204 results on '"Bernd, Wissinger"'
Search Results
2. Splicing defects and CRISPR-Cas9 correction in isogenic homozygous photoreceptor precursors harboring clustered deep-intronic ABCA4 variants
- Author
-
Pietro De Angeli, Arturo Flores-Tufiño, Katarina Stingl, Laura Kühlewein, Eleonora Roschi, Bernd Wissinger, and Susanne Kohl
- Subjects
MT: RNA/DNA Editing ,gene editing ,CRISPR-Cas9 ,inherited retinal disease ,splicing ,Stargardt ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Splicing defects from deep-intronic variants significantly contribute to the mutational spectrum in ABCA4-associated inherited retinal diseases, necessitating functional validation for their pathological classification. Typically, minigene assays in HEK293(T) can qualitatively assess splicing defects, yet they often fail to quantitatively reproduce the resulting mis-splicing patterns, leaving uncertainty on severity and pathogenicity. As a potential cellular model derived from patient cells, photoreceptor precursor cells (PPCs) play a pivotal role in assessing the severity of specific splicing mutations. Nevertheless, the accessibility of biosamples is commonly constrained, and their establishment is costly and laborious. In this study, we combined and investigated the use of a minigene assay and isogenic PPCs, as superior qualitative and more accessible cellular models for the assessment of splicing defects. Specifically, we focused on the clustered c.5196+1013A>G, c.5196+1056A>G, and c.5196+1216C>A deep-intronic variants in intron 36 of ABCA4, comparing their resulting (mis)splicing patterns in minigene-transfected cells and isogenic CRISPR-Cas9-knocked-in PPCs harboring these pathogenic variants in homozygous state. Moreover, we demonstrate the successful correction of these three splicing defects in homozygous mutant PPCs using a single pair of guide RNAs to target Cas9 cleavage, thereby identifying an efficient gene editing strategy for therapeutic applications.
- Published
- 2024
- Full Text
- View/download PDF
3. Extra-viral DNA in adeno-associated viral vector preparations induces TLR9-dependent innate immune responses in human plasmacytoid dendritic cells
- Author
-
Kirsten Bucher, Eduardo Rodríguez-Bocanegra, Bernd Wissinger, Torsten Strasser, Simon J. Clark, Andreas L. Birkenfeld, Dorothea Siegel-Axel, and M. Dominik Fischer
- Subjects
Medicine ,Science - Abstract
Abstract Adeno-associated viral (AAV) vector suspensions produced in either human derived HEK cells or in Spodoptera frugiperda (Sf9) insect cells differ in terms of residual host cell components as well as species-specific post-translational modifications displayed on the AAV capsid proteins. Here we analysed the impact of these differences on the immunogenic properties of the vector. We stimulated human plasmacytoid dendritic cells with various lots of HEK cell-produced and Sf9 cell-produced AAV-CMV-eGFP vectors derived from different manufacturers. We found that AAV8-CMV-eGFP as well as AAV2-CMV-eGFP vectors induced lot-specific but not production platform-specific or manufacturer-specific inflammatory cytokine responses. These could be reduced or abolished by blocking toll-like receptor 9 signalling or by enzymatically reducing DNA in the vector lots using DNase. Successful HEK cell transduction by DNase-treated AAV lots and DNA analyses demonstrated that DNase did not affect the integrity of the vector but degraded extra-viral DNA. We conclude that both HEK- and Sf9-cell derived AAV preparations can contain immunogenic extra-viral DNA components which can trigger lot-specific inflammatory immune responses. This suggests that improved strategies to remove extra-viral DNA impurities may be instrumental in reducing the immunogenic properties of AAV vector preparations.
- Published
- 2023
- Full Text
- View/download PDF
4. Genetic background modulates phenotypic expressivity in OPA1 mutated mice, relevance to DOA pathogenesis
- Author
-
Djamaa Atamena, Venu Gurram, Petnoï Petsophonsakul, Farnoosh Khosrobakhsh, Macarena S. Arrázola, Marlène Botella, Bernd Wissinger, Marion Szelechowski, and Pascale Belenguer
- Subjects
optic atrophy ,OPA1 ,mitochondria ,genetic modifiers ,disease severity ,mouse strains ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Dominant optic atrophy (DOA) is mainly caused by OPA1 mutations and is characterized by the degeneration of retinal ganglion cells (RGCs), whose axons form the optic nerve. The penetrance of DOA is incomplete and the disease is marked by highly variable expressivity, ranging from asymptomatic patients to some who are totally blind or who suffer from multisystemic effects. No clear genotype–phenotype correlation has been established to date. Taken together, these observations point toward the existence of modifying genetic and/or environmental factors that modulate disease severity. Here, we investigated the influence of genetic background on DOA expressivity by switching the previously described DOA mouse model bearing the c.1065 + 5G → A Opa1 mutation from mixed C3H; C57BL/6 J to a pure C57BL/6 J background. We no longer observed retinal and optic nerve abnormalities; the findings indicated no degeneration, but rather a sex-dependent negative effect on RGC connectivity. This highlights the fact that RGC synaptic alteration might precede neuronal death, as has been proposed in other neurodegenerative diseases, providing new clinical considerations for early diagnosis as well as a new therapeutic window for DOA. Furthermore, our results demonstrate the importance of secondary genetic factors in the variability of DOA expressivity and offer a model for screening for aggravating environmental and genetic factors.
- Published
- 2023
- Full Text
- View/download PDF
5. Effective splicing restoration of a deep-intronic ABCA4 variant in cone photoreceptor precursor cells by CRISPR/SpCas9 approaches
- Author
-
Pietro De Angeli, Peggy Reuter, Stefan Hauser, Ludger Schöls, Katarina Stingl, Bernd Wissinger, and Susanne Kohl
- Subjects
MT: RNA/DNA editing ,ABCA4 ,Stargardt disease ,STGD1 ,inherited retinal dystrophy ,deep-intronic variants ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Stargardt disease is an autosomal recessively inherited retinal disorder commonly caused by pathogenic variants in the ABCA4 gene encoding the ATP-binding cassette subfamily A member 4 (ABCA4) protein. Several deep-intronic variants in ABCA4 have been classified as disease causing. By strengthening a cryptic splice site, deep-intronic variant c.5197-557G>T induces the inclusion of a 188-bp intronic sequence in the mature mRNA, resulting in a premature termination codon. Here, we report the design and evaluation of three CRISPR-Cas9 approaches implementing Streptococcus pyogenes Cas9 (single and dual guide RNA) or Streptococcus pyogenes Cas9 nickase (dual guide RNA) for their potential to correct c.5197-557G>T-induced aberrant splicing in minigene splicing assays and patient-derived cone photoreceptor precursor cells. The different strategies were able to rescue correct splicing by up to 83% and increase the overall correctly spliced transcripts by 1.8-fold, demonstrating the successful CRISPR-Cas9-mediated rescue in patient-derived photoreceptor precursor cells of an ABCA4 splicing defect. The results provide initial evidence of possible permanent splicing correction for Stargardt disease, expanding the therapeutic toolbox to counteract deep-intronic pathogenic variants in ABCA4.
- Published
- 2022
- Full Text
- View/download PDF
6. Whole genome sequencing for USH2A-associated disease reveals several pathogenic deep-intronic variants that are amenable to splice correction
- Author
-
Janine Reurink, Nicole Weisschuh, Alejandro Garanto, Adrian Dockery, L. Ingeborgh van den Born, Isabelle Fajardy, Lonneke Haer-Wigman, Susanne Kohl, Bernd Wissinger, G. Jane Farrar, Tamar Ben-Yosef, Fatma Kivrak Pfiffner, Wolfgang Berger, Marianna E. Weener, Lubica Dudakova, Petra Liskova, Dror Sharon, Manar Salameh, Ashley Offenheim, Elise Heon, Giorgia Girotto, Paolo Gasparini, Anna Morgan, Arthur A. Bergen, Jacoline B. ten Brink, Caroline C.W. Klaver, Lisbeth Tranebjærg, Nanna D. Rendtorff, Sascha Vermeer, Jeroen J. Smits, Ronald J.E. Pennings, Marco Aben, Jaap Oostrik, Galuh D.N. Astuti, Jordi Corominas Galbany, Hester Y. Kroes, Milan Phan, Wendy A.G. van Zelst-Stams, Alberta A.H.J. Thiadens, Joke B.G.M. Verheij, Mary J. van Schooneveld, Suzanne E. de Bruijn, Catherina H.Z. Li, Carel B. Hoyng, Christian Gilissen, Lisenka E.L.M. Vissers, Frans P.M. Cremers, Hannie Kremer, Erwin van Wijk, and Susanne Roosing
- Subjects
USH2A ,Usher syndrome ,retinitis pigmentosa ,usherin ,whole genome sequencing ,minigene splice assay ,Genetics ,QH426-470 - Abstract
Summary: A significant number of individuals with a rare disorder such as Usher syndrome (USH) and (non-)syndromic autosomal recessive retinitis pigmentosa (arRP) remain genetically unexplained. Therefore, we assessed subjects suspected of USH2A-associated disease and no or mono-allelic USH2A variants using whole genome sequencing (WGS) followed by an improved pipeline for variant interpretation to provide a conclusive diagnosis.One hundred subjects were screened using WGS to identify causative variants in USH2A or other USH/arRP-associated genes. In addition to the existing variant interpretation pipeline, a particular focus was put on assessing splice-affecting properties of variants, both in silico and in vitro. Also structural variants were extensively addressed. For variants resulting in pseudoexon inclusion, we designed and evaluated antisense oligonucleotides (AONs) using minigene splice assays and patient-derived photoreceptor precursor cells.Biallelic variants were identified in 49 of 100 subjects, including novel splice-affecting variants and structural variants, in USH2A or arRP/USH-associated genes. Thirteen variants were shown to affect USH2A pre-mRNA splicing, including four deep-intronic USH2A variants resulting in pseudoexon inclusion, which could be corrected upon AON treatment.We have shown that WGS, combined with a thorough variant interpretation pipeline focused on assessing pre-mRNA splicing defects and structural variants, is a powerful method to provide subjects with a rare genetic condition, a (likely) conclusive genetic diagnosis. This is essential for the development of future personalized treatments and for patients to be eligible for such treatments.
