288 results on '"Cremers, Fp"'
Search Results
2. 6442 Retinal Phenotypes in Patients Homozygous for the G1961E Mutation in the ABCA4 gene
- Author
-
Burke, Tomas, primary, Fishman, GA, additional, Zernant, J, additional, Schuber, C, additional, Tsang, SH, additional, Smith, RT, additional, Ayyagari, R, additional, Koenekoop, RK, additional, Umfress, A, additional, Ciccarelli, ML, additional, Baldi, A, additional, Iannaccone, A, additional, Cremers, FP, additional, Klaver, CCW, additional, and Allikmets, R, additional
- Published
- 2012
- Full Text
- View/download PDF
3. The ABCA4 2588G>C Stargardt mutation: single origin and increasingfrequency from South-West to North-East Europe.
- Author
-
Maugeri, A, Flothmann, K, Hemmrich, N, Ingvast, S, Jorge, P, Paloma, E, Patel, R, Rozet, JM, Tammur, J, Testa, F, Balcells, S, Bird, AC, Brunner, HG, Hoyng, CB, Metspalu, A, Simonelli, F, Allikmets, R, Bhattacharya, SS, D'Urso, M, Gonzalez-Duarte, R, Kaplan, J, te Meerman, GJ, Santos, R, Schwartz, M, Van Camp, G, Wadelius, C, Weber, BH, Cremers, FP, Maugeri, A, Flothmann, K, Hemmrich, N, Ingvast, S, Jorge, P, Paloma, E, Patel, R, Rozet, JM, Tammur, J, Testa, F, Balcells, S, Bird, AC, Brunner, HG, Hoyng, CB, Metspalu, A, Simonelli, F, Allikmets, R, Bhattacharya, SS, D'Urso, M, Gonzalez-Duarte, R, Kaplan, J, te Meerman, GJ, Santos, R, Schwartz, M, Van Camp, G, Wadelius, C, Weber, BH, and Cremers, FP
- Published
- 2002
4. The spectrum of retinal dystrophies caused by mutations in the peripherin/RDS gene.
- Author
-
Boon CJ, den Hollander AI, Hoyng CB, Cremers FP, Klevering BJ, Keunen JE, Boon, Camiel J F, den Hollander, Anneke I, Hoyng, Carel B, Cremers, Frans P M, Klevering, B Jeroen, and Keunen, Jan E E
- Abstract
Peripherin/rds is an integral membrane glycoprotein, mainly located in the rod and cone outer segments. The relevance of this protein to photoreceptor outer segment morphology was first demonstrated in retinal degeneration slow (rds) mice. Thus far, over 90 human peripherin/RDS gene mutations have been identified. These mutations have been associated with a variety of retinal dystrophies, in which there is a remarkable inter- and intrafamilial variation of the retinal phenotype. In this paper, we discuss the characteristics of the peripherin/RDS gene and its protein product. An overview is presented of the broad spectrum of clinical phenotypes caused by human peripherin/RDS gene mutations, ranging from various macular dystrophies to widespread forms of retinal dystrophy such as retinitis pigmentosa. Finally, we review the proposed genotype-phenotype correlation and the pathophysiologic mechanisms underlying this group of retinal dystrophies. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
5. Mutations in IMPG1 Cause Vitelliform Macular Dystrophies
- Author
-
Hélène Dollfus, Susanne Kohl, Maxime Hebrard, Philippe Brabet, Isabelle Meunier, Audrey Sénéchal, Elfride De Baere, Carmen Ayuso García, Christina Zeitz, Béatrice Bocquet, Sandro Banfi, Guylène Le Meur, Claire Marie Dhaenens, Delphine Allorge, Frans P.M. Cremers, Francesca Simonelli, Michel Weber, Joe G. Hollyfield, Saddek Mohand-Said, Christian P. Hamel, Gaël Manes, Marta Corton, Xavier Zanlonghi, Robert K. Koenekoop, Gilles Labesse, Almudena Avila-Fernandez, José-Alain Sahel, Isabelle Audo, Manes, G, Meunier, I, Avila Fernández, A, Banfi, Sandro, Le Meur, G, Zanlonghi, X, Corton, M, Simonelli, Francesca, Brabet, P, Labesse, G, Audo, I, Mohand Said, S, Zeitz, C, Sahel, Ja, Weber, M, Dollfus, H, Dhaenens, Cm, Allorge, D, De Baere, E, Koenekoop, Rk, Kohl, S, Cremers, Fp, Hollyfield, Jg, Sénéchal, A, Hebrard, M, Bocquet, B, Ayuso García, C, and Hamel, Cp
- Subjects
Adult ,Male ,Proband ,Genetics and epigenetic pathways of disease [NCMLS 6] ,genetic structures ,Fundus Oculi ,Molecular Sequence Data ,Inheritance Patterns ,Vitelliform macular dystrophy ,Biology ,Interphotoreceptor matrix ,medicine.disease_cause ,Compound heterozygosity ,Young Adult ,Report ,medicine ,Genetics ,Chromosomes, Human ,Humans ,Genetic Predisposition to Disease ,Genetics(clinical) ,Amino Acid Sequence ,Eye Proteins ,Genetics (clinical) ,Extracellular Matrix Proteins ,Mutation ,Base Sequence ,Genetic heterogeneity ,Middle Aged ,Macular dystrophy ,medicine.disease ,eye diseases ,Pedigree ,Vitelliform Macular Dystrophy ,Phenotype ,Female ,Proteoglycans ,sense organs ,Retinal Dystrophies - Abstract
Item does not contain fulltext Vitelliform macular dystrophies (VMD) are inherited retinal dystrophies characterized by yellow, round deposits visible upon fundus examination and encountered in individuals with juvenile Best macular dystrophy (BMD) or adult-onset vitelliform macular dystrophy (AVMD). Although many BMD and some AVMD cases harbor mutations in BEST1 or PRPH2, the underlying genetic cause remains unknown for many affected individuals. In a large family with autosomal-dominant VMD, gene mapping and whole-exome sequencing led to the identification of a c.713T>G (p.Leu238Arg) IMPG1 mutation, which was subsequently found in two other families with autosomal-dominant VMD and the same phenotype. IMPG1 encodes the SPACR protein, a component of the rod and cone photoreceptor extracellular matrix domains. Structural modeling indicates that the p.Leu238Arg substitution destabilizes the conserved SEA1 domain of SPACR. Screening of 144 probands who had various forms of macular dystrophy revealed three other IMPG1 mutations. Two individuals from one family affected by autosomal-recessive VMD were homozygous for the splice-site mutation c.807+1G>T, and two from another family were compound heterozygous for the mutations c.461T>C (p.Leu154Pro) and c.1519C>T (p.Arg507( *)). Most cases had a normal or moderately decreased electrooculogram Arden ratio. We conclude that IMPG1 mutations cause both autosomal-dominant and -recessive forms of VMD, thus indicating that impairment of the interphotoreceptor matrix might be a general cause of VMD.
- Published
- 2013
- Full Text
- View/download PDF
6. Retinal Phenotypes in Patients Homozygous for the G1961E Mutation in the ABCA4 Gene
- Author
-
Tomas R. Burke, Robert K. Koenekoop, Caroline C W Klaver, Alfonso Baldi, F. P. M. Cremers, Jana Zernant, Alessandro Iannaccone, Radha Ayyagari, Carl Schubert, Allison C. Umfress, R. T. Smith, Rando Allikmets, Maria Laura Ciccarelli, Stephen H. Tsang, Gerald A. Fishman, Burke, Tr, Fishman, Ga, Zernant, J, Shubert, C, Tsang, Sh, Smith, Rt, Ayyagari, R, Koenekoop, Rk, Umfress, A, Ciccarelli, Ml, Baldi, Alfonso, Iannaccone, A, Cremers, Fp, Klaver, Cc, Allikmets, R., and Ophthalmology
- Subjects
Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,biology ,Genetics and epigenetic pathways of disease [NCMLS 6] ,Fundus photography ,ABCA4 ,Articles ,Macular degeneration ,Fundus (eye) ,medicine.disease ,Fluorescein angiography ,eye diseases ,Stargardt disease ,Mutation (genetic algorithm) ,medicine ,biology.protein ,Age of onset - Abstract
PURPOSE. We evaluated the pathogenicity of the G1961E mutation in the ABCA4 gene, and present the range of retinal phenotypes associated with this mutation in homozygosity in a patient cohort with ABCA4-associated phenotypes. METHODS. Patients were enrolled from the ABCA4 disease database at Columbia University or by inquiry from collaborating physicians. Only patients homozygous for the G1961E mutation were enrolled. The entire ABCA4 gene open reading frame, including all exons and flanking intronic sequences, was sequenced in all patients. Phenotype data were obtained from clinical history and examination, fundus photography, infrared imaging, fundus autofluorescence, fluorescein angiography, and spectral domain-optical coherence tomography. Additional functional data were obtained using the full-field electroretinogram, and static or kinetic perimetry. RESULTS. We evaluated 12 patients homozygous for the G1961E mutation. All patients had evidence of retinal pathology consistent with the range of phenotypes observed in ABCA4 disease. The latest age of onset was recorded at 64 years, in a patient diagnosed initially with age-related macular degeneration (AMD). Of 6 patients in whom severe structural (with/ without functional) fundus changes were detected, 5 had additional, heterozygous or homozygous, variants detected in the ABCA4 gene. CONCLUSIONS. Homozygous G1961E mutation in ABCA4 results in a range of retinal pathology. The phenotype usually is at the milder end of the disease spectrum, with severe phenotypes linked to the presence of additional ABCA4 variants. Our report also highlights that milder, late-onset Stargardt disease may be confused with AMD. (Invest Ophthalmol Vis Sci. 2012; 53:4458–4467) DOI:10.1167/iovs.11-9166
- Published
- 2012
- Full Text
- View/download PDF
7. Development of a genotyping microarray for Usher syndrome
- Author
-
Andrew R. Webster, Robert K. Koenekoop, Zubin Saihan, P.M. Kelley, Elfride De Baere, Arjan P.M. de Brouwer, Maigi Külm, Elene Haralambous, Eeva-Marja Sankila, Erwin van Wijk, Johannes Fleischhauer, Sandro Banfi, Michael D. Weston, William J. Kimberling, Dominique Weil, Gareth J. McKay, Bart P. Leroy, José M. Millán, Lies H. Hoefsloot, Francesca Simonelli, Cor W. R. J. Cremers, Tarja Joensuu, Thomas Rosenberg, Giuliana Silvestri, Heleen te Brinke, Bernd Wissinger, Maria Bitner-Glindzicz, Hannie Kremer, Wolfgang Berger, Frans P.M. Cremers, Cremers, Fp, Kimberling, Wj, Kulm, M, DE BROUWER, A, VAN WIJK, E, TE BRINKE, H, Cremers, Cw, Hoefsloot, Lh, Banfi, Sandro, Simonelli, Francesca, Fleischhauer, Jc, Berger, W, Kelley, Pm, Haralambous, E, BITNER GLINDZICZ, M, Webster, Ar, Saihan, Z, DE BAERE, E, Leroy, Bp, Silvestri, G, Mckay, G, Koenekoop, Rk, Millan, Jm, Rosenberg, T, Joensuu, T, Sankila, Em, Weil, D, Weston, Md, Wissinger, B, and Kremer, H.
- Subjects
RECESSIVE RETINITIS-PIGMENTOSA ,Microarray ,Genetics and epigenetic pathways of disease [NCMLS 6] ,Genotype ,Hearing loss ,Usher syndrome ,LEBER CONGENITAL AMAUROSIS ,Biology ,USH2A GENE ,EAR SENSORY CELLS ,Genomic disorders and inherited multi-system disorders [IGMD 3] ,03 medical and health sciences ,SYNDROME TYPE 1F ,MUTATION DETECTION ,SYNDROME-TYPE-II ,Retinitis pigmentosa ,Genetics ,medicine ,Medicine and Health Sciences ,Perception and Action [DCN 1] ,otorhinolaryngologic diseases ,Neurosensory disorders [UMCN 3.3] ,Humans ,Genotyping ,Genetics (clinical) ,030304 developmental biology ,DNA Primers ,Oligonucleotide Array Sequence Analysis ,0303 health sciences ,Genetic heterogeneity ,HEARING-LOSS ,030305 genetics & heredity ,Genetic Variation ,DNA ,medicine.disease ,ARRAYED PRIMER EXTENSION ,eye diseases ,3. Good health ,MYOSIN VIIA GENE ,Europe ,Genetic defects of metabolism [UMCN 5.1] ,sense organs ,medicine.symptom ,Functional Neurogenomics [DCN 2] ,Usher Syndromes ,PCDH15 ,Letter to JMG - Abstract
Contains fulltext : 52397.pdf (Publisher’s version ) (Closed access) BACKGROUND: Usher syndrome, a combination of retinitis pigmentosa (RP) and sensorineural hearing loss with or without vestibular dysfunction, displays a high degree of clinical and genetic heterogeneity. Three clinical subtypes can be distinguished, based on the age of onset and severity of the hearing impairment, and the presence or absence of vestibular abnormalities. Thus far, eight genes have been implicated in the syndrome, together comprising 347 protein-coding exons. METHODS: To improve DNA diagnostics for patients with Usher syndrome, we developed a genotyping microarray based on the arrayed primer extension (APEX) method. Allele-specific oligonucleotides corresponding to all 298 Usher syndrome-associated sequence variants known to date, 76 of which are novel, were arrayed. RESULTS: Approximately half of these variants were validated using original patient DNAs, which yielded an accuracy of >98%. The efficiency of the Usher genotyping microarray was tested using DNAs from 370 unrelated European and American patients with Usher syndrome. Sequence variants were identified in 64/140 (46%) patients with Usher syndrome type I, 45/189 (24%) patients with Usher syndrome type II, 6/21 (29%) patients with Usher syndrome type III and 6/20 (30%) patients with atypical Usher syndrome. The chip also identified two novel sequence variants, c.400C>T (p.R134X) in PCDH15 and c.1606T>C (p.C536S) in USH2A. CONCLUSION: The Usher genotyping microarray is a versatile and affordable screening tool for Usher syndrome. Its efficiency will improve with the addition of novel sequence variants with minimal extra costs, making it a very useful first-pass screening tool.
- Published
- 2007
8. Genotyping microarray (gene chip) for theABCR(ABCA4) gene
- Author
-
M. Hawlina, Jana Zernant, Carel B. Hoyng, Alessandra Maugeri, James R. Lupski, Francesco Testa, Metka Ravnik-Glavač, M.R. Meltzer, Maigi Külm, Neeme Tõnisson, Rafael C. Caruso, Rando Allikmets, Peter Gouras, Amy Hutchinson, K. Jaakson, D. Glavac, F.P.M. Cremers, Francesca Simonelli, Richard A. Lewis, Jaakson, K, Zernant, J, Külm, M, Hutchinson, A, Tonisson, N, Glavac, D, Ravnik Glavac, M, Hawlina, M, Meltzer, Mr, Caruso, Rc, Testa, Francesco, Maugeri, A, Hoyng, Cb, Gouras, P, Simonelli, Francesca, Lewis, Ra, Lupski, Jr, Cremers, Fp, and Allikmets, R.
