FGF1, FGF2, and their FGFR1 receptor were strongly expressed in the primary uveal melanomas. All five uveal melanoma cell lines expressed and secreted FGF2. They also expressed FGFR1. Cell proliferation was strongly reduced by the antisense oligonucleotide-mediated depletion of endogenous FGF2, immunoneutralization of secreted FGF2, and pharmacologic inhibition of FGFR1. The FGF2/FGFR1-mediated signaling pathway was identified by showing that inhibition of either FGF2 or FGFR1 reduced ERK1/2 activation, cell proliferation, and survival. CONCLUSIONS: The FGF/FGFR/ERK signaling pathway may be a target for therapeutic strategies against uveal melanoma.


Subject(s)
Autocrine Communication/physiology , Cell Proliferation , Fibroblast Growth Factor 2/metabolism , Melanoma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Uveal Neoplasms/metabolism , Apoptosis , Blotting, Western , Cell Survival , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Flow Cytometry , Humans , Melanoma/pathology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins B-raf/physiology , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Thionucleotides/pharmacology , Tumor Cells, Cultured , Uveal Neoplasms/pathology
12.
Nature ; 456(7219): 255-8, 2008 Nov 13.
Article in English | MEDLINE | ID: mdl-18849963

ABSTRACT

Although the cochlea is an amplifier and a remarkably sensitive and finely tuned detector of sounds, it also produces conspicuous mechanical and electrical waveform distortions. These distortions reflect nonlinear mechanical interactions within the cochlea. By allowing one tone to suppress another (masking effect), they contribute to speech intelligibility. Tones can also combine to produce sounds with frequencies not present in the acoustic stimulus. These sounds compose the otoacoustic emissions that are extensively used to screen hearing in newborns. Because both cochlear amplification and distortion originate from the outer hair cells-one of the two types of sensory receptor cells-it has been speculated that they stem from a common mechanism. Here we show that the nonlinearity underlying cochlear waveform distortions relies on the presence of stereocilin, a protein defective in a recessive form of human deafness. Stereocilin was detected in association with horizontal top connectors, lateral links that join adjacent stereocilia within the outer hair cell's hair bundle. These links were absent in stereocilin-null mutant mice, which became progressively deaf. At the onset of hearing, however, their cochlear sensitivity and frequency tuning were almost normal, although masking was much reduced and both acoustic and electrical waveform distortions were completely lacking. From this unique functional situation, we conclude that the main source of cochlear waveform distortions is a deflection-dependent hair bundle stiffness resulting from constraints imposed by the horizontal top connectors, and not from the intrinsic nonlinear behaviour of the mechanoelectrical transducer channel.


Subject(s)
Cochlea/physiology , Hair Cells, Auditory/metabolism , Proteins/genetics , Proteins/metabolism , Acoustic Stimulation , Animals , Female , Gene Expression Regulation , Hair Cells, Auditory/cytology , Hair Cells, Auditory/ultrastructure , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Knockout
13.
Development ; 135(8): 1427-37, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18339676

ABSTRACT

The planar polarity and staircase-like pattern of the hair bundle are essential to the mechanoelectrical transduction function of inner ear sensory cells. Mutations in genes encoding myosin VIIa, harmonin, cadherin 23, protocadherin 15 or sans cause Usher syndrome type I (USH1, characterized by congenital deafness, vestibular dysfunction and retinitis pigmentosa leading to blindness) in humans and hair bundle disorganization in mice. Whether the USH1 proteins are involved in common hair bundle morphogenetic processes is unknown. Here, we show that mouse models for the five USH1 genetic forms share hair bundle morphological defects. Hair bundle fragmentation and misorientation (25-52 degrees mean kinociliary deviation, depending on the mutant) were detected as early as embryonic day 17. Abnormal differential elongation of stereocilia rows occurred in the first postnatal days. In the emerging hair bundles, myosin VIIa, the actin-binding submembrane protein harmonin-b, and the interstereocilia-kinocilium lateral link components cadherin 23 and protocadherin 15, all concentrated at stereocilia tips, in accordance with their known in vitro interactions. Soon after birth, harmonin-b switched from the tip of the stereocilia to the upper end of the tip link, which also comprises cadherin 23 and protocadherin 15. This positional change did not occur in mice deficient for cadherin 23 or protocadherin 15. We suggest that tension forces applied to the early lateral links and to the tip link, both of which can be anchored to actin filaments via harmonin-b, play a key role in hair bundle cohesion and proper orientation for the former, and in stereociliary elongation for the latter.