- Published
- 2023
- Full Text
- View/download PDF
7. Autosomal dominant optic atrophy: A novel treatment for OPA1 splice defects using U1 snRNA adaption
- Author
-
Christoph Jüschke, Thomas Klopstock, Claudia B. Catarino, Marta Owczarek-Lipska, Bernd Wissinger, and John Neidhardt
- Subjects
gene therapy ,U1 snRNA ,ExSpeU1 ,splicing ,Dominant Optic Atrophy ,DOA ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Autosomal dominant optic atrophy (ADOA) is frequently caused by mutations in the optic atrophy 1 (OPA1) gene, with haploinsufficiency being the major genetic pathomechanism. Almost 30% of the OPA1-associated cases suffer from splice defects. We identified a novel OPA1 mutation, c.1065+5G>A, in patients with ADOA. In patient-derived fibroblasts, the mutation led to skipping of OPA1 exon 10, reducing the OPA1 protein expression by approximately 50%. We developed a molecular treatment to correct the splice defect in OPA1 using engineered U1 splice factors retargeted to different locations in OPA1 exon 10 or intron 10. The strongest therapeutic effect was detected when U1 binding was engineered to bind to intron 10 at position +18, a position predicted by bioinformatics to be a promising binding site. We were able to significantly silence the effect of the mutation (skipping of exon 10) and simultaneously increase the expression level of normal transcripts. Retargeting U1 to the canonical splice donor site did not lead to a detectable splice correction. This proof-of-concept study indicates for the first time the feasibility of splice mutation correction as a treatment option for ADOA. Increasing the amount of correctly spliced OPA1 transcripts may suffice to overcome the haploinsufficiency.
- Published
- 2021
- Full Text
- View/download PDF
8. First submicroscopic inversion of the OPA1 gene identified in dominant optic atrophy – a case report
- Author
-
Nicole Weisschuh, Pascale Mazzola, Tilman Heinrich, Tobias Haack, Bernd Wissinger, Felix Tonagel, and Carina Kelbsch
- Subjects
Case report ,Inversion ,Complex rearrangement ,Dominant optic atrophy ,OPA1 ,Non-homologous end joining (NHEJ) ,Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background Dominant optic atrophy (DOA) is an inherited optic neuropathy that mainly affects visual acuity, central visual fields and color vision due to a progressive loss of retinal ganglion cells and their axons that form the optic nerve. Approximately 45–90% of affected individuals with DOA harbor pathogenic variants in the OPA1 gene. The mutation spectrum of OPA1 comprises nonsense, canonical and non-canonical splice site, frameshift and missense as well as copy number variants, but intragenic inversions have not been reported so far. Case presentation We report a 33-year-old male with characteristic clinical features of DOA. Whole-genome sequencing identified a structural variant of 2.4 kb comprising an inversion of 937 bp at the OPA1 locus. Fine mapping of the breakpoints to single nucleotide level revealed that the structural variation was an inversion flanked by two deletions. As this rearrangement inverts the entire first exon of OPA1, it was classified as likely pathogenic. Conclusions We report the first DOA case harboring an inversion in the OPA1 gene. Our study demonstrates that copy-neutral genomic rearrangements have to be considered as a possible cause of disease in DOA cases.
- Published
- 2020
- Full Text
- View/download PDF
9. In Vivo Efficacy and Safety Evaluations of Therapeutic Splicing Correction Using U1 snRNA in the Mouse Retina
- Author
-
Sebastian Swirski, Oliver May, Malte Ahlers, Bernd Wissinger, Martin Greschner, Christoph Jüschke, and John Neidhardt
- Subjects
subretinal injection ,gene therapy ,U1 snRNA ,U6 snRNA ,AAV ,splice correction ,Cytology ,QH573-671 - Abstract
Efficacy and safety considerations constitute essential steps during development of in vivo gene therapies. Herein, we evaluated efficacy and safety of splice factor-based treatments to correct mutation-induced splice defects in an Opa1 mutant mouse line. We applied adeno-associated viruses to the retina. The viruses transduced retinal cells with an engineered U1 snRNA splice factor designed to correct the Opa1 splice defect. We found the treatment to be efficient in increasing wild-type Opa1 transcripts. Correspondingly, Opa1 protein levels increased significantly in treated eyes. Measurements of retinal morphology and function did not reveal therapy-related side-effects supporting the short-term safety of the treatment. Alterations of potential off-target genes were not detected. Our data suggest that treatments of splice defects applying engineered U1 snRNAs represent a promising in vivo therapeutic approach. The therapy increased wild-type Opa1 transcripts and protein levels without detectable morphological, functional or genetic side-effects in the mouse eye. The U1 snRNA-based therapy can be tailored to specific disease gene mutations, hence, raising the possibility of a wider applicability of this promising technology towards treatment of different inherited retinal diseases.
- Published
- 2023
- Full Text
- View/download PDF
10. Novel likely pathogenic variants in TMEM126A identified in non-syndromic autosomal recessive optic atrophy: two case reports
- Author
-
Katja Kloth, Matthis Synofzik, Christoph Kernstock, Simone Schimpf-Linzenbold, Frank Schuettauf, Axel Neu, Bernd Wissinger, and Nicole Weisschuh
- Subjects
Optic atrophy ,Autosomal recessive ,TMEM126A ,Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background Reports on autosomal recessive optic atrophy (arOA) are sparse and so far, only one gene has been specifically associated with non-syndromic arOA, namely TMEM126A. To date, all reports of pathogenic TMEM126A variants are from affected individuals of Maghrebian origin, who all carry an identical nonsense variant. Here we report two novel variants in the TMEM126A gene from non-Maghreb individuals, both found in affected individuals with an arOA phenotype. Case presentation We report three affected individuals from two families. The proband of family A, a 24-year-old Turkish woman, was diagnosed with visual loss in early childhood but a diagnosis of optic atrophy was only made at 14 years. A diagnostic gene panel revealed a splice donor variant (c.86 + 2 T > C) in homozygous state in the TMEM126A gene. Analysis of this variant based on RNA from whole blood revealed a single aberrant transcript lacking exon 2, presumably representing a functional null allele. Two siblings from family B, a 16-year old Iraqi girl and her 14-year old brother, were diagnosed with optic atrophy in early childhood. A missense variant p.(S36 L) in the TMEM126A gene was identified in homozygous state in a gene panel-based diagnostic setting in both siblings. This missense variant is ultra rare in the general population, affects a highly evolutionarily conserved amino acid and segregates with the disease within the family. The three probands reported in this study had a relatively mild clinical course without any evidence of a syndromic (e.g. neurological) comorbidity, which is in line with previous studies. Conclusions We provide additional evidence for the implication of biallelic pathogenic TMEM126A variants in arOA. Our findings extend both the mutational spectrum and geographic presence of TMEM126A in arOA. Screening of the entire gene should be considered in affected individuals presenting with features resembling arOA and also from non-Maghrebian descent.
- Published
- 2019
- Full Text
- View/download PDF
11. Mutation spectrum of the OPA1 gene in a large cohort of patients with suspected dominant optic atrophy: Identification and classification of 48 novel variants.
- Author
-
Nicole Weisschuh, Simone Schimpf-Linzenbold, Pascale Mazzola, Sinja Kieninger, Ting Xiao, Ulrich Kellner, Teresa Neuhann, Carina Kelbsch, Felix Tonagel, Helmut Wilhelm, Susanne Kohl, and Bernd Wissinger
- Subjects
Medicine ,Science - Abstract
Autosomal dominant optic atrophy is one of the most common inherited optic neuropathies. This disease is genetically heterogeneous, but most cases are due to pathogenic variants in the OPA1 gene: depending on the population studied, 32-90% of cases harbor pathogenic variants in this gene. The aim of this study was to provide a comprehensive overview of the entire spectrum of likely pathogenic variants in the OPA1 gene in a large cohort of patients. Over a period of 20 years, 755 unrelated probands with a diagnosis of bilateral optic atrophy were referred to our laboratory for molecular genetic investigation. Genetic testing of the OPA1 gene was initially performed by a combined analysis using either single-strand conformation polymorphism or denaturing high performance liquid chromatography followed by Sanger sequencing to validate aberrant bands or melting profiles. The presence of copy number variations was assessed using multiplex ligation-dependent probe amplification. Since 2012, genetic testing was based on next-generation sequencing platforms. Genetic screening of the OPA1 gene revealed putatively pathogenic variants in 278 unrelated probands which represent 36.8% of the entire cohort. A total of 156 unique variants were identified, 78% of which can be considered null alleles. Variant c.2708_2711del/p.(V903Gfs*3) was found to constitute 14% of all disease-causing alleles. Special emphasis was placed on the validation of splice variants either by analyzing cDNA derived from patients´ blood samples or by heterologous splice assays using minigenes. Splicing analysis revealed different aberrant splicing events, including exon skipping, activation of exonic or intronic cryptic splice sites, and the inclusion of pseudoexons. Forty-eight variants that we identified were novel. Nine of them were classified as pathogenic, 34 as likely pathogenic and five as variant of uncertain significance. Our study adds a significant number of novel variants to the mutation spectrum of the OPA1 gene and will thereby facilitate genetic diagnostics of patients with suspected dominant optic atrophy.