- Subjects
Genotype ,Microarray ,DNA Mutational Analysis ,Population ,ABCA4 ,Retinal Diseases ,Genetics ,medicine ,Humans ,Neurosensory disorders [UMCN 3.3] ,education ,Genotyping ,Genetics (clinical) ,Oligonucleotide Array Sequence Analysis ,education.field_of_study ,Polymorphism, Genetic ,biology ,Genetic Variation ,Reproducibility of Results ,medicine.disease ,Stargardt disease ,Genetic defects of metabolism [UMCN 5.1] ,Gene chip analysis ,biology.protein ,ATP-Binding Cassette Transporters ,Allelic heterogeneity ,DNA microarray - Abstract
Item does not contain fulltext Genetic variation in the ABCR (ABCA4) gene has been associated with five distinct retinal phenotypes, including Stargardt disease/fundus flavimaculatus (STGD/FFM), cone-rod dystrophy (CRD), and age-related macular degeneration (AMD). Comparative genetic analyses of ABCR variation and diagnostics have been complicated by substantial allelic heterogeneity and by differences in screening methods. To overcome these limitations, we designed a genotyping microarray (gene chip) for ABCR that includes all approximately 400 disease-associated and other variants currently described, enabling simultaneous detection of all known ABCR variants. The ABCR genotyping microarray (the ABCR400 chip) was constructed by the arrayed primer extension (APEX) technology. Each sequence change in ABCR was included on the chip by synthesis and application of sequence-specific oligonucleotides. We validated the chip by screening 136 confirmed STGD patients and 96 healthy controls, each of whom we had analyzed previously by single strand conformation polymorphism (SSCP) technology and/or heteroduplex analysis. The microarray was >98% effective in determining the existing genetic variation and was comparable to direct sequencing in that it yielded many sequence changes undetected by SSCP. In STGD patient cohorts, the efficiency of the array to detect disease-associated alleles was between 54% and 78%, depending on the ethnic composition and degree of clinical and molecular characterization of a cohort. In addition, chip analysis suggested a high carrier frequency (up to 1:10) of ABCR variants in the general population. The ABCR genotyping microarray is a robust, cost-effective, and comprehensive screening tool for variation in one gene in which mutations are responsible for a substantial fraction of retinal disease. The ABCR chip is a prototype for the next generation of screening and diagnostic tools in ophthalmic genetics, bridging clinical and scientific research.
- Published
- 2003
- Full Text
- View/download PDF
9. Mutations in MFSD8, encoding a lysosomal membrane protein, are associated with nonsyndromic autosomal recessive macular dystrophy
- Author
-
Carel B. Hoyng, Frans P.M. Cremers, L. Ingeborgh van den Born, Sandro Banfi, Riccardo Sangermano, Susanne Roosing, Marijke N. Zonneveld-Vrieling, Caroline C W Klaver, Janneke J.C. van Lith-Verhoeven, Anneke I. den Hollander, Robert K. Koenekoop, Ophthalmology, Roosing, S, van den Born, Li, Sangermano, R, Banfi, Sandro, Koenekoop, Rk, Zonneveld Vrieling, Mn, Klaver, Cc, van Lith Verhoeven, Jj, Cremers, Fp, den Hollander, Ai, and Hoyng, Cb
- Subjects
Adult ,Male ,Proband ,Pathology ,medicine.medical_specialty ,Achromatopsia ,genetic structures ,DNA Mutational Analysis ,Sensory disorders Radboud Institute for Health Sciences [Radboudumc 12] ,Genome-wide association study ,Compound heterozygosity ,Macular Degeneration ,Cone dystrophy ,Electroretinography ,medicine ,Humans ,Exome ,Sensory disorders Radboud Institute for Molecular Life Sciences [Radboudumc 12] ,Exome sequencing ,Aged ,Genetics ,business.industry ,Lysosome-Associated Membrane Glycoproteins ,Membrane Transport Proteins ,Middle Aged ,Macular dystrophy ,medicine.disease ,eye diseases ,Pedigree ,Ophthalmology ,Mutation ,Visual Field Tests ,Female ,Neuronal ceroid lipofuscinosis ,sense organs ,business ,Tomography, Optical Coherence ,Genome-Wide Association Study - Abstract
Purpose: This study aimed to identify the genetic defects in 2 families with autosomal recessive macular dystrophy with central cone involvement. Design: Case series. Participants: Two families and a cohort of 244 individuals with various inherited maculopathies and cone disorders. Methods: Genome-wide linkage analysis and exome sequencing were performed in 1 large family with 5 affected individuals. In addition, exome sequencing was performed in the proband of a second family. Subsequent analysis of the identified mutations in 244 patients was performed by Sanger sequencing or restriction enzyme digestion. The medical history of individuals carrying the MFSD8 variants was reviewed and additional ophthalmic examinations were performed, including electroretinography (ERG), multifocal ERG (mfERG), perimetry, optical coherence tomography (OCT), fundus autofluorescence, and fundus photography. Main Outcome Measures: MFSD8 variants, age at diagnosis, visual acuity, fundus appearance, color vision defects, visual field, ERG, mfERG, fundus autofluorescence, and OCT findings. Results: Compound heterozygous variants in MFSD8, a gene encoding a lysosomal transmembrane protein, were identified in 2 families with macular dystrophy with a normal or subnormal ERG, but reduced mfERG. In both families, a heterozygous missense variant p.Glu336Gln was identified, which was predicted to have a mild effect on the protein. In the first family, a protein-truncating variant (p.Glu381*) was identified on the other allele, and in the second family, a variant (c.1102G>C) was identified that results in a splicing defect leading to skipping of exon 11 (p.Lys333Lysfs*3). The p.Glu336Gln allele was found to be significantly enriched in patients with maculopathies and cone disorders (6/488) compared with ethnically matched controls (35/18 682; P < 0.0001), suggesting that it may act as a genetic modifier. Conclusions: In this study, we identified variants in MFSD8 as a novel cause of nonsyndromic autosomal recessive macular dystrophy with central cone involvement. Affected individuals showed no neurologic features typical for variant late-infantile neuronal ceroid lipofuscinosis (vLINCL), a severe and devastating multisystem lysosomal storage disease previously associated with mutations in MFSD8. We propose a genotype-phenotype model in which a combination of a severe and a mild variant cause nonsyndromic macular dystrophy with central cone involvement, and 2 severe mutations cause vLINCL. (C) 2015 by the American Academy of Ophthalmology.
- Published
- 2015
- Full Text
- View/download PDF
10. Union makes strength: a worldwide collaborative genetic and clinical study to provide a comprehensive survey of RD3 mutations and delineate the associated phenotype
- Author
-
Bernd Wissinger, Renate C. Zekveld-Vroon, Alejandro Estrada-Cuzcano, Sandro Banfi, Jean-Michel Rozet, Ditta Zobor, Esther Pomares, Isabelle Perrault, Huanan Ren, Shiqiang Li, Susanne Kohl, Frans P.M. Cremers, André Mégarbané, Anneke I. den Hollander, Ralph J. Florijn, Rui Chen, Marc Jeanpierre, Irma Lopez, Francesco Testa, Nisrine Aboussair, Roser Gonzàlez-Duarte, Blanca C. Flores, Corinne Leowski, Francesca Simonelli, Edwin M. Stone, Josseline Kaplan, Qingjiong Zhang, Catherine Edelson, Arthur A.B. Bergen, Jean Andorf, Arnold Munnich, Cristina Villanueva, Nathalie Delphin, Robert K. Koenekoop, Xia Wang, Universitat de Barcelona, Perrault, I, Estrada Cuzcano, A, Lopez, I, Kohl, S, Li, S, Testa, Francesco, Zekveld Vroon, R, Wang, X, Pomares, E, Andorf, J, Aboussair, N, Banfi, Sandro, Delphin, N, den Hollander, Ai, Edelson, C, Florijn, R, Jean Pierre, M, Leowski, C, Megarbane, A, Villanueva, C, Flores, B, Munnich, A, Ren, H, Zobor, D, Bergen, A, Chen, R, Cremers, Fp, Gonzalez Duarte, R, Koenekoop, Rk, Simonelli, Francesca, Stone, E, Wissinger, B, Zhang, Q, Kaplan, J, Rozet, Jm, Human Genetics, ANS - Amsterdam Neuroscience, and ARD - Amsterdam Reproduction and Development
- Subjects
Male ,Retinal degeneration ,Photoreceptors ,Genetic Screens ,genetic structures ,Genetics and epigenetic pathways of disease [NCMLS 6] ,Leber Congenital Amaurosis ,lcsh:Medicine ,Social and Behavioral Sciences ,medicine.disease_cause ,Linkage Disequilibrium ,Fotoreceptors ,Cohort Studies ,0302 clinical medicine ,Sociology ,Human Families ,lcsh:Science ,Child ,Genetics ,0303 health sciences ,Mutation ,Multidisciplinary ,Retinal Degeneration ,Pedigree ,3. Good health ,Europe ,Phenotype ,Child, Preschool ,Medicine ,Retinal Disorders ,GUCY2D ,Female ,Research Article ,Adult ,Canada ,China ,Adolescent ,Nonsense mutation ,Genetic Counseling ,Biology ,Retina ,Frameshift mutation ,Young Adult ,03 medical and health sciences ,Genetic Mutation ,medicine ,Humans ,Allele ,Eye Proteins ,030304 developmental biology ,Polymorphism, Genetic ,lcsh:R ,Mutació (Biologia) ,Infant ,Human Genetics ,Heterozygote advantage ,Mutation (Biology) ,medicine.disease ,United States ,eye diseases ,Ophthalmology ,Genetics of Disease ,Genetic Polymorphism ,Pediatric Ophthalmology ,030221 ophthalmology & optometry ,lcsh:Q ,sense organs ,Genetics and epigenetic pathways of disease Genomic disorders and inherited multi-system disorders [NCMLS 6] ,Population Genetics ,Founder effect - Abstract
Contains fulltext : 117915.pdf (Publisher’s version ) (Open Access) Leber congenital amaurosis (LCA) is the earliest and most severe retinal degeneration (RD), and the most common cause of incurable blindness diagnosed in children. It is occasionally the presenting symptom of multisystemic ciliopathies which diagnosis will require a specific care of patients. Nineteen LCA genes are currently identified and three of them account for both non-syndromic and syndromic forms of the disease. RD3 (LCA12) was implicated as a LCA gene based on the identification of homozygous truncating mutations in two LCA families despite the screening of large cohorts of patients. Here we provide a comprehensive survey of RD3 mutations and of their clinical expression through the screening of a cohort of 852 patients originating worldwide affected with LCA or early-onset and severe RD. We identified three RD3 mutations in seven unrelated consanguineous LCA families - i.e., a 2 bp deletion and two nonsense mutations - predicted to cause complete loss of function. Five families originating from the Southern Shores of the Mediterranean segregated a similar mutation (c.112C>T, p.R38*) suggesting that this change may have resulted from an ancient founder effect. Considering the low frequency of RD3 carriers, the recurrence risk for LCA in non-consanguineous unions is negligible for both heterozygote and homozygote RD3 individuals. The LCA12 phenotype in our patients is highly similar to those of patients with mutant photoreceptor-specific guanylate cyclase (GUCY2D/LCA1). This observation is consistent with the report of the role of RD3 in trafficking of GUCYs and gives further support to a common mechanism of photoreceptor degeneration in LCA12 and LCA1, i.e., inability to increase cytoplasmic cGMP concentration in outer segments and thus to recover the dark-state. Similar to LCA1, LCA12 patients have no extraocular symptoms despite complete inactivation of both RD3 alleles, supporting the view that extraocular investigations in LCA infants with RD3 mutations should be avoided.
- Published
- 2013
11. Mutations in IMPG2, Encoding Interphotoreceptor Matrix Proteoglycan 2, Cause Autosomal-Recessive Retinitis Pigmentosa
- Author
-
Ellen A.W. Blokland, Frans P.M. Cremers, Lina Zelinger, Dikla Bandah-Rozenfeld, Dirk J. Lefeber, Tim M. Strom, Karlien L.M. Coene, Francesco Testa, Inbar Erdinest, Caroline C W Klaver, Francesca Simonelli, Anneke I. den Hollander, Krysta Voesenek, L. Ingeborgh van den Born, Anna M. Siemiatkowska, Raheel Qamar, Rob W.J. Collin, Muhammad Imran Khan, Dror Sharon, Sandro Banfi, Eyal Banin, Hematology, Ophthalmology, Bandah Rozenfeld, D, Collin, Rw, Banin, E, Ingeborgh van den Born, L, Coene, Kl, Siemiatkowska, Am, Zelinger, L, Khan, Mi, Lefeber, Dj, Erdinest, I, Testa, Francesco, Simonelli, Francesca, Voesenek, K, Blokland, Ea, Strom, Tm, Klaver, Cc, Qamar, R, Banfi, Sandro, Cremers, Fp, Sharon, D, and den Hollander, Ai
- Subjects
Adult ,Male ,Genetics and epigenetic pathways of disease [NCMLS 6] ,Fundus Oculi ,Genetic Linkage ,Nonsense mutation ,DNA Mutational Analysis ,Molecular Sequence Data ,Genes, Recessive ,Gene mutation ,Biology ,Interphotoreceptor matrix ,Neuroinformatics [DCN 3] ,medicine.disease_cause ,Article ,Genomic disorders and inherited multi-system disorders [IGMD 3] ,Exon ,Chromosome Segregation ,Retinitis pigmentosa ,Chlorocebus aethiops ,medicine ,Genetics ,Missense mutation ,Animals ,Humans ,Genetics(clinical) ,Amino Acid Sequence ,Genetics (clinical) ,Aged ,Mutation ,Base Sequence ,Homozygote ,Chromosome Mapping ,Glycostation disorders [IGMD 4] ,Middle Aged ,medicine.disease ,Pedigree ,COS Cells ,Mutation testing ,Female ,Mutant Proteins ,Proteoglycans ,Retinitis Pigmentosa ,Subcellular Fractions - Abstract
Contains fulltext : 89392.pdf (Publisher’s version ) (Closed access) Retinitis pigmentosa (RP) is a heterogeneous group of inherited retinal diseases caused by progressive degeneration of the photoreceptor cells. Using autozygosity mapping, we identified two families, each with three affected siblings sharing large overlapping homozygous regions that harbored the IMPG2 gene on chromosome 3. Sequence analysis of IMPG2 in the two index cases revealed homozygous mutations cosegregating with the disease in the respective families: three affected siblings of Iraqi Jewish ancestry displayed a nonsense mutation, and a Dutch family displayed a 1.8 kb genomic deletion that removes exon 9 and results in the absence of seven amino acids in a conserved SEA domain of the IMPG2 protein. Transient transfection of COS-1 cells showed that a construct expressing the wild-type SEA domain is properly targeted to the plasma membrane, whereas the mutant lacking the seven amino acids appears to be retained in the endoplasmic reticulum. Mutation analysis in ten additional index cases that were of Dutch, Israeli, Italian, and Pakistani origin and had homozygous regions encompassing IMPG2 revealed five additional mutations; four nonsense mutations and one missense mutation affecting a highly conserved phenylalanine residue. Most patients with IMPG2 mutations showed an early-onset form of RP with progressive visual-field loss and deterioration of visual acuity. The patient with the missense mutation, however, was diagnosed with maculopathy. The IMPG2 gene encodes the interphotoreceptor matrix proteoglycan IMPG2, which is a constituent of the interphotoreceptor matrix. Our data therefore show that mutations in a structural component of the interphotoreceptor matrix can cause arRP.