Subject(s)
Cochlea/embryology , Usher Syndromes/embryology , Usher Syndromes/genetics , Animals , Cadherin Related Proteins , Cadherins/genetics , Cadherins/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Cycle Proteins , Cochlea/abnormalities , Cochlea/ultrastructure , Cytoskeletal Proteins , Disease Models, Animal , Dyneins/genetics , Dyneins/physiology , Female , Humans , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Mice , Mice, Knockout , Mice, Mutant Strains , Microscopy, Electron, Scanning , Myosin VIIa , Myosins/genetics , Myosins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Phenotype , Pregnancy , Protein Precursors/genetics , Protein Precursors/physiology , Usher Syndromes/physiopathology
14.
J Neurosci ; 27(24): 6478-88, 2007 Jun 13.
Article in English | MEDLINE | ID: mdl-17567809

ABSTRACT

Several lines of evidence indicate that very large G-protein-coupled receptor 1 (Vlgr1) makes up the ankle links that connect the stereocilia of hair cells at their base. Here, we show that the transmembrane protein usherin, the putative transmembrane protein vezatin, and the PDZ (postsynaptic density-95/Discs large/zona occludens-1) domain-containing submembrane protein whirlin are colocalized with Vlgr1 at the stereocilia base in developing cochlear hair cells and are absent in Vlgr1-/- mice that lack the ankle links. Direct in vitro interactions between these four proteins further support their involvement in a molecular complex associated with the ankle links and scaffolded by whirlin. In addition, the delocalization of these proteins in myosin VIIa defective mutant mice as well as the myosin VIIa tail direct interactions with vezatin, whirlin, and, we show, Vlgr1 and usherin, suggest that myosin VIIa conveys proteins of the ankle-link complex to the stereocilia. Adenylyl cyclase 6, which was found at the base of stereocilia, was both overexpressed and mislocated in Vlgr1-/- mice. In postnatal day 7 Vlgr1-/- mice, mechanoelectrical transduction currents evoked by displacements of the hair bundle toward the tallest stereocilia (i.e., in the excitatory direction) were reduced in outer but not inner hair cells. In both cell types, stimulation of the hair bundle in the opposite direction paradoxically resulted in significant transduction currents. The absence of ankle-link-mediated cohesive forces within hair bundles lacking Vlgr1 may account for the electrophysiological results. However, because some long cadherin-23 isoforms could no longer be detected in Vlgr1-/- mice shortly after birth, the loss of some apical links could be involved too. The premature disappearance of these cadherin isoforms in the Vlgr1-/- mutant argues in favor of a signaling function of the ankle links in hair bundle differentiation.


Subject(s)
Cochlea/cytology , Gene Expression Regulation, Developmental/physiology , Hair Cells, Auditory/metabolism , Adenylyl Cyclases/metabolism , Animals , Animals, Newborn , Carrier Proteins/metabolism , Chelating Agents/pharmacology , Cilia/physiology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Embryo, Mammalian , Extracellular Matrix Proteins/metabolism , Hair Cells, Auditory/ultrastructure , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Scanning/methods , Organ Culture Techniques , Patch-Clamp Techniques , Receptors, G-Protein-Coupled/deficiency , Subtilisin/pharmacology
15.
Hum Mol Genet ; 14(24): 3921-32, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16301217