- Published
- 2021
- Full Text
- View/download PDF
12. Gene and Protein Expression in Subjects With a Nystagmus-Associated AHR Mutation
- Author
-
Natalia Borovok, Celeste Weiss, Rajech Sharkia, Michal Reichenstein, Bernd Wissinger, Abdussalam Azem, and Muhammad Mahajnah
- Subjects
aryl hydrocarbon receptor ,human mutation ,infantile nystagmus ,gene expression ,protein expression ,CYP1A1 ,Genetics ,QH426-470 - Abstract
Recently, a consanguineous family was identified in Israel with three children affected by Infantile Nystagmus and Foveal Hypoplasia, following an autosomal recessive mode of inheritance. A homozygous stop mutation c.1861C > T; p.Q621∗ in the aryl hydrocarbon receptor (AHR) gene (AHR; MIM 600253) was identified that co-segregated with the disease in the larger family. AHR is the first gene to be identified causing an autosomal recessive Infantile Nystagmus-related disease in humans. The goal of this study is to delineate the molecular basis of this newly discovered human genetic disorder associated with a rare AHR gene mutation. The gene and protein expression levels of AHR and selected AHR targets from leukocyte cultures of healthy subjects and the patients were analyzed. We observed significant variation between mRNA and protein expression of CYP1A1, CYP1B1, and TiPARP under rest and AHR-induced conditions. The CYP1A1 enzymatic activity in induced leukocytes also differs significantly between the patients and healthy volunteers. Intriguingly, the heterozygous subjects demonstrate CYP1A1 and TiPARP gene and protein expression similar to homozygous patients. In contrast, CYP1B1 inducibility and expression vary between hetero- and homozygous subjects. Similarity and differences in gene and protein expression between heterozygotes and homozygous patients can give us a hint as to which metabolic pathway/s might be involved in the Nystagmus etiology. Thus, we have a unique human model for AHR deficiency that will allow us the opportunity to study the biochemical basis of this rare human mutation, as well as the involvement of AHR in other physiological processes.
- Published
- 2020
- Full Text
- View/download PDF
13. Foveal Therapy in Blue Cone Monochromacy: Predictions of Visual Potential From Artificial Intelligence
- Author
-
Alexander Sumaroka, Artur V. Cideciyan, Rebecca Sheplock, Vivian Wu, Susanne Kohl, Bernd Wissinger, and Samuel G. Jacobson
- Subjects
machine learning ,random forest ,optical coherence tomography ,chromatic perimetry ,retinal degeneration ,rods ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Novel therapeutic approaches for treating inherited retinal degenerations (IRDs) prompt a need to understand which patients with impaired vision have the anatomical potential to gain from participation in a clinical trial. We used supervised machine learning to predict foveal function from foveal structure in blue cone monochromacy (BCM), an X-linked congenital cone photoreceptor dysfunction secondary to mutations in the OPN1LW/OPN1MW gene cluster. BCM patients with either disease-associated large deletion or missense mutations were studied and results compared with those from subjects with other forms of IRD and various degrees of preserved central structure and function. A machine learning technique was used to associate foveal sensitivities and best-corrected visual acuities to foveal structure in IRD patients. Two random forest (RF) models trained on IRD data were applied to predict foveal function in BCM. A curve fitting method was also used and results compared with those of the RF models. The BCM and IRD patients had a comparable range of foveal structure. IRD patients had peak sensitivity at the fovea. Machine learning could successfully predict foveal sensitivity (FS) results from segmented or un-segmented optical coherence tomography (OCT) input. Application of machine learning predictions to BCM at the fovea showed differences between predicted and measured sensitivities, thereby defining treatment potential. The curve fitting method provided similar results. Given a measure of visual acuity (VA) and foveal outer nuclear layer thickness, the question of how many lines of acuity would represent the best efficacious result for each BCM patient could be answered. We propose that foveal vision improvement potential in BCM is predictable from retinal structure using machine learning and curve fitting approaches. This should allow estimates of maximal efficacy in patients being considered for clinical trials and also guide decisions about dosing.
- Published
- 2020
- Full Text
- View/download PDF
14. A 73,128 bp de novo deletion encompassing the OPN1LW/OPN1MW gene cluster in sporadic Blue Cone Monochromacy: a case report
- Author
-
Elena Buena-Atienza, Fadi Nasser, Susanne Kohl, and Bernd Wissinger
- Subjects
De novo mutations ,Sporadic cases ,Blue Cone Monochromacy ,Colour vision deficiency ,Alu-mediated recombination ,Retinal dystrophy ,Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background Blue Cone Monochromacy (BCM) is a rare congenital cone dysfunction disorder with X-linked recessive mode of inheritance. BCM is caused by mutations at the OPN1LW/MW cone opsin gene cluster including deletions of the locus control region (LCR) and/or parts of the gene cluster. We aimed at investigating the clinical presentation, genetic cause and inheritance underlying a sporadic case of BCM. Case presentation We report a 24-year-old male presenting with congenital photophobia, nystagmus and colour vision abnormalities. There was no history of retinal dystrophy in the family. Clinical diagnosis of BCM was supported by genetic investigations of the patient and his family members. Molecular genetic analysis of the OPN1LW/OPN1MW gene cluster revealed a novel deletion of about 73 kb in the patient encompassing the LCR. The deletion was absent in the X-chromosomes of both the mother and transmitting grandfather. Conclusions The present report provides the clinical findings and the genetic basis underlying a sporadic BCM case which is caused by a de novo deletion within the OPN1LW/MW gene cluster originating from the mother’s germline due to Alu-repeat mediated recombination. This is the first report of a de novo deletion resulting in BCM, highlighting the importance to consider BCM and perform genetic testing for this condition in male patients with cone dysfunction also in the absence of a positive family history.
- Published
- 2018
- Full Text
- View/download PDF
15. Abstracts from the 8th International Conference on cGMP Generators, Effectors and Therapeutic Implications
- Author
-
G. Todd Milne, on behalf of the Ironwood team, Peter Sandner, Kathleen A. Lincoln, Paul C. Harrison, Hongxing Chen, Hong Wang, Holly Clifford, Hu Sheng Qian, Diane Wong, Chris Sarko, Ryan Fryer, Jeremy Richman, Glenn A. Reinhart, Carine M. Boustany, Steven S. Pullen, Henriette Andresen, Lise Román Moltzau, Alessandro Cataliotti, Finn Olav Levy, Robert Lukowski, Sandra Frankenreiter, Andreas Friebe, Timothy Calamaras, Robert Baumgartner, Angela McLaughlin, Mark Aronovitz, Wendy Baur, Guang-Rong Wang, Navin Kapur, Richard Karas, Robert Blanton, Stefan Hell, Scott A. Waldman, Jieru E. Lin, Francheska Colon-Gonzalez, Gilbert W. Kim, Erik S. Blomain, Dante Merlino, Adam Snook, Jeanette Erdmann, Jana Wobst, Thorsten Kessler, Heribert Schunkert, Ulrich Walter, Oliver Pagel, Elena Walter, Stepan Gambaryan, Albert Smolenski, Kerstin Jurk, Rene Zahedi, James R. Klinger, Raymond L. Benza, Paul A. Corris, David Langleben, Robert Naeije, Gérald Simonneau, Christian Meier, Pablo Colorado, Mi Kyung Chang, Dennis Busse, Marius M. Hoeper, Jaime L. Masferrer, Sarah Jacobson, Guang Liu, Renee Sarno, Sylvie Bernier, Ping Zhang, Roger Flores-Costa, Mark Currie, Katherine Hall, Dorit Möhrle, Katrin Reimann, Steffen Wolter, Markus Wolters, Evanthia Mergia, Nicole Eichert, Hyun-Soon Geisler, Peter Ruth, Robert Feil, Ulrike Zimmermann, Doris Koesling, Marlies Knipper, Lukas Rüttiger, Yasutake Tanaka, Atsuko Okamoto, Takashi Nojiri, Motofumi Kumazoe, Takeshi Tokudome, Koichi Miura, Jun Hino, Hiroshi Hosoda, Mikiya Miyazato, Kenji Kangawa, Vikas Kapil, Amrita Ahluwalia, Nazareno Paolocci, Philip Eaton, James C. Campbell, Philipp Henning, Eugen Franz, Banumathi Sankaran, Friedrich W. Herberg, Choel Kim, M. Wittwer, Q. Luo, V. Kaila, S. A. Dames, Andrew Tobin, Mahmood Alam, Olena Rudyk, Susanne Krasemann, Kristin