- Published
- 2010
12. The ABCA4 2588G>C Stargardt mutation: single origin and increasing frequency from South-West to North-East Europe
- Author
-
Francesco Testa, N Hemmrich, G. Van Camp, R Santoss, J. C. Kaplan, S. Ingvast, Kris Flothmann, Francesca Simonelli, E Paloma, Carel B. Hoyng, Michele D'Urso, Alessandra Maugeri, Andres Metspalu, Reshma Patel, Paula Jorge, C Wadelius, Roser Gonzàlez-Duarte, Shomi S. Bhattacharya, Han G. Brunner, A C Bird, J Tammur, F. P. M. Cremers, Gjt Meerman, Susana Balcells, Jean-Michel Rozet, Marianne Schwartz, Bhf Weber, Rando Allikmets, Maugeri, A, Flothmann, K, Hemmrich, N, Ingvast, S, Jorge, P, Paloma, E, Patel, R, Rozet, Jm, Tammur, J, Testa, Francesco, Balcells, S, Bird, Ac, Brunner, Hg, Hoyng, Cb, Metspalu, A, Simonelli, Francesca, Allikmets, R, Bhattacharya, S, D'Urso, M, Gonzàlez Duarte, R, Kaplan, J, Te Meerman, Gj, Santos, R, Schwartz, M, Van Camp, G, Wadelius, C, Weber, Bh, and Cremers, Fp
- Subjects
carrier frequency ,EXPRESSION ,Heterozygote ,FOUNDER ,RECESSIVE RETINITIS-PIGMENTOSA ,Elucidation of hereditary disorders and their molecular diagnosis ,Molecular Sequence Data ,ABCA4 ,Locus (genetics) ,CONE-ROD DYSTROPHY ,PHENOTYPE ,Gene Frequency ,TRANSPORTER GENE ,Genetics ,Humans ,Point Mutation ,experimenteel en klinisch onderzoek en behandeling. [Erfelijke en verworven vitreo-retinale aandoeningen] ,Allele ,Gene ,Allele frequency ,Alleles ,Genetics (clinical) ,PHOTORECEPTORS ,Base Sequence ,biology ,Point mutation ,Haplotype ,ABCR ,Heterozygote advantage ,MACULAR DEGENERATION ,United States ,Europe ,retinal dystrophies ,Mutation ,biology.protein ,founder mutation ,POPULATIONS ,ATP-Binding Cassette Transporters ,experimental and clinical research and treatment. [Hereditary and acquired vitreo-retinal disorders] ,DISEASE GENE ABCR ,Opheldering van erfelijke ziekten en hun moleculaire diagnostiek ,STGD - Abstract
Item does not contain fulltext Inherited retinal dystrophies represent the most important cause of vision impairment in adolescence, affecting approximately 1 out of 3000 individuals. Mutations of the photoreceptor-specific gene ABCA4 (ABCR) are a common cause of retinal dystrophy. A number of mutations have been repeatedly reported for this gene, notably the 2588G>C mutation which is frequent in both patients and controls. Here we ascertained the frequency of the 2588G>C mutation in a total of 2343 unrelated random control individuals from 11 European countries and 241 control individuals from the US, as well as in 614 patients with STGD both from Europe and the US. We found an overall carrier frequency of 1 out of 54 in Europe, compared with 1 out of 121 in the US, confirming that the 2588G>C ABCA4 mutation is one of the most frequent autosomal recessive mutations in the European population. Carrier frequencies show an increasing gradient in Europe from South-West to North-East. The lowest carrier frequency, 0 out of 199 (0%), was found in Portugal; the highest, 11 out of 197 (5.5%), was found in Sweden. Haplotype analysis in 16 families segregating the 2588G>C mutation showed four intragenic polymorphisms invariably present in all 16 disease chromosomes and sharing of the same allele for several markers flanking the ABCA4 locus in most of the disease chromosomes. These results indicate a single origin of the 2588G>C mutation which, to our best estimate, occurred between 2400 and 3000 years ago.
- Published
- 2002
13. Homozygosity Mapping and Targeted Sanger Sequencing Identifies Three Novel CRB1 (Crumbs homologue 1) Mutations in Iranian Retinal Degeneration Families
- Author
-
Ghofrani M, Yahyaei M, Brunner HG, Cremers FP, Movasat M, Imran Khan M, and Keramatipour M
- Abstract
Background: Inherited retinal diseases (IRDs) are a group of genetic disorders with high degrees of clinical, genetic and allelic heterogeneity. IRDs generally show progressive retinal cell death resulting in gradual vision loss. IRDs constitute a broad spectrum of disorders including retinitis pigmentosa and Leber congenital amaurosis. In this study, we performed genotyping studies to identify the underlying mutations in three Iranian families., Method: Having employed homozygosity mapping and Sanger sequencing, we identified the underlying mutations in the crumbs homologue 1 gene. The CRB1 protein is a part of a macromolecular complex with a vital role in retinal cell polarity, morphogenesis, and maintenance., Results: We identified a novel homozygous variant (c.1053_1061del; p.Gly352_Cys354del) in one family, a combination of a novel (c.2086T>C; p.Cys696Arg) and a known variant (c.2234C>T, p.Thr745Met) in another family and a homozygous novel variant (c.3090T>A; p.Asn1030Lys) in a third family., Conclusion: This study shows that mutations in CRB1 are relatively common in Iranian non-syndromic IRD patients.
- Published
- 2017
- Full Text
- View/download PDF
14. Diagnostic exome sequencing in 266 Dutch patients with visual impairment.
- Author
-
Haer-Wigman L, van Zelst-Stams WA, Pfundt R, van den Born LI, Klaver CC, Verheij JB, Hoyng CB, Breuning MH, Boon CJ, Kievit AJ, Verhoeven VJ, Pott JW, Sallevelt SC, van Hagen JM, Plomp AS, Kroes HY, Lelieveld SH, Hehir-Kwa JY, Castelein S, Nelen M, Scheffer H, Lugtenberg D, Cremers FP, Hoefsloot L, and Yntema HG
- Subjects
- Adolescent, Adult, Case-Control Studies, Child, Eye Diseases, Hereditary pathology, Humans, Netherlands, Vision Disorders pathology, Exome, Eye Diseases, Hereditary genetics, Inheritance Patterns, Vision Disorders genetics
- Abstract
Inherited eye disorders have a large clinical and genetic heterogeneity, which makes genetic diagnosis cumbersome. An exome-sequencing approach was developed in which data analysis was divided into two steps: the vision gene panel and exome analysis. In the vision gene panel analysis, variants in genes known to cause inherited eye disorders were assessed for pathogenicity. If no causative variants were detected and when the patient consented, the entire exome data was analyzed. A total of 266 Dutch patients with different types of inherited eye disorders, including inherited retinal dystrophies, cataract, developmental eye disorders and optic atrophy, were investigated. In the vision gene panel analysis (likely), causative variants were detected in 49% and in the exome analysis in an additional 2% of the patients. The highest detection rate of (likely) causative variants was in patients with inherited retinal dystrophies, for instance a yield of 63% in patients with retinitis pigmentosa. In patients with developmental eye defects, cataract and optic atrophy, the detection rate was 50, 33 and 17%, respectively. An exome-sequencing approach enables a genetic diagnosis in patients with different types of inherited eye disorders using one test. The exome approach has the same detection rate as targeted panel sequencing tests, but offers a number of advantages. For instance, the vision gene panel can be frequently and easily updated with additional (novel) eye disorder genes. Determination of the genetic diagnosis improved the clinical diagnosis, regarding the assessment of the inheritance pattern as well as future disease perspective.
- Published
- 2017
- Full Text
- View/download PDF
15. In Silico Functional Meta-Analysis of 5,962 ABCA4 Variants in 3,928 Retinal Dystrophy Cases.
- Author
-
Cornelis SS, Bax NM, Zernant J, Allikmets R, Fritsche LG, den Dunnen JT, Ajmal M, Hoyng CB, and Cremers FP
- Subjects
- Alleles, Gene Frequency, Genotype, Humans, Phenotype, Retinal Dystrophies pathology, Severity of Illness Index, ATP-Binding Cassette Transporters genetics, Computer Simulation, Mutation, Retinal Dystrophies genetics
- Abstract
Variants in the ABCA4 gene are associated with a spectrum of inherited retinal diseases (IRDs), most prominently with autosomal recessive (ar) Stargardt disease (STGD1) and ar cone-rod dystrophy. The clinical outcome to a large degree depends on the severity of the variants. To provide an accurate prognosis and to select patients for novel treatments, functional significance assessment of nontruncating ABCA4 variants is important. We collected all published ABCA4 variants from 3,928 retinal dystrophy cases in a Leiden Open Variation Database, and compared their frequency in 3,270 Caucasian IRD cases with 33,370 non-Finnish European control individuals. Next to the presence of 270 protein-truncating variants, 191 nontruncating variants were significantly enriched in the patient cohort. Furthermore, 30 variants were deemed benign. Assessing the homozygous occurrence of frequent variants in IRD cases based on the allele frequencies in control individuals confirmed the mild nature of the p.[Gly863Ala, Gly863del] variant and identified three additional mild variants (p.(Ala1038Val), c.5714+5G>A, and p.(Arg2030Gln)). The p.(Gly1961Glu) variant was predicted to act as a mild variant in most cases. Based on these data, in silico analyses, and American College of Medical Genetics and Genomics guidelines, we provide pathogenicity classifications on a five-tier scale from benign to pathogenic for all variants in the ABCA4-LOVD database., (© 2017 WILEY PERIODICALS, INC.)
- Published
- 2017
- Full Text
- View/download PDF
16. Putative digenic inheritance of heterozygous RP1L1 and C2orf71 null mutations in syndromic retinal dystrophy.
- Author
-
Liu YP, Bosch DG, Siemiatkowska AM, Rendtorff ND, Boonstra FN, Möller C, Tranebjærg L, Katsanis N, and Cremers FP
- Subjects
- Adult, Animals, Comparative Genomic Hybridization, DNA Mutational Analysis, Disease Models, Animal, Embryo, Nonmammalian, Exome genetics, Female, Gene Silencing, Heterozygote, Humans, Pedigree, Retinitis Pigmentosa pathology, Rhodopsin genetics, Sequence Analysis, DNA, Zebrafish embryology, Zebrafish Proteins genetics, Eye Proteins genetics, Genetic Predisposition to Disease, Inheritance Patterns, Mutation genetics, Retinitis Pigmentosa genetics
- Abstract
Background: Retinitis pigmentosa (RP) is the most common cause of inherited retinal degeneration and can occur in non-syndromic and syndromic forms. Syndromic RP is accompanied by other symptoms such as intellectual disability, hearing loss, or congenital abnormalities. Both forms are known to exhibit complex genetic interactions that can modulate the penetrance and expressivity of the phenotype., Materials and Methods: In an individual with atypical RP, hearing loss, ataxia and cerebellar atrophy, whole exome sequencing was performed. The candidate pathogenic variants were tested by developing an in vivo zebrafish model and assaying for retinal and cerebellar integrity., Results: Exome sequencing revealed a complex heterozygous protein-truncating mutation in RP1L1, p.[(Lys111Glnfs*27; Gln2373*)], and a heterozygous nonsense mutation in C2orf71, p.(Ser512*). Mutations in both genes have previously been implicated in autosomal recessive non-syndromic RP, raising the possibility of a digenic model in this family. Functional testing in a zebrafish model for two key phenotypes of the affected person showed that the combinatorial suppression of rp1l1 and c2orf71l induced discrete pathology in terms of reduction of eye size with concomitant loss of rhodopsin in the photoreceptors, and disorganization of the cerebellum., Conclusions: We propose that the combination of heterozygous loss-of-function mutations in these genes drives syndromic retinal dystrophy, likely through the genetic interaction of at least two loci. Haploinsufficiency at each of these loci is insufficient to induce overt pathology.
- Published
- 2017
- Full Text
- View/download PDF
17. Deletions Overlapping VCAN Exon 8 Are New Molecular Defects for Wagner Disease.
- Author
-
Burin-des-Roziers C, Rothschild PR, Layet V, Chen JM, Ghiotti T, Leroux C, Cremers FP, Brézin AP, and Valleix S
- Subjects
- Chromosome Breakpoints, DNA Mutational Analysis, Female, High-Throughput Nucleotide Sequencing, Humans, Male, Pedigree, Real-Time Polymerase Chain Reaction, Translocation, Genetic, Versicans genetics, Exons, Retinal Degeneration diagnosis, Retinal Degeneration genetics, Sequence Deletion, Versicans deficiency
- Abstract
Wagner disease is a rare nonsyndromic autosomal-dominant vitreoretinopathy, associated with splice mutations specifically targeting VCAN exon 8. We report the extensive genetic analysis of two Wagner probands, previously found negative for disease-associated splice mutations. Next-generation sequencing (NGS), quantitative real-time PCR, and long-range PCR identified two deletions (3.4 and 10.5 kb) removing at least one exon-intron boundary of exon 8, and both correlating with an imbalance of VCAN mRNA isoforms. We showed that the 10.5-kb deletion occurred de novo, causing somatic mosaicism in the proband's mother who had an unusually mild asymmetrical phenotype. Therefore, exon 8 deletions are novel VCAN genetic defects responsible for Wagner disease, and VCAN mosaic mutations may be involved in the pathogenesis of Wagner disease with attenuated phenotype. NGS is then an effective screening tool for genetic diagnosis of Wagner disease, improving the chance of identifying all disease-causative variants as well as mosaic mutations in VCAN., (© 2016 WILEY PERIODICALS, INC.)
- Published
- 2017
- Full Text
- View/download PDF
18. Asymmetric Inter-Eye Progression in Stargardt Disease.
- Author
-
Lambertus S, Bax NM, Groenewoud JM, Cremers FP, van der Wilt GJ, Klevering BJ, Theelen T, and Hoyng CB
- Subjects
- ATP-Binding Cassette Transporters genetics, ATP-Binding Cassette Transporters metabolism, Adolescent, Adult, Aged, Atrophy pathology, Child, Child, Preschool, Disease Progression, Electroretinography, Female, Fluorescein Angiography, Follow-Up Studies, Fundus Oculi, Genetic Testing methods, Humans, Macular Degeneration diagnosis, Macular Degeneration genetics, Macular Degeneration physiopathology, Male, Middle Aged, Retinal Pigment Epithelium physiopathology, Retrospective Studies, Stargardt Disease, Time Factors, Tomography, Optical Coherence methods, Young Adult, Macular Degeneration congenital, Retinal Pigment Epithelium pathology, Visual Acuity
- Abstract
Purpose: Asymmetry in disease progression between left and right eyes can occur in Stargardt disease (STGD1), and this needs to be considered in novel therapeutic trials with a fellow-eye paired controlled design. This study investigated the inter-eye discordance of best-corrected visual acuity (BCVA) and progression of RPE atrophy in STGD1., Methods: We performed a retrospective cohort study collecting 68 STGD1 patients (136 eyes) with ≥1 ABCA4 variants and ≥0.5-year follow-up on BCVA and fundus autofluorescence. We compared inter-eye correlations of RPE atrophy progression between early-onset (≤10 years), intermediate-onset (11-44), and late-onset (≥45) STGD1 and ABCA4 variant combinations by χ2 tests. We identified associations of discordant baseline BCVA and RPE atrophy with discordant RPE atrophy progression by odds ratios (OR). We defined discordance by differences >1.5 interquartile ranges ± first/third interquartiles., Results: Progression of RPE atrophy correlated moderately between eyes (ρ = 0.766), which decreased with later onset (P = 9.8 × 10-7) and lower pathogenicity of ABCA4 combinations (P = 0.007). Twelve patients (17.6%) had discordant inter-eye RPE atrophy progression, associated with baseline discordance of RPE atrophy (OR, 6.50 [1.35-31.34]), but not BCVA (OR, 0.33 [0.04-2.85])., Conclusions: Lower inter-eye correlations are more likely found in late-onset STGD1 and patients carrying low pathogenic ABCA4 combinations. To achieve the highest power in a therapeutic trial, early-phase studies should minimize inter-eye discordance by selecting early-onset STGD1 patients carrying severe ABCA4 variants without evidence of asymmetry at baseline.