ABSTRACT

Usher syndrome type IIa (USH2A) combines moderate to severe congenital hearing impairment and retinitis pigmentosa. It is the most common genetic form of USH. USH2A encodes usherin, which was previously defined as a basement membrane protein. A much larger USH2A transcript predicted to encode a transmembrane (TM) isoform was recently reported. Here, we address the role of TM usherin in the inner ear. Analysis of the usherin alternative transcripts in the murine inner ear revealed the existence of several predicted TM usherin isoforms with modular ectodomains of different lengths. In addition, we identified in the usherin cytoplasmic region a predicted 24 amino acid peptide, derived from a newly defined exon that is predominantly expressed in the inner ear but not in the retina. In mouse and rat inner ears, we show that TM usherin is present at the base of the differentiating stereocilia, which make up the mechanosensitive hair bundles receptive to sound. The usherin immunolabeling is transient in the hair bundles of cochlear hair cells (HCs), but persists in mature hair bundles of vestibular HCs. Several lines of evidence support the involvement of TM usherin in the composition of the ankle links, a subset of filamentous lateral links connecting stereocilia at the base. By co-immunoprecipitation and in vitro binding assays, we establish that the usherin cytodomain can bind to whirlin and harmonin, two PDZ domain-containing proteins that are defective in genetic forms of isolated deafness and USH type I, respectively. These PDZ proteins are suitable to provide the anchoring of interstereocilia lateral links to the F-actin core of stereocilia. Our results strongly suggest that congenital deafness in USH type I and type II shares similar pathogenic mechanisms, i.e. the disruption of hair bundle links-mediated adhesion forces that are essential for the proper organization of growing hair bundles.


Subject(s)
Ear, Inner/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Hair Cells, Auditory/metabolism , Usher Syndromes/physiopathology , Alternative Splicing , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cell Cycle Proteins , Cilia/metabolism , Cilia/pathology , Cytoskeletal Proteins , Ear, Inner/cytology , Hair Cells, Auditory/pathology , Humans , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Homology, Amino Acid
16.
Med Sci (Paris) ; 21(8-9): 737-40, 2005.
Article in French | MEDLINE | ID: mdl-16115459

ABSTRACT

Defects in myosin VIIa, the PDZ-domain-containing protein harmonin, cadherin 23, protocadherin 15, and the putative scaffolding protein sans, underlie five genetic forms of Usher syndrome type I (USH1), the most frequent cause of hereditary deafness-blindness in humans. Mice mutants defective for any of these proteins have a severe hearing impairment and display similar inner ear phenotypes characterized by the abnormal spreading of the sensory cells' stereocilia. These are highly specialized mechanoreceptive organelles derived from microvilli, that normally form a well-structured hair bundle at the apex of inner ear sensory cells. All the USH1 proteins, except sans, have been detected in the growing stereocilia. Moreover, biochemical studies have started to unravel the multiple direct molecular interactions between USH1 proteins. In particular, harmonin can bind to the other four USH1 proteins and to F-actin. Finally, cell biology studies have provided the first insights into the functions of these proteins, and revealed that cadherin 23, and probably protocadherin 15 also, are associated with transient lateral links that interconnect growing stereocilia. These connectors play a critical role in the differentiating hair bundle.


Subject(s)
Ear, Inner/pathology , Hair Cells, Auditory, Inner/pathology , Hearing Loss, Sensorineural/pathology , Animals , Cell Differentiation , Humans , Mice
17.
J Biol Chem ; 279(30): 31769-79, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15145934

ABSTRACT

The B-Raf(V599E)-mediated constitutive activation of ERK1/2 is involved in establishing the transformed phenotype of some uveal melanoma cells (Calipel, A., Lefevre, G., Pouponnot, C., Mouriaux, F., Eychene, A., and Mascarelli, F. (2003) J. Biol. Chem. 278, 42409-42418). We have shown that stem cell factor (SCF) is involved in the proliferation of normal uveal melanocytes and that c-Kit is expressed in 75% of primary uveal melanomas. This suggests that the acquisition of autonomous growth during melanoma progression may involve the SCF/c-Kit axis. We used six human uveal melanoma tumor-derived cell lines and normal uveal melanocytes to characterize the SCF/c-Kit system and to assess its specific role in transformation. We investigated the possible roles of activating mutations in c-KIT, the overexpression of this gene, and ligand-dependent c-Kit overactivation in uveal melanoma cell tumorigenesis. Four cell lines (92.1, SP6.5, Mel270, and TP31) expressed both SCF and c-Kit, and none harbored the c-KIT mutations in exons 9, 11, 13, and 17 that have been shown to induce SCF-independent c-Kit activation. Melanoma cell proliferation was strongly inhibited by small interfering RNA-mediated depletion of c-Kit in these cells, despite the presence of (V599E)B-Raf in SP6.5 and TP31 cells. We characterized the signaling pathways involved in SCF/c-Kit-mediated cell growth and survival in normal and tumoral melanocytes and found that constitutive ERK1/2 activation played a key role in both the SCF/c-Kit autocrine loop and the gain of function of (V599E)B-Raf for melanoma cell proliferation and transformation. We also provide the first evidence that Glivec/STI571, a c-Kit tyrosine kinase inhibitor, could be used to treat uveal melanomas.