Hartmann, Oleksandra Prysyazhna, Min Zhang, Lan Zhao, Astrid Weiss, Ralph Schermuly, Amie J. Moyes, Sandy M. Chu, Reshma S. Baliga, Adrian J. Hobbs, Stylianos Michalakis, Regine Mühlfriedel, Christian Schön, Dominik M. Fischer, Barbara Wilhelm, Ditta Zobor, Susanne Kohl, Tobias Peters, Eberhart Zrenner, Karl Ulrich Bartz-Schmidt, Marius Ueffing, Bernd Wissinger, Mathias Seeliger, Martin Biel, RD-CURE consortium, Mark J. Ranek, Kristen M. Kokkonen, Dong I. Lee, Ronald J. Holewinski, Vineet Agrawal, Cornelia Virus, Donté A. Stevens, Masayuki Sasaki, Huaqun Zhang, Mathew M. Mannion, Peter P. Rainer, Richard C. Page, Jonathan C. Schisler, Jennifer E. Van Eyk, Monte S. Willis, David A. Kass, Manuela Zaccolo, Michael Russwurm, Jan Giesen, Corina Russwurm, Ernst-Martin Füchtbauer, Nadja I. Bork, Viacheslav O. Nikolaev, Luis Agulló, Martin Floor, Jordi Villà-Freixa, Ornella Manfra, Gaia Calamera, Nicoletta C. Surdo, Silja Meier, Alexander Froese, Kjetil Wessel Andressen, Annemarie Aue, Fabian Schwiering, Dieter Groneberg, Gzona Bajraktari, Jürgen Burhenne, Walter E. Haefeli, Johanna Weiss, Katharina Beck, Barbara Voussen, Alexander Vincent, Sean P. Parsons, Jan D. Huizinga, Fabiola Zakia Mónica, Edward Seto, Ferid Murad, Ka Bian, Joseph R. Burgoyne, Daniel Richards, Marianne Bjørnerem, Andrea Hembre Ulsund, Jeong Joo Kim, Sonia Donzelli, Mara Goetz, Kjestine Schmidt, Konstantina Stathopoulou, Jenna Scotcher, Christian Dees, Hariharan Subramanian, Elke Butt, Alisa Kamynina, S. Bruce King, Cor de Witt, Lars I. Leichert, Friederike Cuello, Hyazinth Dobrowinski, Moritz Lehners, Michael Paolillo Hannes Schmidt, Susanne Feil, Lai Wen, Martin Thunemann, Marcus Olbrich, Harald Langer, Meinrad Gawaz, Cor de Wit, Daniela Bertinetti, Hossein-Ardeschir Ghofrani, Friedrich Grimminger, Ekkehard Grünig, Yigao Huang, Pavel Jansa, Zhi Cheng Jing, David Kilpatrick, Stephan Rosenkranz, Flavia Menezes, Arno Fritsch, Sylvia Nikkho, Reiner Frey, Marc Humbert, Manuela Harloff, Joerg Reinders, Jens Schlossmann, Joon Jung, Jessica A. Wales, Cheng-Yu Chen, Linda Breci, Andrzej Weichsel, Sylvie G. Bernier, Robert Solinga, James E. Sheppeck, Paul A. Renhowe, William R. Montfort, Liying Qin, Ying-Ju Sung, Darren Casteel, Alexander Kollau, Andrea Neubauer, Astrid Schrammel, Bernd Mayer, Mika Takai, Chieri Takeuchi, Mai Kadomatsu, Shun Hiroi, Kanako Takamatsu, Hirofumi Tachibana, Marissa Opelt, Emrah Eroglu, Markus Waldeck-Weiermair, Roland Malli, Wolfgang F. Graier, John T. Fassett, Selene J. Sollie, Maria Hernandez-Valladares, Frode Berven, Kjetil W. Andressen, Miki Arai, Yutaka Suzuki, Meinoshin Okumura, Shinpei Kawaoka, Stefanie Peters, Hannes Schmidt, B. Selin Kenet, Sarah Helena Nies, Katharina Frank, Fritz G. Rathjen, Olga N. Petrova, Isabelle Lamarre, Michel Négrerie, Jerid W. Robinson, Jeremy R. Egbert, Julia Davydova, Laurinda A. Jaffe, Lincoln R. Potter, Nicholas Blixt, Leia C. Shuhaibar, Gordon L. Warren, Kim C. Mansky, Simone Romoli, Tobias Bauch, Karoline Dröbner, Frank Eitner, Mihály Ruppert, Tamás Radovits, Sevil Korkmaz-Icöz, Shiliang Li, Péter Hegedűs, Sivakanan Loganathan, Balázs Tamás Németh, Attila Oláh, Csaba Mátyás, Kálmán Benke, Béla Merkely, Matthias Karck, Gábor Szabó, Ulrike Scheib, Matthias Broser, Shatanik Mukherjee, Katja Stehfest, Christine E. Gee, Heinz G. Körschen, Thomas G. Oertner, Peter Hegemann, Deborah M. Dickey, Alexandre Dumoulin, Ralf Kühn, Laurinda Jaffe, Sophie Schobesberger, Peter Wright, Claire Poulet, Catherine Mansfield, Sian E. Harding, Julia Gorelik, Gerald Wölkart, Antonius C. F. Gorren, Gerburg K. Schwaerzer, Darren E. Casteel, Nancy D. Dalton, Yusu Gu, Shunhui Zhuang, Dianna M. Milewicz, Kirk L. Peterson, Renate Pilz, Aikaterini I. Argyriou, Garyfalia Makrynitsa, Ioannis I. Alexandropoulos, Andriana Stamopoulou, Marina Bantzi, Athanassios Giannis, Stavros Topouzis, Andreas Papapetropoulos, Georgios A. Spyroulias, Dennis J. Stuehr, Arnab Ghosh, Yue Dai, Saurav Misra, Boris Tchernychev, Inmaculada Silos-Santiago, Gerhard Hannig, Vu Thao-Vi Dao, Martin Deile, Pavel I. Nedvetsky, Andreas Güldner, César Ibarra-Alvarado, Axel Gödecke, Harald H. H. W. Schmidt, Angelos Vachaviolos, Andrea Gerling, Stefan Z. Lutz, Hans-Ulrich Häring, Marcel A. Krüger, Bernd J. Pichler, Michael J. Shipston, Sara Vandenwijngaert, Clara D. Ledsky, Obiajulu Agha, Dongjian Hu, Ibrahim J. Domian, Emmanuel S. Buys, Christopher Newton-Cheh, Donald B. Bloch, Nadine Mauro, Jonas Keppler, Wilson A. Ferreira, Hanan Chweih, Pamela L. Brito, Camila B. Almeida, Carla F. F. Penteado, Sara S. O. Saad, Fernando F. Costa, Paul S. Frenette, Damian Brockschnieder, Johannes-Peter Stasch, Nicola Conran, Daniel P. Zimmer, Jenny Tobin, Courtney Shea, Kimberly Long, Kim Tang, Peter Germano, James Wakefield, Ali Banijamali, G-Yoon Jamie Im, Albert T. Profy, and Mark G. Currie
- Subjects
Therapeutics. Pharmacology ,RM1-950 ,Toxicology. Poisons ,RA1190-1270 - Published
- 2017
- Full Text
- View/download PDF
16. CDHR1 mutations in retinal dystrophies
- Author
-
Katarina Stingl, Anja K. Mayer, Pablo Llavona, Lejla Mulahasanovic, Günther Rudolph, Samuel G. Jacobson, Eberhart Zrenner, Susanne Kohl, Bernd Wissinger, and Nicole Weisschuh
- Subjects
Medicine ,Science - Abstract
Abstract We report ophthalmic and genetic findings in patients with autosomal recessive retinitis pigmentosa (RP), cone-rod dystrophy (CRD) or cone dystrophy (CD) harboring potential pathogenic variants in the CDHR1 gene. Detailed ophthalmic examination was performed in seven sporadic and six familial subjects. Mutation screening was done using a customized next generation sequencing panel targeting 105 genes implicated in inherited retinal disorders. In one family, homozygosity mapping with subsequent candidate gene analysis was performed. Stringent filtering for rare and potentially disease causing variants following a model of autosomal recessive inheritance led to the identification of eleven different CDHR1 variants in nine index cases. All variants were novel at the time of their identification. In silico analyses confirmed their pathogenic potential. Minigene assays were performed for two non-canonical splice site variants and revealed missplicing for the mutant alleles. Mutations in CDHR1 are a rare cause of retinal dystrophy. Our study further expands the mutational spectrum of this gene and the associated clinical presentation.
- Published
- 2017
- Full Text
- View/download PDF
17. Genome-Wide Association Study Identifies Two Common Loci Associated with Pigment Dispersion Syndrome/Pigmentary Glaucoma and Implicates Myopia in its Development
- Author
-
Mark J. Simcoe, Ameet Shah, Baojian Fan, Hélène Choquet, Nicole Weisschuh, Naushin H. Waseem, Chen Jiang, Ronald B. Melles, Robert Ritch, Omar A. Mahroo, Bernd Wissinger, Eric Jorgenson, Janey L. Wiggs, David F. Garway-Heath, Pirro G. Hysi, and Christopher J. Hammond
- Subjects
Ophthalmology ,Myopia ,Humans ,Polymorphism, Single Nucleotide ,Glaucoma, Open-Angle ,Intraocular Pressure ,Genome-Wide Association Study - Abstract
To identify genetic variants associated with pigment dispersion syndrome (PDS) and pigmentary glaucoma (PG) in unrelated patients and to further understand the genetic and potentially causal relationships between PDS and associated risk factors.A 2-stage genome-wide association meta-analysis with replication and subsequent in silico analyses including Mendelian randomization.A total of 574 cases with PG or PDS and 52 627 controls of European descent.Genome-wide association analyses were performed in 4 cohorts and meta-analyzed in 3 stages: (1) a discovery meta-analysis was performed in 3 cohorts, (2) replication was performed in the fourth cohort, and (3) all 4 cohorts were meta-analyzed to increase statistical power. Two-sample Mendelian randomization was used to determine whether refractive error and intraocular pressure exert causal effects over PDS.The association of genetic variants with PDS and whether myopia exerts causal effects over PDS.Significant association was present at 2 novel loci for PDS/PG. These loci and follow-up analyses implicate the genes gamma secretase activator protein (GSAP) (lead single nucleotide polymorphism [SNP]: rs9641220, P = 6.0×10Common SNPs relating to the GSAP and GRM5/TYR genes are associated risk factors for the development of PDS and PG. Although myopia is a known risk factor, this study uses genetic data to demonstrate that myopia is, in part, a cause of PDS and PG.