- Published
- 2016
- Full Text
- View/download PDF
19. The expanding clinical phenotype of Bosch-Boonstra-Schaaf optic atrophy syndrome: 20 new cases and possible genotype-phenotype correlations.
- Author
-
Chen CA, Bosch DG, Cho MT, Rosenfeld JA, Shinawi M, Lewis RA, Mann J, Jayakar P, Payne K, Walsh L, Moss T, Schreiber A, Schoonveld C, Monaghan KG, Elmslie F, Douglas G, Boonstra FN, Millan F, Cremers FP, McKnight D, Richard G, Juusola J, Kendall F, Ramsey K, Anyane-Yeboa K, Malkin E, Chung WK, Niyazov D, Pascual JM, Walkiewicz M, Veluchamy V, Li C, Hisama FM, de Vries BB, and Schaaf C
- Subjects
- Adolescent, Adult, Autism Spectrum Disorder complications, Autism Spectrum Disorder physiopathology, Child, Child, Preschool, Female, Gene Deletion, Humans, Male, Mutation, Missense, Optic Atrophy complications, Optic Atrophy physiopathology, Pedigree, Autism Spectrum Disorder genetics, COUP Transcription Factor I genetics, Genetic Association Studies, Optic Atrophy genetics
- Abstract
Purpose: Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) is an autosomal-dominant disorder characterized by optic atrophy and intellectual disability caused by loss-of-function mutations in NR2F1. We report 20 new individuals with BBSOAS, exploring the spectrum of clinical phenotypes and assessing potential genotype-phenotype correlations., Methods: Clinical features of individuals with pathogenic NR2F1 variants were evaluated by review of medical records. The functional relevance of coding nonsynonymous NR2F1 variants was assessed with a luciferase assay measuring the impact on transcriptional activity. The effects of two start codon variants on protein expression were evaluated by western blot analysis., Results: We recruited 20 individuals with novel pathogenic NR2F1 variants (seven missense variants, five translation initiation variants, two frameshifting insertions/deletions, one nonframeshifting insertion/deletion, and five whole-gene deletions). All the missense variants were found to impair transcriptional activity. In addition to visual and cognitive deficits, individuals with BBSOAS manifested hypotonia (75%), seizures (40%), autism spectrum disorder (35%), oromotor dysfunction (60%), thinning of the corpus callosum (53%), and hearing defects (20%)., Conclusion: BBSOAS encompasses a broad range of clinical phenotypes. Functional studies help determine the severity of novel NR2F1 variants. Some genotype-phenotype correlations seem to exist, with missense mutations in the DNA-binding domain causing the most severe phenotypes.Genet Med 18 11, 1143-1150.
- Published
- 2016
- Full Text
- View/download PDF
20. Mutations in AGBL5, Encoding α-Tubulin Deglutamylase, Are Associated With Autosomal Recessive Retinitis Pigmentosa.
- Author
-
Astuti GD, Arno G, Hull S, Pierrache L, Venselaar H, Carss K, Raymond FL, Collin RW, Faradz SM, van den Born LI, Webster AR, and Cremers FP
- Subjects
- Adolescent, Adult, Alleles, Carboxypeptidases metabolism, Child, Child, Preschool, DNA Mutational Analysis, Exome, Female, Genes, Recessive, Homozygote, Humans, Male, Middle Aged, Pedigree, Phenotype, Retinitis Pigmentosa diagnosis, Retinitis Pigmentosa metabolism, Young Adult, Carboxypeptidases genetics, DNA genetics, Mutation, Retinitis Pigmentosa genetics, Tubulin metabolism
- Abstract
Purpose: AGBL5, encoding ATP/GTP binding protein-like 5, was previously proposed as an autosomal recessive retinitis pigmentosa (arRP) candidate gene based on the identification of missense variants in two families. In this study, we performed next-generation sequencing to reveal additional RP cases with AGBL5 variants, including protein-truncating variants., Methods: Whole-genome sequencing (WGS) or whole-exome sequencing (WES) was performed in three probands. Subsequent Sanger sequencing and segregation analysis were performed in the selected candidate genes. The medical history of individuals carrying AGBL5 variants was reviewed and additional ophthalmic examinations were performed, including fundus photography, fundus autofluorescence imaging, and optical coherence tomography., Results: AGBL5 variants were identified in three unrelated arRP families, comprising homozygous variants in family 1 (c.1775G>A:p.(Trp592*)) and family 2 (complex allele: c.[323C>G; 2659T>C]; p.[(Pro108Arg; *887Argext*1)]), and compound heterozygous variants (c.752T>G:p.(Val251Gly) and c.1504dupG:p.(Ala502Glyfs*15)) in family 3. All affected individuals displayed typical RP phenotypes., Conclusions: Our study convincingly shows that variants in AGBL5 are associated with arRP. The identification of AGBL5 and TTLL5, a previously described RP-associated gene encoding the tubulin tyrosine ligase-like family, member 5 protein, highlights the importance of poly- and deglutamylation in retinal homeostasis. Further studies are required to investigate the underlying disease mechanism associated with AGBL5 variants.
- Published
- 2016
- Full Text
- View/download PDF
21. Mutations in the polyglutamylase gene TTLL5, expressed in photoreceptor cells and spermatozoa, are associated with cone-rod degeneration and reduced male fertility.
- Author
-
Bedoni N, Haer-Wigman L, Vaclavik V, Tran VH, Farinelli P, Balzano S, Royer-Bertrand B, El-Asrag ME, Bonny O, Ikonomidis C, Litzistorf Y, Nikopoulos K, Yioti GG, Stefaniotou MI, McKibbin M, Booth AP, Ellingford JM, Black GC, Toomes C, Inglehearn CF, Hoyng CB, Bax N, Klaver CC, Thiadens AA, Murisier F, Schorderet DF, Ali M, Cremers FP, Andréasson S, Munier FL, and Rivolta C
- Subjects
- Adolescent, Adult, Aged, Animals, Cone-Rod Dystrophies genetics, DNA Mutational Analysis, Disease Models, Animal, Eye Proteins genetics, Female, Homozygote, Humans, Infertility, Male genetics, Male, Mice, Middle Aged, Organ Specificity, Pedigree, Photoreceptor Cells, Vertebrate enzymology, Rats, Sperm Motility, Spermatozoa enzymology, Testis enzymology, Carrier Proteins genetics, Cone-Rod Dystrophies enzymology, Gene Expression, Infertility, Male enzymology, Mutation
- Abstract
Hereditary retinal degenerations encompass a group of genetic diseases characterized by extreme clinical variability. Following next-generation sequencing and autozygome-based screening of patients presenting with a peculiar, recessive form of cone-dominated retinopathy, we identified five homozygous variants [p.(Asp594fs), p.(Gln117*), p.(Met712fs), p.(Ile756Phe), and p.(Glu543Lys)] in the polyglutamylase-encoding gene TTLL5, in eight patients from six families. The two male patients carrying truncating TTLL5 variants also displayed a substantial reduction in sperm motility and infertility, whereas those carrying missense changes were fertile. Defects in this polyglutamylase in humans have recently been associated with cone photoreceptor dystrophy, while mouse models carrying truncating mutations in the same gene also display reduced fertility in male animals. We examined the expression levels of TTLL5 in various human tissues and determined that this gene has multiple viable isoforms, being highly expressed in testis and retina. In addition, antibodies against TTLL5 stained the basal body of photoreceptor cells in rat and the centrosome of the spermatozoon flagellum in humans, suggesting a common mechanism of action in these two cell types. Taken together, our data indicate that mutations in TTLL5 delineate a novel, allele-specific syndrome causing defects in two as yet pathogenically unrelated functions, reproduction and vision.
- Published
- 2016
- Full Text
- View/download PDF
22. Genetic and clinical characterization of Pakistani families with Bardet-Biedl syndrome extends the genetic and phenotypic spectrum.
- Author
-
Maria M, Lamers IJ, Schmidts M, Ajmal M, Jaffar S, Ullah E, Mustafa B, Ahmad S, Nazmutdinova K, Hoskins B, van Wijk E, Koster-Kamphuis L, Khan MI, Beales PL, Cremers FP, Roepman R, Azam M, Arts HH, and Qamar R
- Subjects
- Adolescent, Adult, Cytoskeletal Proteins, DNA Mutational Analysis methods, Female, Genetic Predisposition to Disease, High-Throughput Nucleotide Sequencing methods, Humans, Male, Middle Aged, Pakistan, Pedigree, Phosphate-Binding Proteins, Exome Sequencing methods, Young Adult, ADP-Ribosylation Factors genetics, Bardet-Biedl Syndrome genetics, Genetic Association Studies methods, Microtubule Proteins genetics, Neoplasm Proteins genetics, Proteins genetics
- Abstract
Bardet-Biedl syndrome (BBS) is an autosomal recessive disorder that is both genetically and clinically heterogeneous. To date 19 genes have been associated with BBS, which encode proteins active at the primary cilium, an antenna-like organelle that acts as the cell's signaling hub. In the current study, a combination of mutation screening, targeted sequencing of ciliopathy genes associated with BBS, and whole-exome sequencing was used for the genetic characterization of five families including four with classic BBS symptoms and one BBS-like syndrome. This resulted in the identification of novel mutations in BBS genes ARL6 and BBS5, and recurrent mutations in BBS9 and CEP164. In the case of CEP164, this is the first report of two siblings with a BBS-like syndrome with mutations in this gene. Mutations in this gene were previously associated with nephronophthisis 15, thus the current results expand the CEP164-associated phenotypic spectrum. The clinical and genetic spectrum of BBS and BBS-like phenotypes is not fully defined in Pakistan. Therefore, genetic studies are needed to gain insights into genotype-phenotype correlations, which will in turn improve the clinician's ability to make an early and accurate diagnosis, and facilitate genetic counseling, leading to directly benefiting families with affected individuals.
- Published
- 2016
- Full Text
- View/download PDF
23. Reevaluation of the Retinal Dystrophy Due to Recessive Alleles of RGR With the Discovery of a Cis-Acting Mutation in CDHR1.
- Author
-
Arno G, Hull S, Carss K, Dev-Borman A, Chakarova C, Bujakowska K, van den Born LI, Robson AG, Holder GE, Michaelides M, Cremers FP, Pierce E, Raymond FL, Moore AT, and Webster AR
- Subjects
- Alleles, Cadherin Related Proteins, Cadherins metabolism, DNA Mutational Analysis, Electroretinography, Female, Fluorescein Angiography, Fundus Oculi, Genes, Recessive, Homozygote, Humans, Male, Nerve Tissue Proteins metabolism, Pedigree, Phenotype, Retina pathology, Retinal Dystrophies diagnosis, Retinal Dystrophies metabolism, Tomography, Optical Coherence, Cadherins genetics, DNA genetics, Mutation, Nerve Tissue Proteins genetics, Retina metabolism, Retinal Dystrophies genetics
- Abstract
Purpose: Mutation of RGR, encoding retinal G-protein coupled receptor was originally reported in association with retinal dystrophy in 1999. A single convincing recessive variant segregated perfectly in one family of five affected and two unaffected siblings. At least one further individual, homozygous for the same variant has since been reported. The aim of this report was to reevaluate the findings in consideration of data from a whole genome sequencing (WGS) study of a large cohort of retinal dystrophy families., Methods: Whole genome sequencing was performed on 599 unrelated probands with inherited retinal disease. Detailed phenotyping was performed, including clinical evaluation, electroretinography, fundus photography, fundus autofluorescence imaging (FAF) and spectral-domain optical coherence tomography (OCT)., Results: Overall we confirmed that affected individuals from six unrelated families were homozygous for both the reported RGR p.Ser66Arg variant and a nearby frameshifting deletion in CDHR1 (p.Ile841Serfs119*). All had generalized rod and cone dysfunction with severe macular involvement. An additional proband was heterozygous for the same CDHR1/RGR haplotype but also carried a second null CDHR1 mutation on a different haplotype. A comparison of the clinical presentation of the probands reported here with other CDHR1-related retinopathy patients shows the phenotypes to be similar in presentation, severity, and rod/cone involvement., Conclusions: These data suggest that the recessive retinal disorder previously reported to be due to homozygous mutation in RGR is, at least in part, due to variants in CDHR1 and that the true consequences of RGR knock-out on human retinal structure and function are yet to be determined.
- Published
- 2016
- Full Text
- View/download PDF
24. Comprehensive genotyping reveals RPE65 as the most frequently mutated gene in Leber congenital amaurosis in Denmark.
- Author
-
Astuti GD, Bertelsen M, Preising MN, Ajmal M, Lorenz B, Faradz SM, Qamar R, Collin RW, Rosenberg T, and Cremers FP
- Subjects
- Adult, Child, Denmark, Female, Heterozygote, Humans, Infant, Leber Congenital Amaurosis pathology, Male, Mutation, Missense, Pedigree, RNA Splicing, Leber Congenital Amaurosis genetics, Mutation Rate, cis-trans-Isomerases genetics
- Abstract
Leber congenital amaurosis (LCA) represents the most severe form of inherited retinal dystrophies with an onset during the first year of life. Currently, 21 genes are known to be associated with LCA and recurrent mutations have been observed in AIPL1, CEP290, CRB1 and GUCY2D. In addition, sequence analysis of LRAT and RPE65 may be important in view of treatments that are emerging for patients carrying variants in these genes. Screening of the aforementioned variants and genes was performed in 64 Danish LCA probands. Upon the identification of heterozygous variants, Sanger sequencing was performed of the relevant genes to identify the second allele. In combination with prior arrayed primer extension analysis, this led to the identification of two variants in 42 of 86 cases (49%). Remarkably, biallelic RPE65 variants were identified in 16% of the cases, and one novel variant, p.(D110G), was found in seven RPE65 alleles. We also collected all previously published RPE65 variants, identified in 914 alleles of 539 patients with LCA or early-onset retinitis pigmentosa, and deposited them in the RPE65 Leiden Open Variation Database (LOVD). The in silico pathogenicity assessment of the missense and noncanonical splice site variants, as well as an analysis of their frequency in ~60 000 control individuals, rendered 864 of the alleles to affect function or probably affect function. This comprehensive database can now be used to select patients eligible for gene augmentation or retinoid supplementation therapies.
- Published
- 2016
- Full Text
- View/download PDF
25. Photoreceptor Progenitor mRNA Analysis Reveals Exon Skipping Resulting from the ABCA4 c.5461-10T→C Mutation in Stargardt Disease.