Subject(s)
Melanoma/etiology , Proto-Oncogene Proteins c-kit/physiology , Stem Cell Factor/physiology , Uveal Neoplasms/etiology , Base Sequence , Benzamides , Cell Division , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA Primers/genetics , Gene Expression , Humans , Imatinib Mesylate , Melanocytes/physiology , Melanoma/drug therapy , Melanoma/genetics , Melanoma/physiopathology , Mitogens/metabolism , Mutation , Piperazines/therapeutic use , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/therapeutic use , RNA, Small Interfering/genetics , Signal Transduction , Stem Cell Factor/genetics , Uvea/physiology , Uveal Neoplasms/drug therapy , Uveal Neoplasms/genetics , Uveal Neoplasms/physiopathology
18.
Oncogene ; 22(55): 8813-22, 2003 Dec 04.
Article in English | MEDLINE | ID: mdl-14654778

ABSTRACT

Although there is no current evidence for ras gene mutation in choroidal melanoma, there is an increasing body of evidence indicating that deregulated intracellular signalling pathways are involved in choroidal melanoma pathogenesis. The various components of the linear Raf/MEK/ERK signalling pathway have been implicated in various tumours. We therefore investigated the role of Raf-1 and the MEK/ERK module in the proliferation of human normal choroidal melanocytes (NCM) and cells from the ocular choroidal melanoma (OCM-1) cell line. OCM-1 cells proliferated four times faster than NCM. High basal activation of the MEK/ERK module was observed in unstimulated OCM-1 cells, whereas rapid and persistent activation was detected after serum stimulation, throughout the 24-h period of culture. In contrast, the activation of MEK/ERK was barely detectable in unstimulated NCM and occurred late (6 h) after the stimulation of cell proliferation. Inhibition of Raf-1 and MEK1/2 activation by pharmacological approaches and of the production of Raf-1 and ERK1/2 by antisense oligonucleotide approaches demonstrated that Raf-1 and the MEK/ERK module controlled proliferation in OCM-1 cells, but not in NCM. OCM-1 cells produced very low levels of p27Kip1, whereas NCM produced constant, high levels of p27Kip1. The inhibition of Raf-1 or MEK1/2 induced a large increase in p27Kip1 in OCM-1 cells, associated with an arrest of cell proliferation. Levels of c-Myc production were high and constant in OCM-1 cells and low in NCM, in contrast to what was observed for p27Kip1. The inhibition of both Raf-1 and MEK1/2 induced a decrease in c-Myc production and downregulated c-Myc activity by preventing c-Myc phosphorylation in OCM-1 cells. We conclude that Raf-1 and the MEK/ERK module control the production of both p27Kip1 and c-Myc, and the activation of c-Myc for OCM-1 cell proliferation.


Subject(s)
Cell Cycle Proteins/metabolism , MAP Kinase Kinase Kinase 1 , Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Tumor Suppressor Proteins/metabolism , Cell Division/physiology , Choroid Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Gene Expression Regulation, Neoplastic , Humans , Melanoma/metabolism
19.
J Biol Chem ; 278(43): 42409-18, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-12917419