- Published
- 2022
- Full Text
- View/download PDF
18. DNAJC30 disease-causing gene variants in a large Central European cohort of patients with suspected Leber’s hereditary optic neuropathy and optic atrophy
- Author
-
Sinja Kieninger, Ting Xiao, Nicole Weisschuh, Susanne Kohl, Klaus Rüther, Peter Michael Kroisel, Tobias Brockmann, Steffi Knappe, Ulrich Kellner, Wolf Lagrèze, Pascale Mazzola, Tobias B Haack, Bernd Wissinger, and Felix Tonagel
- Subjects
Genetics ,Humans ,Optic Atrophy, Hereditary, Leber ,HSP40 Heat-Shock Proteins ,DNA, Mitochondrial ,Genetics (clinical) ,Mitochondria - Abstract
BackgroundLeber’s hereditary optic neuropathy (LHON) has been considered a prototypical mitochondriopathy and a textbook example for maternal inheritance linked to certain disease-causing variants in the mitochondrial genome. Recently, an autosomal recessive form of LHON (arLHON) has been described, caused by disease-causing variants in the nuclear encoded gene DNAJC30.Methods and resultsIn this study, we screened the DNAJC30 gene in a large Central European cohort of patients with a clinical diagnosis of LHON or other autosomal inherited optic atrophies (OA). We identified likely pathogenic variants in 35/1202 patients, corresponding to a detection rate of 2.9%. The previously described missense variant c.152A>G;p.(Tyr51Cys) accounts for 90% of disease-associated alleles in our cohort and we confirmed a strong founder effect. Furthermore, we identified two novel pathogenic variants in DNAJC30: the nonsense variant c.610G>T;p.(Glu204*) and the in-frame deletion c.230_232del;p.(His77del). Clinical investigation of the patients with arLHON revealed a younger age of onset, a more frequent bilateral onset and an increased clinically relevant recovery compared with LHON associated with disease-causing variants in the mitochondrial DNA.ConclusionThis study expands previous findings on arLHON and emphasises the importance of DNAJC30 in the genetic diagnostics of LHON and OA in European patients.
- Published
- 2022
- Full Text
- View/download PDF
19. Autosomal dominant optic atrophy: A novel treatment for OPA1 splice defects using U1 snRNA adaption
- Author
-
Bernd Wissinger, Christoph Jüschke, Thomas Klopstock, Marta Owczarek-Lipska, John Neidhardt, Claudia B. Catarino, and Faculteit Medische Wetenschappen/UMCG
- Subjects
EXPRESSION ,endocrine system ,ExSpeU1 ,U1 snRNA ,ISOFORMS ,DOA ,RM1-950 ,Biology ,GENE THERAPEUTIC APPROACH ,medicine.disease_cause ,OPA1 ,DISEASE ,ADOA ,splicing ,Exon ,Drug Discovery ,Optic Atrophy Type 1 ,medicine ,splice ,STRATEGY ,ddc:610 ,Dominant Optic Atrophy ,MUTATION ,Gene ,Genetics ,SPECTRUM ,Mutation ,Intron ,SMALL NUCLEAR-RNA ,medicine.disease ,CORRECT ,gene therapy ,eye diseases ,LONG ,RNA splicing ,Molecular Medicine ,Optic Atrophy 1 ,Therapeutics. Pharmacology ,Haploinsufficiency ,Autosomal Dominant Optic Atrophy - Abstract
Autosomal dominant optic atrophy (ADOA) is frequently caused by mutations in the optic atrophy 1 (OPA1) gene, with haploinsufficiency being the major genetic pathomechanism. Almost 30% of the OPA1-associated cases suffer from splice defects. We identified a novel OPA1 mutation, c.1065+5G>A, in patients with ADOA. In patient-derived fibroblasts, the mutation led to skipping of OPA1 exon 10, reducing the OPA1 protein expression by approximately 50%. We developed a molecular treatment to correct the splice defect in OPA1 using engineered U1 splice factors retargeted to different locations in OPA1 exon 10 or intron 10. The strongest therapeutic effect was detected when U1 binding was engineered to bind to intron 10 at position +18, a position predicted by bioinformatics to be a promising binding site. We were able to significantly silence the effect of the mutation (skipping of exon 10) and simultaneously increase the expression level of normal transcripts. Retargeting U1 to the canonical splice donor site did not lead to a detectable splice correction. This proof-of-concept study indicates for the first time the feasibility of splice mutation correction as a treatment option for ADOA. Increasing the amount of correctly spliced OPA1 transcripts may suffice to overcome the haploinsufficiency.
- Published
- 2021
- Full Text
- View/download PDF
20. KCNV2-Associated Retinopathy: Detailed Retinal Phenotype and Structural Endpoints—KCNV2 Study Group Report 2
- Author
-
Kazushige Tsunoda, Camiel J. F. Boon, Ester Carreño, Xiao Liu, Rachel M. Huckfeldt, Mark E. Pennesi, Andrew R. Webster, Anthony G. Robson, Elise Héon, Gavin Arno, Susanne Kohl, Belen Jimenez-Rolando, Michel Michaelides, Carmen Ayuso, Omar A. Mahroo, Eyal Banin, Samer Khateb, Takaaki Hayashi, Bernd Wissinger, Arif O. Khan, Eberhart Zrenner, Alberta A H J Thiadens, Ajoy Vincent, Nikolas Pontikos, Maria Inmaculada Martin-Merida, Thales Antonio Cabral de Guimaraes, Xuan-Thanh-An Nguyen, Michalis Georgiou, Almudena Avila-Fernandez, Mauricio E Vargas, Emanuel R. de Carvalho, Shaun Michael Leo, Yu Fujinami-Yokokawa, Dror Sharon, Fadi Nasser, Kaoru Fujinami, Blanca Garcia-Sandoval, Ophthalmology, and ANS - Complex Trait Genetics
- Subjects
medicine.medical_specialty ,genetic structures ,Fundus Oculi ,Retina ,Foveola ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Retinal Diseases ,Optical coherence tomography ,Ophthalmology ,medicine ,Humans ,Fluorescein Angiography ,Outer nuclear layer ,Retrospective Studies ,030304 developmental biology ,0303 health sciences ,Retinal pigment epithelium ,medicine.diagnostic_test ,business.industry ,Retinal ,medicine.disease ,eye diseases ,Autofluorescence ,Phenotype ,medicine.anatomical_structure ,chemistry ,Potassium Channels, Voltage-Gated ,030221 ophthalmology & optometry ,Original Article ,sense organs ,business ,Tomography, Optical Coherence ,Retinopathy - Abstract
Highlights • KCNV2-associated retinopathy is a slowly progressive disease with early retinal changes, which are predominantly symmetric between eyes. • Disease course can be unpredictable and may severely affect children and young adults. • Findings suggest a potential window for intervention until 40 years of age, albeit with variability between patients due to macular atrophy., Purpose To describe the detailed retinal phenotype of KCNV2-associated retinopathy. Study design Multicenter international retrospective case series. Methods Review of retinal imaging including fundus autofluorescence (FAF) and optical coherence tomography (OCT), including qualitative and quantitative analyses. Results Three distinct macular FAF features were identified: (1) centrally increased signal (n = 35, 41.7%), (2) decreased autofluorescence (n = 27, 31.1%), and (3) ring of increased signal (n = 37, 44.0%). Five distinct FAF groups were identified based on combinations of those features, with 23.5% of patients changing the FAF group over a mean (range) follow-up of 5.9 years (1.9-13.1 years). Qualitative assessment was performed by grading OCT into 5 grades: (1) continuous ellipsoid zone (EZ) (20.5%); (2) EZ disruption (26.1%); (3) EZ absence, without optical gap and with preserved retinal pigment epithelium complex (21.6%); (4) loss of EZ and a hyporeflective zone at the foveola (6.8%); and (5) outer retina and retinal pigment epithelium complex loss (25.0%). Eighty-six patients had scans available from both eyes, with 83 (96.5%) having the same grade in both eyes, and 36.1% changed OCT grade over a mean follow-up of 5.5 years. The annual rate of outer nuclear layer thickness change was similar for right and left eyes. Conclusions KCNV2-associated retinopathy is a slowly progressive disease with early retinal changes, which are predominantly symmetric between eyes. The identification of a single OCT or FAF measurement as an endpoint to determine progression that applies to all patients may be challenging, although outer nuclear layer thickness is a potential biomarker. Findings suggest a potential window for intervention until 40 years of age.