- Author
-
Sangermano R, Bax NM, Bauwens M, van den Born LI, De Baere E, Garanto A, Collin RW, Goercharn-Ramlal AS, den Engelsman-van Dijk AH, Rohrschneider K, Hoyng CB, Cremers FP, and Albert S
- Subjects
- Adult, DNA Mutational Analysis, Electroretinography, Female, Fibroblasts metabolism, Fluorescein Angiography, HEK293 Cells, Haplotypes, Humans, Induced Pluripotent Stem Cells metabolism, Macular Degeneration diagnosis, Macular Degeneration genetics, Macular Degeneration physiopathology, Male, Middle Aged, Polymorphism, Single Nucleotide, RNA Splice Sites genetics, Reverse Transcriptase Polymerase Chain Reaction, Stargardt Disease, Transfection, Visual Acuity physiology, Visual Fields physiology, Young Adult, ATP-Binding Cassette Transporters genetics, Alternative Splicing, Exons genetics, Macular Degeneration congenital, Photoreceptor Cells, Vertebrate physiology, RNA, Messenger genetics, Stem Cells physiology
- Abstract
Purpose: To elucidate the functional effect of the ABCA4 variant c.5461-10T→C, one of the most frequent variants associated with Stargardt disease (STGD1)., Design: Case series., Participants: Seventeen persons with STGD1 carrying ABCA4 variants and 1 control participant., Methods: Haplotype analysis of 4 homozygotes and 11 heterozygotes for c.5461-10T→C and sequence analysis of the ABCA4 gene for a homozygous proband. Fibroblasts were reprogrammed from 3 persons with STGD1 into induced pluripotent stem cells, which were differentiated into photoreceptor progenitor cells (PPCs). The effect of the c.5461-10T→C variant on RNA splicing by reverse-transcription polymerase chain reaction was analyzed using PPC mRNA. In vitro assays were performed with minigene constructs containing ABCA4 exon 39. We analyzed the natural history and ophthalmologic characteristics of 4 persons homozygous for c.5461-10T→C., Main Outcome Measures: Haplotype and rare variant data for ABCA4, RNA splice defects, age at diagnosis, visual acuity, fundus appearance, visual field, electroretinography (ERG) results, fluorescein angiography results, and fundus autofluorescence findings., Results: The frequent ABCA4 variant c.5461-10T→C has a subtle effect on splicing based on prediction programs. A founder haplotype containing c.5461-10T→C was found to span approximately 96 kb of ABCA4 and did not contain other rare sequence variants. Patient-derived PPCs showed skipping of exon 39 or exons 39 and 40 in the mRNA. HEK293T cell transduction with minigenes carrying exon 39 showed that the splice defects were the result of the c.5461-10T→C variant. All 4 subjects carrying the c.5461-10T→C variant in a homozygous state showed a young age of STGD1 onset, with low visual acuity at presentation and abnormal cone ERG results. All 4 demonstrated severe cone-rod dystrophy before 20 years of age and were legally blind by 25 years of age., Conclusions: The ABCA4 variant c.5461-10T→C is located on a founder haplotype lacking other disease-causing rare sequence variants. In vitro studies revealed that it leads to mRNA exon skipping and ABCA4 protein truncation. Given the severe phenotype in persons homozygous for this variant, we conclude that this variant results in the absence of ABCA4 activity., (Copyright © 2016 American Academy of Ophthalmology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Biallelic Mutations in CRB1 Underlie Autosomal Recessive Familial Foveal Retinoschisis.
- Author
-
Vincent A, Ng J, Gerth-Kahlert C, Tavares E, Maynes JT, Wright T, Tiwari A, Tumber A, Li S, Hanson JV, Bahr A, MacDonald H, Bähr L, Westall C, Berger W, Cremers FP, den Hollander AI, and Héon E
- Subjects
- Adolescent, Adult, DNA Mutational Analysis, Eye Proteins metabolism, Female, Fluorescein Angiography, Fovea Centralis metabolism, Fundus Oculi, Genetic Testing, Genotype, Humans, Male, Membrane Proteins metabolism, Nerve Tissue Proteins metabolism, Pedigree, Phenotype, Retinoschisis diagnosis, Retinoschisis metabolism, Tomography, Optical Coherence, Visual Acuity, Young Adult, DNA genetics, Eye Proteins genetics, Fovea Centralis pathology, Membrane Proteins genetics, Mutation, Nerve Tissue Proteins genetics, Retinoschisis genetics
- Abstract
Purpose: To identify the genetic cause of autosomal recessive familial foveal retinoschisis (FFR)., Methods: A female sibship with FFR was identified (Family-A; 17 and 16 years, respectively); panel based genetic sequencing (132 genes) and comparative genome hybridization (142 genes) were performed. Whole-exome sequencing (WES) was performed on both siblings using the Illumina-HiSeq-2500 platform. A sporadic male (Family-B; 35 years) with FFR underwent WES using Illumina NextSeq500. All three affected subjects underwent detailed ophthalmologic evaluation including fundus photography, autofluorescence imaging, spectral-domain optical coherence tomography (SD-OCT), and full-field electroretinogram (ERG)., Results: Panel-based genetic testing identified two presumed disease causing variants in CRB1 (p.Gly123Cys and p.Cys948Tyr) in Family-A sibship; no deletion or duplication was detected. WES analysis in the sibship identified nine genes with two or more shared nonsynonymous rare coding sequence variants; CRB1 remained a strong candidate gene, and CRB1 variants segregated with the disease. WES in Family-B identified two presumed disease causing variants in CRB1 (p.Ile167_Gly169del and p.Arg764Cys) that segregated with the disease phenotype. Distance visual acuity was 20/40 or better in all three affected except for the left eye of the older subject (Family-B), which showed macular atrophy. Fundus evaluation showed spoke-wheel appearance at the macula in five eyes. The SD-OCT showed macular schitic changes in inner and outer nuclear layers in all cases. The ERG responses were normal in all subjects., Conclusions: This is the first report to implicate CRB1 as the underlying cause of FFR. This phenotype forms the mildest end of the spectrum of CRB1-related diseases.
- Published
- 2016
- Full Text
- View/download PDF
27. Visual Prognosis in USH2A-Associated Retinitis Pigmentosa Is Worse for Patients with Usher Syndrome Type IIa Than for Those with Nonsyndromic Retinitis Pigmentosa.
- Author
-
Pierrache LH, Hartel BP, van Wijk E, Meester-Smoor MA, Cremers FP, de Baere E, de Zaeytijd J, van Schooneveld MJ, Cremers CW, Dagnelie G, Hoyng CB, Bergen AA, Leroy BP, Pennings RJ, van den Born LI, and Klaver CC
- Subjects
- Adolescent, Adult, Aged, Blindness physiopathology, DNA Mutational Analysis, Female, Follow-Up Studies, Genetic Association Studies, Humans, Male, Middle Aged, Mutation, Prognosis, Retinitis Pigmentosa physiopathology, Usher Syndromes physiopathology, Vision, Low physiopathology, Visual Fields physiology, Extracellular Matrix Proteins genetics, Retinitis Pigmentosa genetics, Usher Syndromes genetics, Visual Acuity physiology
- Abstract
Purpose: USH2A mutations are an important cause of retinitis pigmentosa (RP) with or without congenital sensorineural hearing impairment. We studied genotype-phenotype correlations and compared visual prognosis in Usher syndrome type IIa and nonsyndromic RP., Design: Clinic-based, longitudinal, multicenter study., Participants: Consecutive patients with Usher syndrome type IIa (n = 152) and nonsyndromic RP (n = 73) resulting from USH2A mutations from ophthalmogenetic clinics in the Netherlands and Belgium., Methods: Data on clinical characteristics, visual acuity, visual field measurements, retinal imaging, and electrophysiologic features were extracted from medical charts over a mean follow-up of 9 years. Cumulative lifetime risks of low vision and blindness were estimated using Kaplan-Meier survival analysis., Main Outcome Measures: Low vision and blindness., Results: Participant groups had similar distributions of gender (48% vs. 45% males in Usher syndrome type IIa vs. nonsydromic RP; P = 0.8), ethnicity (97% vs. 99% European; P = 0.3), and median follow-up time (6.5 years vs. 3 years; P = 0.3). Usher syndrome type IIa patients demonstrated symptoms at a younger age (median age, 15 years vs. 25 years; P < 0.001), were diagnosed earlier (median age, 26 years vs. 36.5 years; P < 0.001), and became visually impaired 13 years earlier (median age, 41 years vs. 54 years; P < 0.001) based on VF and 18 years earlier based on VA (median age, 54 years vs. 72 years; P < 0.001) than nonsyndromic RP patients. The presence of 2 truncating mutations in USH2A was associated mostly with the syndromic phenotype, whereas other combinations were present in both groups. We found novel variants in Usher syndrome type IIa (25%) and nonsyndromic RP (19%): 29 missense mutations, 10 indels, 14 nonsense mutations, 9 frameshift mutations, and 5 splice-site mutations., Conclusions: Most patients with USH2A-associated RP have severe visual impairment by age 50. However, those with Usher syndrome type IIa have an earlier decline of visual function and a higher cumulative risk of visual impairment than those without nonsyndromic RP. Complete loss of function of the USH2A protein predisposes to Usher syndrome type IIa, but remnant protein function can lead to RP with or without hearing loss., (Copyright © 2016 American Academy of Ophthalmology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
28. Novel genetic causes for cerebral visual impairment.
- Author
-
Bosch DG, Boonstra FN, de Leeuw N, Pfundt R, Nillesen WM, de Ligt J, Gilissen C, Jhangiani S, Lupski JR, Cremers FP, and de Vries BB
- Subjects
- Adolescent, Blindness, Cortical diagnosis, Child, Child, Preschool, Female, Humans, Male, Young Adult, Blindness, Cortical genetics, Genetic Loci, Polymorphism, Single Nucleotide
- Abstract
Cerebral visual impairment (CVI) is a major cause of low vision in children due to impairment in projection and/or interpretation of the visual input in the brain. Although acquired causes for CVI are well known, genetic causes underlying CVI are largely unidentified. DNAs of 25 patients with CVI and intellectual disability, but without acquired (eg, perinatal) damage, were investigated by whole-exome sequencing. The data were analyzed for de novo, autosomal-recessive, and X-linked variants, and subsequently classified into known, candidate, or unlikely to be associated with CVI. This classification was based on the Online Mendelian Inheritance in Man database, literature reports, variant characteristics, and functional relevance of the gene. After classification, variants in four genes known to be associated with CVI (AHDC1, NGLY1, NR2F1, PGAP1) in 5 patients (20%) were identified, establishing a conclusive genetic diagnosis for CVI. In addition, in 11 patients (44%) with CVI, variants in one or more candidate genes were identified (ACP6, AMOT, ARHGEF10L, ATP6V1A, DCAF6, DLG4, GABRB2, GRIN1, GRIN2B, KCNQ3, KCTD19, RERE, SLC1A1, SLC25A16, SLC35A2, SOX5, UFSP2, UHMK1, ZFP30). Our findings show that diverse genetic causes underlie CVI, some of which will provide insight into the biology underlying this disease process.
- Published
- 2016
- Full Text
- View/download PDF
29. Mutations in CTNNA1 cause butterfly-shaped pigment dystrophy and perturbed retinal pigment epithelium integrity.
- Author
-
Saksens NT, Krebs MP, Schoenmaker-Koller FE, Hicks W, Yu M, Shi L, Rowe L, Collin GB, Charette JR, Letteboer SJ, Neveling K, van Moorsel TW, Abu-Ltaif S, De Baere E, Walraedt S, Banfi S, Simonelli F, Cremers FP, Boon CJ, Roepman R, Leroy BP, Peachey NS, Hoyng CB, Nishina PM, and den Hollander AI
- Subjects
- Animals, Female, Humans, Light, Male, Mice, Mice, Mutant Strains, Pedigree, Retinal Dystrophies pathology, Mutation, Missense, Retinal Dystrophies genetics, Retinal Pigment Epithelium pathology, alpha Catenin genetics
- Abstract
Butterfly-shaped pigment dystrophy is an eye disease characterized by lesions in the macula that can resemble the wings of a butterfly. Here we report the identification of heterozygous missense mutations in the CTNNA1 gene (encoding α-catenin 1) in three families with butterfly-shaped pigment dystrophy. In addition, we identified a Ctnna1 missense mutation in a chemically induced mouse mutant, tvrm5. Parallel clinical phenotypes were observed in the retinal pigment epithelium (RPE) of individuals with butterfly-shaped pigment dystrophy and in tvrm5 mice, including pigmentary abnormalities, focal thickening and elevated lesions, and decreased light-activated responses. Morphological studies in tvrm5 mice demonstrated increased cell shedding and the presence of large multinucleated RPE cells, suggesting defects in intercellular adhesion and cytokinesis. This study identifies CTNNA1 gene variants as a cause of macular dystrophy, indicates that CTNNA1 is involved in maintaining RPE integrity and suggests that other components that participate in intercellular adhesion may be implicated in macular disease.
- Published
- 2016
- Full Text
- View/download PDF
30. Cerebral visual impairment and intellectual disability caused by PGAP1 variants.
- Author
-
Bosch DG, Boonstra FN, Kinoshita T, Jhangiani S, de Ligt J, Cremers FP, Lupski JR, Murakami Y, and de Vries BB
- Subjects
- Animals, CHO Cells, Cell Line, Tumor, Child, Cricetinae, Cricetulus, Humans, Intellectual Disability diagnosis, Male, Membrane Proteins metabolism, Phosphoinositide Phospholipase C metabolism, Phosphoric Monoester Hydrolases metabolism, Syndrome, Vision Disorders diagnosis, Visual Perception, Intellectual Disability genetics, Membrane Proteins genetics, Mutation, Phosphoric Monoester Hydrolases genetics, Vision Disorders genetics
- Abstract
Homozygous variants in PGAP1 (post-GPI attachment to proteins 1) have recently been identified in two families with developmental delay, seizures and/or spasticity. PGAP1 is a member of the glycosylphosphatidylinositol anchor biosynthesis and remodeling pathway and defects in this pathway are a subclass of congenital disorders of glycosylation. Here we performed whole-exome sequencing in an individual with cerebral visual impairment (CVI), intellectual disability (ID), and factor XII deficiency and revealed compound heterozygous variants in PGAP1, c.274_276del (p.(Pro92del)) and c.921_925del (p.(Lys308Asnfs*25)). Subsequently, PGAP1-deficient Chinese hamster ovary (CHO)-cell lines were transfected with either mutant or wild-type constructs and their sensitivity to phosphatidylinositol-specific phospholipase C (PI-PLC) treatment was measured. The mutant constructs could not rescue the PGAP1-deficient CHO cell lines resistance to PI-PLC treatment. In addition, lymphoblastoid cell lines (LCLs) of the affected individual showed no sensitivity to PI-PLC treatment, whereas the LCLs of the heterozygous carrier parents were partially resistant. In conclusion, we report novel PGAP1 variants in a boy with CVI and ID and a proven functional loss of PGAP1 and show, to our knowledge, for the first time this genetic association with CVI.
- Published
- 2015
- Full Text
- View/download PDF
31. A Nonsense Mutation in FAM161A Is a Recurrent Founder Allele in Dutch and Belgian Individuals With Autosomal Recessive Retinitis Pigmentosa.