ABSTRACT

The BRAF gene, encoding a mitogen-activated protein kinase kinase kinase, is mutated in several human cancers, with the highest incidence occurring in cutaneous melanoma. The activating V599E mutation accounted for 80% of all mutations detected in cutaneous melanoma cell lines. Reconstitution experiments have shown that this mutation increases ectopically expressed B-Raf kinase activity and induces NIH3T3 cell transformation. Here we used tumor-derived cell lines to characterize the activity of endogenous mutated B-Raf protein and assess its specific role in transformation. We show that three cell lines (OCM-1, MKT-BR, and SP-6.5) derived from human choroidal melanoma, the most frequent primary ocular neoplasm in humans, express B-Raf containing the V599E mutation. These melanoma cells showed a 10-fold increase in endogenous B-RafV599E kinase activity and a constitutive activation of the MEK/ERK pathway that is independent of Ras. This, as well as melanoma cell proliferation, was strongly diminished by siRNA-mediated depletion of the mutant B-Raf protein. Moreover, blocking B-RafV599E-induced ERK activation by different experimental approaches significantly reduced cell proliferation and anchorage-independent growth of melanoma cells. Finally, quantitative immunoblot analysis allowed us to identify signaling and cell cycle proteins that are differentially expressed between normal melanocytes and melanoma cells. Although the expression of signaling molecules was not sensitive to U0126 in melanoma cells, the expression of a cluster of cell cycle proteins remained regulated by the B-RafV599E/MEK/ERK pathway. Our results pinpoint this pathway as an important component in choroidal melanoma cell lines.


Subject(s)
Choroid Neoplasms/pathology , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutation , Oncogene Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Cell Cycle Proteins/analysis , Cell Division , Gene Expression Profiling , Humans , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase 3 , Oncogene Proteins/analysis , Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf , RNA, Small Interfering/pharmacology , Signal Transduction/genetics , Tumor Cells, Cultured , ras Proteins
20.
Invest Ophthalmol Vis Sci ; 44(3): 1320-9, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12601065

ABSTRACT

PURPOSE: Retinal pigment epithelial (RPE) cell death is an important step in the pathogenesis of ocular diseases. JNK1 and P38 kinase, two stress-activated kinases, play key roles relaying stress signals leading to cell death through cyclin D1 and c-Myc. Recently, stress-activated kinases have been shown to regulate cell proliferation. In the current study, the involvement of the JNK1 and P38 kinase signaling pathways in RPE cell proliferation and death was investigated. METHODS: RPE cell proliferation was stimulated with 10% fetal calf serum (FCS). Activation of the JNK1 and P38 kinase cascades and their potential targets was detected by Western blot analysis. Pharmacologic inhibitors and activators, and antisense oligodeoxynucleotides (ODN) directed against the stress kinases were used to analyze the signaling involved in RPE cell death. RESULTS: P38 and JNK1 and their respective upstream activating kinases, MKK3/6 and -4, were all transiently activated in FCS-stimulated RPE cell cultures. Ras controlled only the activation of JNK1, whereas Rho transmitted the activation of both JNK1 and P38, suggesting parallel signaling pathways and cross talk between the two kinases. Pharmacologic inhibition of JNK1 did not affect cell proliferation in FCS-stimulated cells. Inactivation of P38 kinase and antisense ODN-induced downregulation of P38 kinase also had no affect on cell proliferation. Long-term, high-level activation of JNK1 and P38 kinase occurred during serum depletion-induced RPE cell death. Overactivation of JNK1 and P38 kinase was also observed during pharmacologically induced cell death, suggesting that this process is common to RPE cell-death-signaling pathways induced by various stress stimuli. Cell death mediated by the overactivation of JNK1 and P38 kinase was cyclin D1- and c-Myc-independent. CONCLUSIONS: The inhibition of JNK1 or P38 kinase had no effect on FCS-stimulated proliferation of RPE cells, whereas the overactivation of these two enzymes was involved in RPE cell death in FCS-depleted cultures. Parallel upstream signaling pathways and cross talk between the two kinases suggest that the regulation of signaling in RPE cell death is complex.


Subject(s)
Apoptosis , MAP Kinase Kinase 4 , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Pigment Epithelium of Eye/pathology , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division , Cells, Cultured , Down-Regulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Kinase 3 , MAP Kinase Kinase 6 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pigment Epithelium of Eye/enzymology , Protein-Tyrosine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...