- Published
- 2021
21. Molecular and clinical analysis of 27 German patients with Leber congenital amaurosis.
- Author
-
Nicole Weisschuh, Britta Feldhaus, Muhammad Imran Khan, Frans P M Cremers, Susanne Kohl, Bernd Wissinger, and Ditta Zobor
- Subjects
Medicine ,Science - Abstract
Leber congenital amaurosis (LCA) is the earliest and most severe form of all inherited retinal dystrophies (IRD) and the most frequent cause of inherited blindness in children. The phenotypic overlap with other early-onset and severe IRDs as well as difficulties associated with the ophthalmic examination of infants can complicate the clinical diagnosis. To date, 25 genes have been implicated in the pathogenesis of LCA. The disorder is usually inherited in an autosomal recessive fashion, although rare dominant cases have been reported. We report the mutation spectra and frequency of genes in 27 German index patients initially diagnosed with LCA. A total of 108 LCA- and other genes implicated in IRD were analysed using a cost-effective targeted next-generation sequencing procedure based on molecular inversion probes (MIPs). Sequencing and variant filtering led to the identification of putative pathogenic variants in 25 cases, thereby leading to a detection rate of 93%. The mutation spectrum comprises 34 different alleles, 17 of which are novel. In line with previous studies, the genetic results led to a revision of the initial clinical diagnosis in a substantial proportion of cases, demonstrating the importance of genetic testing in IRD. In addition, our detection rate of 93% shows that MIPs are a cost-efficient and sensitive tool for targeted next-generation sequencing in IRD.
- Published
- 2018
- Full Text
- View/download PDF
22. Visual and ocular findings in a family with X-linked cone dysfunction and protanopia
- Author
-
Dag Holmquist, Bernd Wissinger, Jürg Hengstler, David Epstein, Susanne Kohl, Kristina Tear-Fahnehjelm, and Monica Olsson
- Subjects
Male ,medicine.medical_specialty ,Visual acuity ,Adolescent ,genetic structures ,Color vision ,Visual Acuity ,Color Vision Defects ,Amblyopia ,Retina ,Young Adult ,Exon ,chemistry.chemical_compound ,Sickness Impact Profile ,Ophthalmology ,Electroretinography ,Myopia ,medicine ,Humans ,Genetics (clinical) ,Color Perception Tests ,medicine.diagnostic_test ,business.industry ,Rod Opsins ,Genetic Diseases, X-Linked ,Retinal ,Exons ,eye diseases ,Phenotype ,chemistry ,OPN1LW ,Myopia, Degenerative ,Pediatrics, Perinatology and Child Health ,sense organs ,Visual Fields ,medicine.symptom ,Protanopia ,business ,Erg ,Color Perception ,Tomography, Optical Coherence - Abstract
Background: Bornholm eye disease (BED) is a rare X-linked cone dysfunction disorder with high myopia, amblyopia, and color vision defects.Materials and methods: Visual and ocular outcomes in a family where two of five siblings had molecularly confirmed BED are reported. Ophthalmological assessments included best-corrected visual acuity (BCVA), color vision test, and optical coherence tomography (OCT). Medical records, electroretinography (ERG), and genetic analyses were re-evaluated.Results: Two male siblings had confirmed BED with myopia and protanopia. The younger brother had high myopia, subnormal BCVA, and ocular fundi that showed tilted discs, crescent shaped peripapillary atrophy, and visible choroidal vessels. OCT confirmed retinal and choroidal atrophy. The older brother was lightly myopic with normal/subnormal BCVA and subtle findings in the fundi. Both brothers had abnormal ERG recordings with a decreased cone response. They also had a structurally intact OPN1LW/OPN1MW gene cluster. The OPN1LW gene was shown to carry a deleterious variant combination in exon 3 known to result in mis-splicing of opsin mRNA and acknowledged as LIAVA amino acid delineation (Leu153-Ile171-Ala174-Val178-Ala180), while the OPN1MW gene exon 3 showed a non-pathogenic variant combination (MVVVA). Another normal-sighted brother carried another wildtype variant combination (LVAIS) in exon 3 of the OPN1LW gene.Conclusions: The two affected brothers demonstrated a large variability in their phenotypes even though the genotypes were identical. They presented a disease-associated haplotype in exon 3 of OPN1LW that has been described as the molecular cause of BED.
- Published
- 2021
- Full Text
- View/download PDF
23. KCNV2-Associated Retinopathy
- Author
-
Bernd Wissinger, Eberhart Zrenner, Nikolas Pontikos, Maria Inmaculada Martin-Merida, Xuan-Thanh-An Nguyen, Anthony G. Robson, Emanuel R. de Carvalho, Kazushige Tsunoda, Omar A. Mahroo, Alberta A H J Thiadens, Mauricio E Vargas, Fadi Nasser, Kaoru Fujinami, Gavin Arno, Rachel M. Huckfeldt, Ester Carreño, Thales Antonio Cabral de Guimaraes, Ayuso Carmen, Takaaki Hayashi, Michel Michaelides, Elise Héon, Xiao Liu, Dror Sharon, Ajoy Vincent, Mark E. Pennesi, Michalis Georgiou, Arif O. Khan, Andrew R. Webster, Yu Fujinami-Yokokawa, Gema Gordo, Eyal Banin, Shaun Michael Leo, Susanne Kohl, Belen Jimenez-Rolando, Camiel J. F. Boon, Samer Khateb, Ophthalmology, and ANS - Complex Trait Genetics
- Subjects
Male ,Visual acuity ,Photophobia ,genetic structures ,Visual Acuity ,0302 clinical medicine ,Child ,Genetics ,0303 health sciences ,medicine.diagnostic_test ,High-Throughput Nucleotide Sequencing ,Middle Aged ,Phenotype ,Potassium Channels, Voltage-Gated ,Child, Preschool ,Decreased Visual Acuity ,Cohort ,Female ,Original Article ,medicine.symptom ,Erg ,Retinitis Pigmentosa ,Tomography, Optical Coherence ,Retinopathy ,Adult ,Adolescent ,Vision Disorders ,Dark Adaptation ,Refraction, Ocular ,Nyctalopia ,Retina ,03 medical and health sciences ,Exome Sequencing ,medicine ,Electroretinography ,Humans ,Molecular Biology ,Alleles ,030304 developmental biology ,Aged ,Retrospective Studies ,Whole Genome Sequencing ,business.industry ,Infant, Newborn ,Infant ,medicine.disease ,eye diseases ,Ophthalmology ,030221 ophthalmology & optometry ,business - Abstract
Purpose To investigate genetics, electrophysiology, and clinical course of KCNV2-associated retinopathy in a cohort of children and adults. Study design This was a multicenter international clinical cohort study. Methods Review of clinical notes and molecular genetic testing. Full-field electroretinography (ERG) recordings, incorporating the international standards, were reviewed and quantified and compared with age and recordings from control subjects. Results In total, 230 disease-associated alleles were identified from 117 patients, corresponding to 75 different KCNV2 variants, with 28 being novel. The mean age of onset was 3.9 years old. All patients were symptomatic before 12 years of age (range, 0-11 years). Decreased visual acuity was present in all patients, and 4 other symptoms were common: reduced color vision (78.6%), photophobia (53.5%), nyctalopia (43.6%), and nystagmus (38.6%). After a mean follow-up of 8.4 years, the mean best-corrected visual acuity (BCVA ± SD) decreased from 0.81 ± 0.27 to 0.90 ± 0.31 logarithm of minimal angle of resolution. Full-field ERGs showed pathognomonic waveform features. Quantitative assessment revealed a wide range of ERG amplitudes and peak times, with a mean rate of age-associated reduction indistinguishable from the control group. Mean amplitude reductions for the dark-adapted 0.01 ERG, dark-adapted 10 ERG a-wave, and LA 3.0 30 Hz and LA3 ERG b-waves were 55%, 21%, 48%, and 74%, respectively compared with control values. Peak times showed stability across 6 decades. Conclusion In KCNV2-associated retinopathy, full-field ERGs are diagnostic and consistent with largely stable peripheral retinal dysfunction. Report 1 highlights the severity of the clinical phenotype and established a large cohort of patients, emphasizing the unmet need for trials of novel therapeutics., Highlights • The current study established the largest and most characterized cohort of molecularly confirmed patients with KCNV2-associated retinopathy. • Report 1 highlights the genetic background, evidence of electroretinography stability over a broad age range, and the severe phenotype of the disease.