- Author
-
Van Schil K, Klevering BJ, Leroy BP, Pott JW, Bandah-Rozenfeld D, Zonneveld-Vrieling MN, Sharon D, den Hollander AI, Cremers FP, De Baere E, Collin RW, and van den Born LI
- Subjects
- Adult, Alleles, Belgium epidemiology, Codon, Nonsense, DNA Mutational Analysis, Eye Proteins metabolism, Female, Genes, Recessive, Haplotypes, Humans, Male, Middle Aged, Netherlands epidemiology, Pedigree, Retinitis Pigmentosa epidemiology, Retinitis Pigmentosa metabolism, DNA genetics, Eye Proteins genetics, Mutation, Retinitis Pigmentosa genetics
- Abstract
Purpose: To identify mutations in FAM161A underlying autosomal recessive retinitis pigmentosa (arRP) in the Dutch and Belgian populations and to investigate whether common FAM161A-associated phenotypic features could be identified., Methods: Homozygosity mapping, amplification-refractory mutation system (ARMS) analysis, and Sanger sequencing were performed to identify mutations in FAM161A. Microsatellite and SNP markers were genotyped for haplotype analysis. Patients with biallelic mutations underwent detailed ophthalmologic examinations, including measuring best-corrected visual acuity, extensive fundus photography with reflectance and autofluorescence imaging, and optical coherence tomography., Results: Homozygosity mapping in 230 Dutch individuals with suspected arRP yielded five individuals with a homozygous region harboring FAM161A. Sanger sequencing revealed a homozygous nonsense mutation (c.1309A>T; p.[Arg437*]) in one individual. Subsequent ARMS analysis and Sanger sequencing in Dutch and Belgian arRP patients resulted in the identification of seven additional individuals carrying the p.(Arg437*) mutation, either homozygously or compound heterozygously with another mutation. Haplotype analysis identified a shared haplotype block of 409 kb surrounding the p.(Arg437*) mutation in all patients, suggesting a founder effect. Although the age of onset was variable among patients, all eight developed pronounced outer retinal loss with severe visual field defects and a bull's eye-like maculopathy, followed by loss of central vision within 2 decades after the initial diagnosis in five subjects., Conclusions: A founder mutation in FAM161A p.(Arg437*) underlies approximately 2% of arRP cases in the Dutch and Belgian populations. The age of onset of the retinal dystrophy appears variable, but progression can be steep, with almost complete loss of central vision later in life.
- Published
- 2015
- Full Text
- View/download PDF
32. Non-syndromic retinitis pigmentosa due to mutations in the mucopolysaccharidosis type IIIC gene, heparan-alpha-glucosaminide N-acetyltransferase (HGSNAT).
- Author
-
Haer-Wigman L, Newman H, Leibu R, Bax NM, Baris HN, Rizel L, Banin E, Massarweh A, Roosing S, Lefeber DJ, Zonneveld-Vrieling MN, Isakov O, Shomron N, Sharon D, Den Hollander AI, Hoyng CB, Cremers FP, and Ben-Yosef T
- Subjects
- Adult, Aged, Animals, Asymptomatic Diseases, Base Sequence, Exons, Female, Humans, Male, Mice, Mice, Inbred C57BL, Middle Aged, Molecular Sequence Data, Mucopolysaccharidosis III genetics, Pedigree, Retina enzymology, Retinitis Pigmentosa genetics, Acetyltransferases genetics, Mucopolysaccharidosis III enzymology, Point Mutation, Retinitis Pigmentosa enzymology
- Abstract
Retinitis pigmentosa (RP), the most common form of inherited retinal degeneration, is clinically and genetically heterogeneous and can appear as syndromic or non-syndromic. Mucopolysaccharidosis type IIIC (MPS IIIC) is a lethal disorder, caused by mutations in the heparan-alpha-glucosaminide N-acetyltransferase (HGSNAT) gene and characterized by progressive neurological deterioration, with retinal degeneration as a prominent feature. We identified HGSNAT mutations in six patients with non-syndromic RP. Whole exome sequencing (WES) in an Ashkenazi Jewish Israeli RP patient revealed a novel homozygous HGSNAT variant, c.370A>T, which leads to partial skipping of exon 3. Screening of 66 Ashkenazi RP index cases revealed an additional family with two siblings homozygous for c.370A>T. WES in three Dutch siblings with RP revealed a complex HGSNAT variant, c.[398G>C; 1843G>A] on one allele, and c.1843G>A on the other allele. HGSNAT activity levels in blood leukocytes of patients were reduced compared with healthy controls, but usually higher than those in MPS IIIC patients. All patients were diagnosed with non-syndromic RP and did not exhibit neurological deterioration, or any phenotypic features consistent with MPS IIIC. Furthermore, four of the patients were over 60 years old, exceeding by far the life expectancy of MPS IIIC patients. HGSNAT is highly expressed in the mouse retina, and we hypothesize that the retina requires higher HGSNAT activity to maintain proper function, compared with other tissues associated with MPS IIIC, such as the brain. This report broadens the spectrum of phenotypes associated with HGSNAT mutations and highlights the critical function of HGSNAT in the human retina., (© The Author 2015. Published by Oxford University Press.)
- Published
- 2015
- Full Text
- View/download PDF
33. Mutations in the unfolded protein response regulator ATF6 cause the cone dysfunction disorder achromatopsia.
- Author
-
Kohl S, Zobor D, Chiang WC, Weisschuh N, Staller J, Gonzalez Menendez I, Chang S, Beck SC, Garcia Garrido M, Sothilingam V, Seeliger MW, Stanzial F, Benedicenti F, Inzana F, Héon E, Vincent A, Beis J, Strom TM, Rudolph G, Roosing S, Hollander AI, Cremers FP, Lopez I, Ren H, Moore AT, Webster AR, Michaelides M, Koenekoop RK, Zrenner E, Kaufman RJ, Tsang SH, Wissinger B, and Lin JH
- Subjects
- Adolescent, Adult, Aged, 80 and over, Animals, Child, Female, Genetic Association Studies, HEK293 Cells, Humans, Male, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Mutation, Missense, Pedigree, Retinal Cone Photoreceptor Cells pathology, Transcription, Genetic, Unfolded Protein Response, Young Adult, Activating Transcription Factor 6 genetics, Color Vision Defects genetics
- Abstract
Achromatopsia (ACHM) is an autosomal recessive disorder characterized by color blindness, photophobia, nystagmus and severely reduced visual acuity. Using homozygosity mapping and whole-exome and candidate gene sequencing, we identified ten families carrying six homozygous and two compound-heterozygous mutations in the ATF6 gene (encoding activating transcription factor 6A), a key regulator of the unfolded protein response (UPR) and cellular endoplasmic reticulum (ER) homeostasis. Patients had evidence of foveal hypoplasia and disruption of the cone photoreceptor layer. The ACHM-associated ATF6 mutations attenuate ATF6 transcriptional activity in response to ER stress. Atf6(-/-) mice have normal retinal morphology and function at a young age but develop rod and cone dysfunction with increasing age. This new ACHM-related gene suggests a crucial and unexpected role for ATF6A in human foveal development and cone function and adds to the list of genes that, despite ubiquitous expression, when mutated can result in an isolated retinal photoreceptor phenotype.
- Published
- 2015
- Full Text
- View/download PDF
34. Homozygosity mapping and targeted sanger sequencing reveal genetic defects underlying inherited retinal disease in families from pakistan.
- Author
-
Maria M, Ajmal M, Azam M, Waheed NK, Siddiqui SN, Mustafa B, Ayub H, Ali L, Ahmad S, Micheal S, Hussain A, Shah ST, Ali SH, Ahmed W, Khan YM, den Hollander AI, Haer-Wigman L, Collin RW, Khan MI, Qamar R, and Cremers FP
- Subjects
- Consanguinity, DNA Mutational Analysis, Female, Genotype, Homozygote, Humans, Leber Congenital Amaurosis epidemiology, Leber Congenital Amaurosis genetics, Male, Pakistan epidemiology, Pedigree, Polymorphism, Single Nucleotide, Retina metabolism, Retina pathology, Retinal Diseases epidemiology, Retinitis Pigmentosa epidemiology, Retinitis Pigmentosa genetics, Mutation, Retinal Diseases genetics
- Abstract
Background: Homozygosity mapping has facilitated the identification of the genetic causes underlying inherited diseases, particularly in consanguineous families with multiple affected individuals. This knowledge has also resulted in a mutation dataset that can be used in a cost and time effective manner to screen frequent population-specific genetic variations associated with diseases such as inherited retinal disease (IRD)., Methods: We genetically screened 13 families from a cohort of 81 Pakistani IRD families diagnosed with Leber congenital amaurosis (LCA), retinitis pigmentosa (RP), congenital stationary night blindness (CSNB), or cone dystrophy (CD). We employed genome-wide single nucleotide polymorphism (SNP) array analysis to identify homozygous regions shared by affected individuals and performed Sanger sequencing of IRD-associated genes located in the sizeable homozygous regions. In addition, based on population specific mutation data we performed targeted Sanger sequencing (TSS) of frequent variants in AIPL1, CEP290, CRB1, GUCY2D, LCA5, RPGRIP1 and TULP1, in probands from 28 LCA families., Results: Homozygosity mapping and Sanger sequencing of IRD-associated genes revealed the underlying mutations in 10 families. TSS revealed causative variants in three families. In these 13 families four novel mutations were identified in CNGA1, CNGB1, GUCY2D, and RPGRIP1., Conclusions: Homozygosity mapping and TSS revealed the underlying genetic cause in 13 IRD families, which is useful for genetic counseling as well as therapeutic interventions that are likely to become available in the near future.
- Published
- 2015
- Full Text
- View/download PDF
35. Early-onset stargardt disease: phenotypic and genotypic characteristics.
- Author
-
Lambertus S, van Huet RA, Bax NM, Hoefsloot LH, Cremers FP, Boon CJ, Klevering BJ, and Hoyng CB
- Subjects
- Adolescent, Adult, Age of Onset, Child, Child, Preschool, Electroretinography, Female, Fluorescein Angiography, Humans, Infant, Macular Degeneration diagnosis, Macular Degeneration epidemiology, Macular Degeneration genetics, Male, Middle Aged, Ophthalmoscopy, Retrospective Studies, Stargardt Disease, Tomography, Optical Coherence, Visual Acuity physiology, Young Adult, ATP-Binding Cassette Transporters genetics, DNA Mutational Analysis, Genotype, Macular Degeneration congenital, Phenotype
- Abstract
Objective: To describe the phenotype and genotype of patients with early-onset Stargardt disease., Design: Retrospective cohort study., Participants: Fifty-one Stargardt patients with age at onset ≤10 years., Methods: We reviewed patient medical records for age at onset, medical history, initial symptoms, best-corrected visual acuity (BCVA), ophthalmoscopy, fundus photography, fundus autofluorescence (FAF), fluorescein angiography (FA), spectral-domain optical coherence tomography (SD-OCT), and full-field electroretinography (ffERG). The ABCA4 gene was screened for mutations., Main Outcome Measures: Age at onset, BCVA, fundus appearance, FAF, FA, SD-OCT, ffERG, and presence of ABCA4 mutations., Results: The mean age at onset was 7.2 years (range, 1-10). The median times to develop BCVA of 20/32, 20/80, 20/200, and 20/500 were 3, 5, 12, and 23 years, respectively. Initial ophthalmoscopy in 41 patients revealed either no abnormalities or foveal retinal pigment epithelium (RPE) changes in 10 and 9 patients, respectively; the other 22 patients had foveal atrophy, atrophic RPE lesions, and/or irregular yellow-white fundus flecks. On FA, there was a "dark choroid" in 21 out of 29 patients. In 14 out of 50 patients, foveal atrophy occurred before flecks developed. On FAF, there was centrifugal expansion of disseminated atrophic spots, which progressed to the eventual profound chorioretinal atrophy. Spectral-domain OCT revealed early photoreceptor damage followed by atrophy of the outer retina, RPE, and choroid. On ffERG in 26 patients, 15 had normal amplitudes, and 11 had reduced photopic and/or scotopic amplitudes at their first visit. We found no correlation between ffERG abnormalities and the rate of vision loss. Thirteen out of 25 patients had progressive ffERG abnormalities. Finally, genetic screening of 44 patients revealed ≥2 ABCA4 mutations in 37 patients and single heterozygous mutations in 7., Conclusions: In early-onset Stargardt, initial ophthalmoscopy can reveal no abnormalities or minor retinal abnormalities. Yellow-white flecks can be preceded by foveal atrophy and may be visible only on FAF. Although ffERG is insufficient for predicting the rate of vision loss, abnormalities can develop. Over time, visual acuity declines rapidly in parallel with progressive retinal degeneration, resulting in profound chorioretinal atrophy. Thus, early-onset Stargardt lies at the severe end of the spectrum of ABCA4-associated retinal phenotypes., (Copyright © 2015 American Academy of Ophthalmology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
36. Cerebral visual impairment, autism, and pancreatitis associated with a 9 Mbp deletion on 10p12.
- Author
-
Bosch DG, Boonstra FN, Pfundt R, Cremers FP, and de Vries BB
- Subjects
- Adolescent, Adult, Child, Female, Genotype, Humans, Infant, Infant, Newborn, Male, Autistic Disorder complications, Base Pairing genetics, Chromosomes, Human, Pair 10 genetics, Pancreatitis complications, Sequence Deletion genetics, Vision Disorders complications
- Published
- 2015
- Full Text
- View/download PDF
37. Mutations in IFT172 cause isolated retinal degeneration and Bardet-Biedl syndrome.
- Author
-
Bujakowska KM, Zhang Q, Siemiatkowska AM, Liu Q, Place E, Falk MJ, Consugar M, Lancelot ME, Antonio A, Lonjou C, Carpentier W, Mohand-Saïd S, den Hollander AI, Cremers FP, Leroy BP, Gai X, Sahel JA, van den Born LI, Collin RW, Zeitz C, Audo I, and Pierce EA
- Subjects
- Adaptor Proteins, Signal Transducing, Adolescent, Adult, Animals, Cells, Cultured, Cytoskeletal Proteins, Exome, Female, High-Throughput Nucleotide Sequencing, Humans, Male, Mutation, Pedigree, Rats, Retina metabolism, Sequence Analysis, DNA, Young Adult, Zebrafish, Bardet-Biedl Syndrome genetics, Bardet-Biedl Syndrome pathology, Carrier Proteins genetics, Retina pathology, Retinitis Pigmentosa genetics, Retinitis Pigmentosa pathology
- Abstract
Primary cilia are sensory organelles present on most mammalian cells. The assembly and maintenance of primary cilia are facilitated by intraflagellar transport (IFT), a bidirectional protein trafficking along the cilium. Mutations in genes coding for IFT components have been associated with a group of diseases called ciliopathies. These genetic disorders can affect a variety of organs including the retina. Using whole exome sequencing in three families, we identified mutations in Intraflagellar Transport 172 Homolog [IFT172 (Chlamydomonas)] that underlie an isolated retinal degeneration and Bardet-Biedl syndrome. Extensive functional analyses of the identified mutations in cell culture, rat retina and in zebrafish demonstrated their hypomorphic or null nature. It has recently been reported that mutations in IFT172 cause a severe ciliopathy syndrome involving skeletal, renal, hepatic and retinal abnormalities (Jeune and Mainzer-Saldino syndromes). Here, we report for the first time that mutations in this gene can also lead to an isolated form of retinal degeneration. The functional data for the mutations can partially explain milder phenotypes; however, the involvement of modifying alleles in the IFT172-associated phenotypes cannot be excluded. These findings expand the spectrum of disease associated with mutations in IFT172 and suggest that mutations in genes originally reported to be associated with syndromic ciliopathies should also be considered in subjects with non-syndromic retinal dystrophy., (© The Author 2014. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
38. Heterozygous deep-intronic variants and deletions in ABCA4 in persons with retinal dystrophies and one exonic ABCA4 variant.