- Published
- 2021
24. The landscape of submicroscopic structural variants at the
- Author
-
Bernd, Wissinger, Britta, Baumann, Elena, Buena-Atienza, Zeinab, Ravesh, Artur V, Cideciyan, Katarina, Stingl, Isabelle, Audo, Isabelle, Meunier, Beatrice, Bocquet, Elias I, Traboulsi, Alison J, Hardcastle, Jessica C, Gardner, Michel, Michaelides, Kari E, Branham, Thomas, Rosenberg, Sten, Andreasson, Hélène, Dollfus, David, Birch, Andrea L, Vincent, Loreto, Martorell, Jaume, Català Mora, Ulrich, Kellner, Klaus, Rüther, Birgit, Lorenz, Markus N, Preising, Emanuela, Manfredini, Yuri A, Zarate, Raymon, Vijzelaar, Eberhart, Zrenner, Samuel G, Jacobson, and Susanne, Kohl
- Subjects
Multigene Family ,Retinal Cone Photoreceptor Cells ,Rod Opsins ,Humans ,Color Vision Defects ,Gene Deletion - Abstract
Blue cone monochromacy (BCM) is an X-linked retinal disorder characterized by low vision, photoaversion, and poor color discrimination. BCM is due to the lack of long-wavelength-sensitive and middle-wavelength-sensitive cone photoreceptor function and caused by mutations in the
- Published
- 2022
25. Adaptive optics ophthalmoscopy in retinitis pigmentosa ( <scp>RP</scp> ): Typical patterns
- Author
-
Friederike C. Kortuem, Melanie Kempf, Laura Kuehlewein, Fadi Nasser, Constanze Kortuem, Michel Paques, Susanne Kohl, Marius Ueffing, Bernd Wissinger, Eberhart Zrenner, and Katarina Stingl
- Subjects
Ophthalmoscopy ,Ophthalmology ,Humans ,General Medicine ,Retinitis Pigmentosa - Published
- 2022
- Full Text
- View/download PDF
26. Central Visual Function and Genotype-Phenotype Correlations in PDE6A-Associated Retinitis Pigmentosa
- Author
-
Laura, Kuehlewein, Torsten, Straßer, Gunnar, Blumenstock, Katarina, Stingl, M Dominik, Fischer, Barbara, Wilhelm, Eberhart, Zrenner, Bernd, Wissinger, Susanne, Kohl, Nicole, Weisschuh, and Ditta, Zobor
- Subjects
Male ,Cyclic Nucleotide Phosphodiesterases, Type 6 ,Mutation ,Visual Acuity ,Humans ,Female ,Eye Proteins ,Genetic Association Studies ,Retinitis Pigmentosa ,Pedigree - Abstract
Autosomal recessive retinitis pigmentosa (arRP) can be caused by mutations in the phosphodiesterase 6A (PDE6A) gene. Here, we describe the natural course of disease progression with respect to central retinal function (i.e., visual acuity, contrast sensitivity, and color vision) and establish a detailed genotype--phenotype correlation.Forty-four patients (26 females; mean age ± SD, 43 ± 13 years) with a confirmed genetic diagnosis of PDE6A-associated arRP underwent comprehensive ophthalmological examinations including best-corrected visual acuity (BCVA) with Early Treatment Diabetic Retinopathy Study charts, contrast sensitivity (CS) with Pelli-Robson charts at distances of 3 m and 1 m, and color vision testing using Roth 28-Hue and Panel D-15 saturated color cups.The most frequently observed variants were c.998+1GA/p.?, c.304CA/p.R102S, and c.2053GA/p.V685M. Central retinal function in patients homozygous for variant c.304CA/p.R102S was better when compared to patients homozygous for variant c.998+1GA/p.?, although the former were older at baseline. Central retinal function was similar in patients homozygous for variant c.304CA/p.R102S and patients heterozygous for variants c.304CA/p.R102S and c.2053GA/p.V685M, although the latter were younger at baseline. Annual decline rates in central retinal function were small.We conclude that the severity of the different disease-causing PDE6A mutations in humans with respect to central visual function may be ranked as follows: c.2053GA/p.V685M in homozygous state (most severe)c.998+1GA/p.? in homozygous statec.304CA/p.R102S and c.2053GA/p.V685M in compound-heterozygous statec.304CA/p.R102S in homozygous state (mildest). The assessment of treatment efficacy in interventional trials will remain challenging due to small annual decline rates in central retinal function.
- Published
- 2022
27. Spatial and temporal resolution of the photoreceptors rescue dynamics after treatment with voretigene neparvovec
- Author
-
Fabian Wozar, Krunoslav Stingl, Ronja Jung, Friederike Kortüm, Bernd Wissinger, Eberhart Zrenner, Felix F L Reichel, Tobias Peters, Fadi Nasser, Carina Kelbsch, Karl U. Bartz-Schmidt, Katarina Stingl, Barbara Wilhelm, Spyridon Dimopoulos, Susanne Kohl, M. Dominik Fischer, and Melanie Kempf
- Subjects
Male ,medicine.medical_specialty ,Visual acuity ,genetic structures ,Visual Acuity ,Retina ,Pupil ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Ophthalmology ,Retinal Dystrophies ,Pupillary response ,Humans ,Medicine ,Scotopic vision ,Chromatic scale ,business.industry ,Retinal ,Sensory Systems ,medicine.anatomical_structure ,chemistry ,Visual Field Tests ,Female ,sense organs ,medicine.symptom ,business ,Tomography, Optical Coherence ,Photopic vision - Abstract
BackgroundVoretigene neparvovec is a gene therapeutic agent for treatment of retinal dystrophies caused by bi-allelic RPE65 mutations. In this study, we report on a novel and objective evaluation of a retinotopic photoreceptor rescue.MethodsSeven eyes of five patients (14, 21, 23, 24, 36 years, 1 male, 4 females) with bi-allelic RPE65 mutations have been treated with voretigene neparvovec. The clinical examinations included visual acuity testing, dark-adapted full-field stimulus threshold (FST), dark-adapted chromatic perimeter (DAC) with a 30-degree grid, and a 30 degrees grid scotopic and photopic chromatic pupil campimetry (CPC). All evaluations and spectral domain optical coherence tomography were performed at baseline, 1 month and 3 months.ResultsAll except the oldest patient had a measurable improvement of the rod function assessed via FST, DAC or scotopic CPC at 1 month. The visual acuity improved slightly or remained stable in all eyes. A cone function improvement as measured by photopic CPC was observed in three eyes. The gain of the dark-adapted threshold with blue FST and the DAC stimuli (cyan) average correlated strongly with age (R2>0.7). The pupil response improvement in the scotopic CPC correlated with the baseline local retinal volume (R2=0.5).ConclusionsThe presented protocols allow evaluating the individual spatial and temporal effects of gene therapy effects. Additionally, we explored parameters that correlated with the success of the therapy. CPC and DAC present new and fast ways to assess functional changes in retinotopic maps of rod and cone function, measuring complementary aspects of retinal function.
- Published
- 2021
- Full Text
- View/download PDF
28. Biallelic Loss-of-Function NDUFA12 Variants Cause a Wide Phenotypic Spectrum from Leigh/Leigh-Like Syndrome to Isolated Optic Atrophy
- Author
-
Francesca Magrinelli, Elisa Cali, Vinícius Lopes Braga, Uluç Yis, Hoda Tomoum, Hanan Shamseldin, Julian Raiman, Christoph Kernstock, Flávio Moura Rezende Filho, Orlando Graziani Povoas Barsottini, Robert W. Taylor, Elsebet Østergaard, Abdullah Tamim, Karin Schäferhoff, Juliana Maria Ferraz Sallum, Maha S. Zaki, Fernando Kok, Kailash P. Bhatia, Bernd Wissinger, Kate Sergeant, Tobias B. Haack, Rita Horvath, Semra Hiz, Fowzan S. Alkuraya, Henry Houlden, José Luiz Pedroso, and Reza Maroofian
- Subjects
NDUFA12 ,Neurology ,optic atrophy ,Neurology (clinical) ,dystonia ,Leigh syndrome ,phenotypic heterogeneity - Abstract
Background Biallelic loss-of-function NDUFA12 variants have hitherto been linked to mitochondrial complex I deficiency presenting with heterogeneous clinical and radiological features in nine cases only. Objectives To fully characterize, both phenotypically and genotypically, NDUFA12-related mitochondrial disease. Methods We collected data from cases identified by screening genetic databases of several laboratories worldwide and systematically reviewed the literature. Results Nine unreported NDUFA12 cases from six pedigrees were identified, with presentation ranging from movement disorder phenotypes (dystonia and/or spasticity) to isolated optic atrophy. MRI showed basal ganglia abnormalities (n = 6), optic atrophy (n = 2), or was unremarkable (n = 1). All carried homozygous truncating NDUFA12 variants, three of which are novel. Conclusions Our case series expands phenotype-genotype correlations in NDUFA12-associated mitochondrial disease, providing evidence of intra- and inter-familial clinical heterogeneity for the same variant. It confirms NDUFA12 variants should be included in the diagnostic workup of Leigh/Leigh-like syndromes - particularly with dystonia - as well as isolated optic atrophy.
- Published
- 2022
- Full Text
- View/download PDF
29. Comprehensive variant spectrum of the CNGA3 gene in patients affected by achromatopsia
- Author
-
Maria Solaki, Britta Baumann, Peggy Reuter, Sten Andreasson, Isabelle Audo, Carmen Ayuso, Ghassan Balousha, Francesco Benedicenti, David Birch, Pierre Bitoun, Delphine Blain, Beatrice Bocquet, Kari Branham, Jaume Català‐Mora, Elfride De Baere, Helene Dollfus, Mohammed Falana, Roberto Giorda, Irina Golovleva, Irene Gottlob, John R. Heckenlively, Samuel G. Jacobson, Kaylie Jones, Herbert Jägle, Andreas R. Janecke, Ulrich Kellner, Petra Liskova, Birgit Lorenz, Loreto Martorell‐Sampol, André Messias, Isabelle Meunier, Fernanda Belga Ottoni Porto, Eleni Papageorgiou, Astrid S. Plomp, Thomy J. L. de Ravel, Charlotte M. Reiff, Agnes B. Renner, Thomas Rosenberg, Günther Rudolph, Roberto Salati, E. Cumhur Sener, Paul A. Sieving, Franco Stanzial, Elias I. Traboulsi, Stephen H. Tsang, Balázs Varsanyi, Richard G. Weleber, Ditta Zobor, Katarina Stingl, Bernd Wissinger, Susanne Kohl, Human genetics, Amsterdam Reproduction & Development (AR&D), Clinical sciences, and Medical Genetics
- Subjects
NUCLEOTIDE-GATED CHANNELS ,JAPANESE ,analysis ,Cyclic Nucleotide-Gated Cation Channels ,Color Vision Defects ,TOTAL COLOURBLINDNESS ,PATIENT ,MOLECULAR-GENETICS ,variant spectrum ,Medicine and Health Sciences ,Genetics ,in silico analysis ,Humans ,NONSENSE MUTATION ,PAKISTANI FAMILIES ,Color Vision Defects/genetics ,variant classification ,Genetics (clinical) ,Medicinsk genetik ,FUNCTIONAL-ANALYSIS ,UNFOLDED PROTEIN RESPONSE ,CNGA3 ,PHOTORECEPTOR DEGENERATION ,Cyclic Nucleotide-Gated Cation Channels/genetics ,in silico ,cyclic nucleotide-gated ion channel ,Mutation ,Retinal Cone Photoreceptor Cells ,achromatopsia ,ALPHA-SUBUNIT ,Medical Genetics - Abstract
Achromatopsia (ACHM) is a congenital cone photoreceptor disorder characterized by impaired color discrimination, low visual acuity, photosensitivity, and nystagmus. To date, six genes have been associated with ACHM (CNGA3, CNGB3, GNAT2, PDE6C, PDE6H, and ATF6), the majority of these being implicated in the cone phototransduction cascade. CNGA3 encodes the CNGA3 subunit of the cyclic nucleotide-gated ion channel in cone photoreceptors and is one of the major disease-associated genes for ACHM. Herein, we provide a comprehensive overview of the CNGA3 variant spectrum in a cohort of 1060 genetically confirmed ACHM patients, 385 (36.3%) of these carrying "likely disease-causing" variants in CNGA3. Compiling our own genetic data with those reported in the literature and in public databases, we further extend the CNGA3 variant spectrum to a total of 316 variants, 244 of which we interpreted as "likely disease-causing" according to ACMG/AMP criteria. We report 48 novel "likely disease-causing" variants, 24 of which are missense substitutions underlining the predominant role of this mutation class in the CNGA3 variant spectrum. In addition, we provide extensive in silico analyses and summarize reported functional data of previously analyzed missense, nonsense and splicing variants to further advance the pathogenicity assessment of the identified variants.