- Author
-
Bax NM, Sangermano R, Roosing S, Thiadens AA, Hoefsloot LH, van den Born LI, Phan M, Klevering BJ, Westeneng-van Haaften C, Braun TA, Zonneveld-Vrieling MN, de Wijs I, Mutlu M, Stone EM, den Hollander AI, Klaver CC, Hoyng CB, and Cremers FP
- Subjects
- Exons, Female, Genetic Heterogeneity, Genetic Predisposition to Disease, High-Throughput Nucleotide Sequencing, Humans, Introns, Macular Degeneration genetics, Male, Pedigree, Sequence Analysis, DNA, Sequence Deletion, Stargardt Disease, ATP-Binding Cassette Transporters genetics, Genetic Association Studies methods, Macular Degeneration congenital, Retinitis Pigmentosa genetics
- Abstract
Variants in ABCA4 are responsible for autosomal-recessive Stargardt disease and cone-rod dystrophy. Sequence analysis of ABCA4 exons previously revealed one causative variant in each of 45 probands. To identify the "missing" variants in these cases, we performed multiplex ligation-dependent probe amplification-based deletion scanning of ABCA4. In addition, we sequenced the promoter region, fragments containing five deep-intronic splice variants, and 15 deep-intronic regions containing weak splice sites. Heterozygous deletions spanning ABCA4 exon 5 or exons 20-22 were found in two probands, heterozygous deep-intronic variants were identified in six probands, and a deep-intronic variant was found together with an exon 20-22 deletion in one proband. Based on ophthalmologic findings and characteristics of the identified exonic variants present in trans, the deep-intronic variants V1 and V4 were predicted to be relatively mild and severe, respectively. These findings are important for proper genetic counseling and for the development of variant-specific therapies., (© 2014 WILEY PERIODICALS, INC.)
- Published
- 2015
- Full Text
- View/download PDF
39. The RD5000 database: facilitating clinical, genetic, and therapeutic studies on inherited retinal diseases.
- Author
-
van Huet RA, Oomen CJ, Plomp AS, van Genderen MM, Klevering BJ, Schlingemann RO, Klaver CC, van den Born LI, and Cremers FP
- Subjects
- DNA Mutational Analysis, Databases, Factual, Humans, DNA genetics, Genetic Predisposition to Disease, Mutation, Retina pathology, Retinal Diseases diagnosis, Retinal Diseases genetics, Retinal Diseases therapy
- Abstract
Inherited retinal diseases (IRDs) represent a clinical and genetic heterogeneous group of chorioretinal disorders. The frequency of persons affected by an IRD due to mutations in the same gene varies from 1 in 10,000 to less than 1 in a million. To perform meaningful genotype-phenotype analyses for rare genetic conditions, it is necessary to collect data from sizable populations. Although several standardized functional tests are used widely, ophthalmologic data usually are stored in local databases and not in multicenter databases that are linked with other centers. To be able to register ophthalmologic data of all Dutch patients with IRDs into one database, we developed the RD5000 database (RD5000db), which can harbor all ophthalmologic and selected genetic data. Authorization rights for the management, data entry, and data sharing have been set up, rendering this database into a user-friendly, secure, and widely used repository that will facilitate future studies into molecular genetics and therapies for IRDs. The RD5000db database has the potential to grow into a European standard for the registration of data from IRDs., (Copyright 2014 The Association for Research in Vision and Ophthalmology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
40. Chromosomal aberrations in cerebral visual impairment.
- Author
-
Bosch DG, Boonstra FN, Reijnders MR, Pfundt R, Cremers FP, and de Vries BB
- Subjects
- Blindness, Cortical epidemiology, Blindness, Cortical physiopathology, Child, Child, Preschool, Chromosome Mapping, Cohort Studies, Down Syndrome complications, Female, Humans, Male, Visual Acuity genetics, Blindness, Cortical genetics, Chromosome Aberrations, Genetic Predisposition to Disease, Visually Impaired Persons
- Abstract
Background: Cerebral visual impairment (CVI) is a disorder in projection and/or interpretation of the visual input in the brain and accounts for 27% of the visually impaired children., Aim: A large cohort of patients with CVI was investigated in order to ascertain the relevance of chromosomal aberrations in the etiology of this disorder., Methods: 607 patients with CVI and a visual acuity ≤0.3 were assessed for the presence of a chromosomal aberration retrospectively. The observed aberrations were classified for pathogenicity., Results: A total of 98 chromosomal aberrations were found in 79 persons (13%) of the cohort. In nine persons it was not possible to classify the clinical implication of the aberration, due to lack of detailed information. In 70 persons it was possible to classify the aberration for causality: in 41 patients the aberration was associated with CVI, in 16 it was unknown and in 13 the aberration was unlikely to be associated with CVI. For four aberrations, present in 26 patients, the association with CVI has been reported before: trisomy 21, 1p36 deletion syndrome, 17p13.3 deletion syndrome (Miller-Dieker syndrome) and 22q13.3 deletion syndrome (Phelan-McDermid syndrome). The chromosomal aberrations in another 15 patients were for the first time associated with CVI., Conclusions: Chromosomal aberrations associated with CVI were found in 7% (41/607) of patients, of which 37% (15/41) have not been reported before in association with CVI. Therefore, in patients with CVI chromosomal investigations should be routinely performed to warrant a good clinical diagnosis and counseling., (Copyright © 2014 European Paediatric Neurology Society. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
41. Oral 9-cis retinoid for childhood blindness due to Leber congenital amaurosis caused by RPE65 or LRAT mutations: an open-label phase 1b trial.
- Author
-
Koenekoop RK, Sui R, Sallum J, van den Born LI, Ajlan R, Khan A, den Hollander AI, Cremers FP, Mendola JD, Bittner AK, Dagnelie G, Schuchard RA, and Saperstein DA
- Subjects
- Acyltransferases deficiency, Acyltransferases genetics, Administration, Oral, Adolescent, Adult, Blindness genetics, Child, Diterpenes, Humans, Leber Congenital Amaurosis genetics, Mutation genetics, Prospective Studies, Retinyl Esters, Visual Acuity drug effects, Vitamin A administration & dosage, Young Adult, cis-trans-Isomerases deficiency, cis-trans-Isomerases genetics, Blindness drug therapy, Leber Congenital Amaurosis drug therapy, Vitamin A analogs & derivatives
- Abstract
Background: Leber congenital amaurosis, caused by mutations in RPE65 and LRAT, is a severe form of inherited retinal degeneration leading to blindness. We aimed to assess replacement of the missing chromophore 11-cis retinal with oral QLT091001 (synthetic 9-cis-retinyl acetate) in these patients., Methods: In our open-label, prospective, phase 1b trial, we enrolled patients (aged ≥6 years) with Leber congenital amaurosis and RPE65 or LRAT mutations at McGill University's Montreal Children's Hospital. Patients received 7 days of oral QLT091001 (10-40 mg/m(2) per day). We assessed patients at baseline and days 7, 9, 14, and 30, and then 2 months and every 2 months thereafter for up to 2·2 years for safety outcomes and visual function endpoints including Goldmann visual fields (GVF), visual acuity, and functional MRI assessment. We regarded patients as having an improvement in vision if we noted at least a 20% improvement in retinal area on GVF compared with baseline or a visual acuity improvement of five or more letters compared with baseline in two consecutive study visits (or any improvement from no vision at baseline). This study is registered with ClinicalTrials.gov, number NCT01014052., Findings: Between December, 2009, and June, 2011, we enrolled and treated 14 patients aged 6-38 years who were followed up until March, 2012. Ten (71%) of 14 patients had an improvement in GVF areas (mean increase in retinal area of 28-683%). Six (43%) patients had an improvement in visual acuity (mean increase of 2-30 letters). Self-reported or parent-reported improvements in activities of daily living supported these findings. After 2 years, 11 (79%) patients had returned to their baseline GVF retinal area and ten (71%) had returned to baseline visual acuity letter values. Thus, three (21%) patients had a sustained GVF response and four (30%) had a sustained visual acuity response. Four patients had functional MRI scans, which correlated with visual response or absence of response to treatment. No serious adverse events occurred, although we noted transient headaches (11 patients), photophobia (11 patients), reduction in serum HDL concentrations (four patients), and increases in serum triglycerides (eight patients) and aspartate aminotransferase concentrations (two patients)., Interpretation: Non-invasive oral QLT091001 therapy is well tolerated, and can rapidly improve visual function in some patients with Leber congenital amaurosis and RPE65 and LRAT mutations., Funding: QLT, Foundation Fighting Blindness Canada, CIHR, FRSQ, Reseau Vision.
- Published
- 2014
- Full Text
- View/download PDF
42. Foveal sparing in Stargardt disease.
- Author
-
van Huet RA, Bax NM, Westeneng-Van Haaften SC, Muhamad M, Zonneveld-Vrieling MN, Hoefsloot LH, Cremers FP, Boon CJ, Klevering BJ, and Hoyng CB
- Subjects
- ATP-Binding Cassette Transporters metabolism, Adult, Aged, DNA Mutational Analysis, Electroretinography, Female, Fluorescein Angiography, Fovea Centralis metabolism, Fundus Oculi, Humans, Macular Degeneration diagnosis, Macular Degeneration genetics, Macular Degeneration metabolism, Male, Middle Aged, Ophthalmoscopy, Retinal Pigment Epithelium metabolism, Rod Cell Outer Segment, Stargardt Disease, Tomography, Optical Coherence, Visual Acuity, ATP-Binding Cassette Transporters genetics, DNA genetics, Fovea Centralis pathology, Mutation, Retinal Pigment Epithelium pathology
- Abstract
Purpose: To provide a clinical and genetic description of a patient cohort with Stargardt disease (STGD1) with identifiable foveal sparing., Methods: Patients with retinal atrophy (defined as an absence of autofluorescence) that surrounded the fovea by at least 180° and did not include the fovea were defined as having foveal sparing; eyes with visual acuity (VA) worse than 20/200 were excluded. We reviewed the medical files and extracted data regarding medical history, VA, ophthalmoscopy, static perimetry, fundus photography, spectral-domain optical coherence tomography (SD-OCT), fluorescein angiography (FA), fundus autofluorescence (FAF), and electroretinography (ERG). We screened each patient's ABCA4 gene for mutations., Results: Seventeen eyes with foveal sparing were identified in 13 unrelated patients. In 4 eyes, the fovea gradually became atrophic after the initial foveal sparing. The mean age at onset was 51 years (range, 32-67 years). Visual acuity was 20/40 or better in all foveal sparing eyes and was 20/25 or better in 41%. Fundus autofluorescence imaging revealed hyperautofluorescent flecks and parafoveal retinal atrophy; SD-OCT revealed sharply delineated atrophy; and perimetry revealed parafoveal scotomas with intact foveal sensitivity. Finally, genetic screening identified mutations in 19 of the 26 ABCA4 gene alleles., Conclusions: Foveal sparing occurs mainly in patients with late-onset STGD1 and represents the milder end of the clinical spectrum in STGD1. The anatomy, metabolism, and biochemistry of the retina, as well as genetic variations in genes other than ABCA4, can influence the etiology of foveal sparing. Identifying these fovea-protecting factors will facilitate the future development of strategies designed to treat STGD1., (Copyright 2014 The Association for Research in Vision and Ophthalmology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
43. Causes and consequences of inherited cone disorders.
- Author
-
Roosing S, Thiadens AA, Hoyng CB, Klaver CC, den Hollander AI, and Cremers FP
- Subjects
- Animals, Color Vision Defects congenital, Disease Models, Animal, Humans, Retinal Degeneration congenital, Color Vision Defects genetics, Eye Proteins genetics, Mutation, Retinal Cone Photoreceptor Cells physiology, Retinal Degeneration genetics
- Abstract
Hereditary cone disorders (CDs) are characterized by defects of the cone photoreceptors or retinal pigment epithelium underlying the macula, and include achromatopsia (ACHM), cone dystrophy (COD), cone-rod dystrophy (CRD), color vision impairment, Stargardt disease (STGD) and other maculopathies. Forty-two genes have been implicated in non-syndromic inherited CDs. Mutations in the 5 genes implicated in ACHM explain ∼93% of the cases. On the contrary, only 21% of CRDs (17 genes) and 25% of CODs (8 genes) have been elucidated. The fact that the large majority of COD and CRD-associated genes are yet to be discovered hints towards the existence of unknown cone-specific or cone-sensitive processes. The ACHM-associated genes encode proteins that fulfill crucial roles in the cone phototransduction cascade, which is the most frequently compromised (10 genes) process in CDs. Another 7 CD-associated proteins are required for transport processes towards or through the connecting cilium. The remaining CD-associated proteins are involved in cell membrane morphogenesis and maintenance, synaptic transduction, and the retinoid cycle. Further novel genes are likely to be identified in the near future by combining large-scale DNA sequencing and transcriptomics technologies. For 31 of 42 CD-associated genes, mammalian models are available, 14 of which have successfully been used for gene augmentation studies. However, gene augmentation for CDs should ideally be developed in large mammalian models with cone-rich areas, which are currently available for only 11 CD genes. Future research will aim to elucidate the remaining causative genes, identify the molecular mechanisms of CD, and develop novel therapies aimed at preventing vision loss in individuals with CD in the future., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
44. Disruption of the basal body protein POC1B results in autosomal-recessive cone-rod dystrophy.
- Author
-
Roosing S, Lamers IJ, de Vrieze E, van den Born LI, Lambertus S, Arts HH, Peters TA, Hoyng CB, Kremer H, Hetterschijt L, Letteboer SJ, van Wijk E, Roepman R, den Hollander AI, and Cremers FP
- Subjects
- Amino Acid Sequence, Animals, Basal Bodies, Base Sequence, Cell Cycle Proteins metabolism, Cells, Cultured, Exome genetics, Eye Proteins genetics, Female, Gene Knockdown Techniques, HEK293 Cells, Humans, Male, Molecular Sequence Data, Morpholinos genetics, Mutation, Missense, Netherlands, Photoreceptor Connecting Cilium metabolism, Retinal Photoreceptor Cell Outer Segment physiology, Retinal Pigment Epithelium metabolism, Retinal Pigment Epithelium pathology, Sequence Analysis, DNA, Turkey, Vision Disorders genetics, Zebrafish, Cell Cycle Proteins genetics, Eye Proteins metabolism, Retinal Cone Photoreceptor Cells pathology, Retinal Rod Photoreceptor Cells pathology, Retinitis Pigmentosa genetics
- Abstract
Exome sequencing revealed a homozygous missense mutation (c.317C>G [p.Arg106Pro]) in POC1B, encoding POC1 centriolar protein B, in three siblings with autosomal-recessive cone dystrophy or cone-rod dystrophy and compound-heterozygous POC1B mutations (c.199_201del [p.Gln67del] and c.810+1G>T) in an unrelated person with cone-rod dystrophy. Upon overexpression of POC1B in human TERT-immortalized retinal pigment epithelium 1 cells, the encoded wild-type protein localized to the basal body of the primary cilium, whereas this localization was lost for p.Arg106Pro and p.Gln67del variant forms of POC1B. Morpholino-oligonucleotide-induced knockdown of poc1b translation in zebrafish resulted in a dose-dependent small-eye phenotype, impaired optokinetic responses, and decreased length of photoreceptor outer segments. These ocular phenotypes could partially be rescued by wild-type human POC1B mRNA, but not by c.199_201del and c.317C>G mutant human POC1B mRNAs. Yeast two-hybrid screening of a human retinal cDNA library revealed FAM161A as a binary interaction partner of POC1B. This was confirmed in coimmunoprecipitation and colocalization assays, which both showed loss of FAM161A interaction with p.Arg106Pro and p.Gln67del variant forms of POC1B. FAM161A was previously implicated in autosomal-recessive retinitis pigmentosa and shown to be located at the base of the photoreceptor connecting cilium, where it interacts with several other ciliopathy-associated proteins. Altogether, this study demonstrates that POC1B mutations result in a defect of the photoreceptor sensory cilium and thus affect cone and rod photoreceptors., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
45. Exome sequencing extends the phenotypic spectrum for ABHD12 mutations: from syndromic to nonsyndromic retinal degeneration.
- Author
-
Nishiguchi KM, Avila-Fernandez A, van Huet RA, Corton M, Pérez-Carro R, Martín-Garrido E, López-Molina MI, Blanco-Kelly F, Hoefsloot LH, van Zelst-Stams WA, García-Ruiz PJ, Del Val J, Di Gioia SA, Klevering BJ, van de Warrenburg BP, Vazquez C, Cremers FP, García-Sandoval B, Hoyng CB, Collin RW, Rivolta C, and Ayuso C
- Subjects
- Adult, Aged, Ataxia diagnosis, Ataxia physiopathology, Audiometry, Cataract diagnosis, Cataract physiopathology, Electroretinography, Female, Genes, Recessive, Humans, Magnetic Resonance Imaging, Male, Middle Aged, Monoacylglycerol Lipases chemistry, Pedigree, Phenotype, Polyneuropathies diagnosis, Polyneuropathies physiopathology, Protein Structure, Secondary, Retinitis Pigmentosa diagnosis, Retinitis Pigmentosa physiopathology, Sequence Analysis, DNA, Visual Acuity physiology, Visual Fields physiology, Ataxia genetics, Cataract genetics, Exome genetics, Monoacylglycerol Lipases genetics, Mutation, Missense, Polyneuropathies genetics, Retinitis Pigmentosa genetics
- Abstract
Objective: To identify the genetic causes underlying autosomal recessive retinitis pigmentosa (arRP) and to describe the associated phenotype., Design: Case series., Participants: Three hundred forty-seven unrelated families affected by arRP and 33 unrelated families affected by retinitis pigmentosa (RP) plus noncongenital and progressive hearing loss, ataxia, or both, respectively., Methods: A whole exome sequencing (WES) analysis was performed in 2 families segregating arRP. A mutational screening was performed in 378 additional unrelated families for the exon-intron boundaries of the ABHD12 gene. To establish a genotype-phenotype correlation, individuals who were homozygous or compound heterozygotes of mutations in ABHD12 underwent exhaustive clinical examinations by ophthalmologists, neurologists, and otologists., Main Outcome Measures: DNA sequence variants, best-corrected visual acuity, visual field assessments, electroretinogram responses, magnetic resonance imaging, and audiography., Results: After a WES analysis, we identified 4 new mutations (p.Arg107Glufs*8, p.Trp159*, p.Arg186Pro, and p.Thr202Ile) in ABHD12 in 2 families (RP-1292 and W08-1833) previously diagnosed with nonsyndromic arRP, which cosegregated with the disease among the family members. Another homozygous mutation (p.His372Gln) was detected in 1 affected individual (RP-1487) from a cohort of 378 unrelated arRP and syndromic RP patients. After exhaustive clinical examinations by neurologists and otologists, the 4 affected members of the RP-1292 had no polyneuropathy or ataxia, and the sensorineural hearing loss and cataract were attributed to age or the normal course of the RP, whereas the affected members of the families W08-1833 and RP-1487 showed clearly symptoms associated with polyneuropathy, hearing loss, cerebellar ataxia, RP, and early-onset cataract (PHARC) syndrome., Conclusions: Null mutations in the ABHD12 gene lead to PHARC syndrome, a neurodegenerative disease including polyneuropathy, hearing loss, cerebellar ataxia, RP, and early-onset cataract. Our study allowed us to report 5 new mutations in ABHD12. This is the first time missense mutations have been described for this gene. Furthermore, these findings are expanding the spectrum of phenotypes associated with ABHD12 mutations ranging from PHARC syndrome to a nonsyndromic form of retinal degeneration., (Copyright © 2014 American Academy of Ophthalmology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
46. Nonpenetrance of the most frequent autosomal recessive leber congenital amaurosis mutation in NMNAT1.
- Author
-
Siemiatkowska AM, Schuurs-Hoeijmakers JH, Bosch DG, Boonstra FN, Riemslag FC, Ruiter M, de Vries BB, den Hollander AI, Collin RW, and Cremers FP
- Subjects
- Humans, Leber Congenital Amaurosis genetics, Mutation, Nicotinamide-Nucleotide Adenylyltransferase genetics
- Abstract
Importance: The NMNAT1 gene was recently found to be mutated in a subset of patients with Leber congenital amaurosis and macular atrophy. The most prevalent NMNAT1 variant was p.Glu257Lys, which was observed in 38 of 106 alleles (35.8%). On the basis of functional assays, it was deemed a severe variant., Observations: The p.Glu257Lys variant was 80-fold less frequent in a homozygous state in patients with Leber congenital amaurosis than predicted based on its heterozygosity frequency in the European American population. Moreover, we identified this variant in a homozygous state in a patient with no ocular abnormalities., Conclusions and Relevance: On the basis of these results, the p.Glu257Lys variant is considered not fully penetrant. Homozygotes of the p.Glu257Lys variant in most persons are therefore not associated with ocular disease. Consequently, genetic counselors should exercise great caution in the interpretation of this variant.
- Published
- 2014
- Full Text
- View/download PDF
47. A missense mutation in the splicing factor gene DHX38 is associated with early-onset retinitis pigmentosa with macular coloboma.
- Author
-
Ajmal M, Khan MI, Neveling K, Khan YM, Azam M, Waheed NK, Hamel CP, Ben-Yosef T, De Baere E, Koenekoop RK, Collin RW, Qamar R, and Cremers FP
- Subjects
- Base Sequence, DNA Mutational Analysis, Female, Genes, Recessive, Genetic Association Studies, Genetic Linkage, Genetic Predisposition to Disease, Humans, Male, Pedigree, Polymorphism, Single Nucleotide, RNA Splicing Factors, Coloboma genetics, DEAD-box RNA Helicases genetics, Macula Lutea abnormalities, Mutation, Missense, Retinitis Pigmentosa genetics
- Abstract
Background: Retinitis pigmentosa (RP) is the most frequent inherited retinal disease, which shows a relatively high incidence of the autosomal-recessive form in Pakistan., Methods: Genome-wide high-density single-nucleotide polymorphism (SNP) microarrays were used to identify homozygous regions shared by affected individuals of one consanguineous family. DNA of three affected and two healthy siblings was used for SNP genotyping. Genotyping data were then analysed by Homozygosity Mapper. DNA of the proband was further analysed employing exome sequencing., Results: Homozygosity mapping revealed a single homozygous region on chromosome 16, shared by three affected individuals. Subsequent exome sequencing identified a novel missense mutation, c.995G>A; p.(Gly332Asp), in DHX38. This mutation was found to be present in a homozygous state in four affected individuals while two healthy siblings and the parents of the affected persons were heterozygous for this mutation. This variant thereby yields a logarithm of the odds (LOD) score of 3.25, which is highly suggestive for linkage. This variant was neither detected in 180 ethnically matched control individuals, nor in 7540 Africans or Caucasians and an in-house database that contained the exome data of 400 individuals., Conclusions: By combining genome-wide homozygosity mapping and exome sequencing, a novel missense mutation was identified in the DHX38 gene that encodes the pre-mRNA splicing factor PRP16, in a Pakistani family with early-onset autosomal-recessive RP. The phenotype is different from those associated with other retinal pre-mRNA splicing factors and DHX38 is the first pre-mRNA splicing gene that is putatively associated with autosomal-recessive inherited RP., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://group.bmj.com/group/rights-licensing/permissions.)
- Published
- 2014
- Full Text
- View/download PDF
48. Genomic approaches for the discovery of genes mutated in inherited retinal degeneration.
- Author
-
Siemiatkowska AM, Collin RW, den Hollander AI, and Cremers FP
- Subjects
- DNA Copy Number Variations genetics, Exome genetics, Gene Expression Profiling methods, Gene Frequency genetics, Genetic Linkage genetics, Genetic Techniques, Genetic Testing methods, Genome-Wide Association Study methods, Humans, Genomics methods, Mutation genetics, Retinal Degeneration genetics
- Abstract
In view of their high degree of genetic heterogeneity, inherited retinal diseases (IRDs) pose a significant challenge for identifying novel genetic causes. Thus far, more than 200 genes have been found to be mutated in IRDs, which together contain causal variants in >80% of the cases. Accurate genetic diagnostics is particularly important for isolated cases, in which X-linked and de novo autosomal dominant variants are not uncommon. In addition, new gene- or mutation-specific therapies are emerging, underlining the importance of identifying causative mutations in each individual. Sanger sequencing of selected genes followed by cost-effective targeted next-generation sequencing (NGS) can identify defects in known IRD-associated genes in the majority of the cases. Exome NGS in combination with genetic linkage or homozygosity mapping studies can aid the identification of the remaining causal genes. As these are thought to be mutated in <1% of the cases, validation through functional modeling in, for example, zebrafish and/or replication through the genotyping of large patient cohorts is required. In the near future, whole genome NGS in combination with transcriptome NGS may reveal mutations that are currently hidden in the noncoding regions of the human genome., (Copyright © 2014 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
49. Novel compound heterozygous NMNAT1 variants associated with Leber congenital amaurosis.
- Author
-
Siemiatkowska AM, van den Born LI, van Genderen MM, Bertelsen M, Zobor D, Rohrschneider K, van Huet RA, Nurohmah S, Klevering BJ, Kohl S, Faradz SM, Rosenberg T, den Hollander AI, Collin RW, and Cremers FP
- Subjects
- Adolescent, Adult, Aged, Child, Child, Preschool, Female, Humans, Male, Middle Aged, Mutation genetics, Young Adult, Genetic Association Studies, Genetic Predisposition to Disease, Heterozygote, Leber Congenital Amaurosis enzymology, Leber Congenital Amaurosis genetics, Nicotinamide-Nucleotide Adenylyltransferase genetics
- Abstract
Purpose: The gene encoding nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) was recently found to be mutated in a subset of patients with Leber congenital amaurosis (LCA) with macular atrophy. The aim of this study was to determine the occurrence and frequency of NMNAT1 mutations and associated phenotypes in different types of inherited retinal dystrophies., Methods: DNA samples of 161 patients with LCA without genetic diagnosis were analyzed for variants in NMNAT1 using Sanger sequencing. Variants in exon 5 of NMNAT1, which harbors the majority of the previously identified mutations, were screened in 532 additional patients with retinal dystrophies. This cohort encompassed 108 persons with isolated or autosomal recessive cone-rod dystrophy (CRD), 271 with isolated or autosomal recessive retinitis pigmentosa (RP), and 49 with autosomal dominant RP, as well as 104 persons with LCA in whom the causative mutation was previously identified., Results: Compound heterozygous alterations were found in six patients with LCA and in one person with early-onset RP. All except one carried the common p.E257K variant on one allele. Macular atrophy was absent in one patient, who carried this variant in combination with a truncating mutation on the other allele. The p.E257K alteration was also found in a heterozygous state in five individuals with LCA and one with RP while no mutation was detected on the other allele. Two individuals with LCA carried other NMNAT1 variants in a heterozygous state, whereas no NMNAT1 variants in exon 5 were identified in individuals with CRD. The p.E257K variant was found to be enriched in a heterozygous state in individuals with LCA (0.94%) compared to Caucasian controls (0.18%), although the difference was statistically insignificant (p=0.12)., Conclusions: Although macular atrophy can occur in LCA and CRD, no NMNAT1 mutations were found in the latter cohort. NMNAT1 variants were also not found in a large group of patients with sporadic or autosomal recessive RP. The enrichment of p.E257K in a heterozygous state in patients with LCA versus controls suggests that this allele could act as a modifier in other genetic subtypes of LCA.
- Published
- 2014
50. IMPG2-associated retinitis pigmentosa displays relatively early macular involvement.
- Author
-
van Huet RA, Collin RW, Siemiatkowska AM, Klaver CC, Hoyng CB, Simonelli F, Khan MI, Qamar R, Banin E, Cremers FP, Theelen T, den Hollander AI, van den Born LI, and Klevering BJ
- Subjects
- Adult, Age of Onset, Aged, Color Perception Tests, Corneal Dystrophies, Hereditary diagnosis, Corneal Dystrophies, Hereditary genetics, DNA Mutational Analysis, Electroretinography, Female, Humans, Male, Middle Aged, Night Blindness diagnosis, Night Blindness genetics, Ophthalmoscopy, Pedigree, Retinitis Pigmentosa diagnosis, Tomography, Optical Coherence, Vision Disorders diagnosis, Vision Disorders genetics, Visual Acuity physiology, Visual Field Tests, Visual Fields physiology, Young Adult, Genes, Recessive, Mutation, Proteoglycans genetics, Retinitis Pigmentosa genetics
- Abstract
Purpose: To provide the first detailed clinical description in patients with RP caused by recessive mutations in IMPG2., Methods: This international collaborative study includes 17 RP patients with inherited retinal disease caused by mutations in IMPG2. The patients were clinically (re-)examined, including extensive medical history taking, slit-lamp biomicroscopy, ophthalmoscopy, perimetry, ERG, optical coherence tomography (OCT), fundus autofluorescence (FAF) imaging, fundus photography, and color vision tests. The main outcome measures included mean age at onset, initial symptom, best-corrected visual acuity, fundus appearance, perimetry results, ERG responses, OCT images, FAF imaging, color vision test reports and DNA sequence variants., Results: The mean age at onset was 10.5 years (range, 4-20 years). Initial symptoms included night blindness in 59% of patients, a decreased visual acuity in 35%, and visual field loss in 6%. Fundus abnormalities were typical of RP: optic disc pallor, attenuated vessels, bone spicules, and generalized atrophy of the retina and choriocapillaris. Additionally, we observed macular abnormalities in all patients, ranging from subtle mottling of the macular pigment epithelium (two patients) and a bull's eye maculopathy (seven patients) to macular chorioretinal atrophy (seven patients)., Conclusions: Mutations in IMPG2 cause a severe form of RP with symptoms manifesting in the first 2 decades of life. IMPG2-associated RP is frequently accompanied by macular involvement, ranging from mild pigment alterations to profound chorioretinal atrophy. The resulting decrease in central vision in combination with the severe tunnel vision leads to severe visual impairment in patients with IMPG2-associated RP., (Copyright 2014 The Association for Research in Vision and Ophthalmology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.