- Published
- 2022
- Full Text
- View/download PDF
30. Antisense Oligonucleotide Mediated Splice Correction of a Deep Intronic Mutation in OPA1
- Author
-
Tobias Bonifert, Irene Gonzalez Menendez, Florian Battke, Yvonne Theurer, Matthis Synofzik, Ludger Schöls, and Bernd Wissinger
- Subjects
antisense oligonucleotides ,deep intronic mutation ,OPA1 ,optic neuropathies ,splice correction ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Inherited optic neuropathies (ION) present an important cause of blindness in the European working-age population. Recently we reported the discovery of four independent families with deep intronic mutations in the main inherited optic neuropathies gene OPA1. These deep intronic mutations cause mis-splicing of the OPA1 pre-messenger-RNA transcripts by creating cryptic acceptor splice sites. As a rescue strategy we sought to prevent mis-splicing of the mutant pre-messenger-RNA by applying 2′O-methyl-antisense oligonucleotides (AONs) with a full-length phosphorothioate backbone that target the cryptic acceptor splice sites and the predicted novel branch point created by the deep intronic mutations, respectively. Transfection of patient-derived primary fibroblasts with these AONs induced correct splicing of the mutant pre-messenger-RNA in a time and concentration dependent mode of action, as detected by pyrosequencing of informative heterozygous variants. The treatment showed strong rescue effects (≃55%) using the cryptic acceptor splice sites targeting AON and moderate rescue (≃16%) using the branch point targeting AON. The highest efficacy of Splice correction could be observed 4 days after treatment however, significant effects were still seen 14 days post-transfection. Western blot analysis revealed increased amounts of OPA1 protein with maximum amounts at ≃3 days post-treatment. In summary, we provide the first mutation-specific in vitro rescue strategy for OPA1 deficiency using synthetic AONs.
- Published
- 2016
- Full Text
- View/download PDF
31. Mutation Detection in Patients with Retinal Dystrophies Using Targeted Next Generation Sequencing.
- Author
-
Nicole Weisschuh, Anja K Mayer, Tim M Strom, Susanne Kohl, Nicola Glöckle, Max Schubach, Sten Andreasson, Antje Bernd, David G Birch, Christian P Hamel, John R Heckenlively, Samuel G Jacobson, Christina Kamme, Ulrich Kellner, Erdmute Kunstmann, Pietro Maffei, Charlotte M Reiff, Klaus Rohrschneider, Thomas Rosenberg, Günther Rudolph, Rita Vámos, Balázs Varsányi, Richard G Weleber, and Bernd Wissinger
- Subjects
Medicine ,Science - Abstract
Retinal dystrophies (RD) constitute a group of blinding diseases that are characterized by clinical variability and pronounced genetic heterogeneity. The different nonsyndromic and syndromic forms of RD can be attributed to mutations in more than 200 genes. Consequently, next generation sequencing (NGS) technologies are among the most promising approaches to identify mutations in RD. We screened a large cohort of patients comprising 89 independent cases and families with various subforms of RD applying different NGS platforms. While mutation screening in 50 cases was performed using a RD gene capture panel, 47 cases were analyzed using whole exome sequencing. One family was analyzed using whole genome sequencing. A detection rate of 61% was achieved including mutations in 34 known and two novel RD genes. A total of 69 distinct mutations were identified, including 39 novel mutations. Notably, genetic findings in several families were not consistent with the initial clinical diagnosis. Clinical reassessment resulted in refinement of the clinical diagnosis in some of these families and confirmed the broad clinical spectrum associated with mutations in RD genes.
- Published
- 2016
- Full Text
- View/download PDF
32. Oscillatory Potentials in Achromatopsia as a Tool for Understanding Cone Retinal Functions
- Author
-
Stingl, Giulia Righetti, Melanie Kempf, Christoph Braun, Ronja Jung, Susanne Kohl, Bernd Wissinger, Eberhart Zrenner, Katarina Stingl, and Krunoslav
- Subjects
genetic structures ,sense organs ,ERG ,achromatopsia ,oscillatory potentials ,cone functions ,Morlet wavelet transform ,time-frequency analysis - Abstract
Achromatopsia (ACHM) is an inherited autosomal recessive disease lacking cone photoreceptors functions. In this study, we characterize the time-frequency representation of the full-field electroretinogram (ffERG) component oscillatory potentials (OPs), to investigate the connections between photoreceptors and the inner retinal network using ACHM as a model. Time-frequency characterization of OPs was extracted from 52 controls and 41 achromat individuals. The stimulation via ffERG was delivered under dark-adaptation (DA, 3.0 and 10.0 cd·s·m−2) to assess mixed rod-cone responses. The ffERG signal was subsequently analyzed using a continuous complex Morlet transform. Time-frequency maps of both DA conditions show the characterization of OPs, disclosing in both groups two distinct time-frequency windows (~70–100 Hz and >100 Hz) within 50 ms. Our main result indicates a significant cluster (p < 0.05) in both conditions of reduced relative power (dB) in ACHM people compared to controls, mainly at the time-frequency window >100 Hz. These results suggest that the strongly reduced but not absent activity of OPs above 100 Hz is mostly driven by cones and only in small part by rods. Thus, the lack of cone modulation of OPs gives important insights into interactions between photoreceptors and the inner retinal network and can be used as a biomarker for monitoring cone connection to the inner retina.
- Published
- 2021
- Full Text
- View/download PDF
33. Dominant
- Author
-
Majida, Charif, Naïg, Gueguen, Marc, Ferré, Zouhair, Elkarhat, Salim, Khiati, Morgane, LeMao, Arnaud, Chevrollier, Valerie, Desquiret-Dumas, David, Goudenège, Céline, Bris, Selma, Kane, Jennifer, Alban, Stéphanie, Chupin, Céline, Wetterwald, Leonardo, Caporali, Francesca, Tagliavini, Chiara, LaMorgia, Michele, Carbonelli, Neringa, Jurkute, Abdelhamid, Barakat, Philippe, Gohier, Christophe, Verny, Magalie, Barth, Vincent, Procaccio, Dominique, Bonneau, Xavier, Zanlonghi, Isabelle, Meunier, Nicole, Weisschuh, Simone, Schimpf-Linzenbold, Felix, Tonagel, Ulrich, Kellner, Patrick, Yu-Wai-Man, Valerio, Carelli, Bernd, Wissinger, Patrizia, Amati-Bonneau, Pascal, Reynier, and Guy, Lenaers
- Subjects
mitochondria ,AcademicSubjects/SCI01870 ,Original Article ,AcademicSubjects/MED00310 ,Krebs cycle ,eye diseases ,ACO2 ,aconitase 2 ,optic neuropathy - Abstract
Biallelic mutations in ACO2, encoding the mitochondrial aconitase 2, have been identified in individuals with neurodegenerative syndromes, including infantile cerebellar retinal degeneration and recessive optic neuropathies (locus OPA9). By screening European cohorts of individuals with genetically unsolved inherited optic neuropathies, we identified 61 cases harbouring variants in ACO2, among whom 50 carried dominant mutations, emphasizing for the first time the important contribution of ACO2 monoallelic pathogenic variants to dominant optic atrophy. Analysis of the ophthalmological and clinical data revealed that recessive cases are affected more severely than dominant cases, while not significantly earlier. In addition, 27% of the recessive cases and 11% of the dominant cases manifested with extraocular features in addition to optic atrophy. In silico analyses of ACO2 variants predicted their deleterious impacts on ACO2 biophysical properties. Skin derived fibroblasts from patients harbouring dominant and recessive ACO2 mutations revealed a reduction of ACO2 abundance and enzymatic activity, and the impairment of the mitochondrial respiration using citrate and pyruvate as substrates, while the addition of other Krebs cycle intermediates restored a normal respiration, suggesting a possible short-cut adaptation of the tricarboxylic citric acid cycle. Analysis of the mitochondrial genome abundance disclosed a significant reduction of the mitochondrial DNA amount in all ACO2 fibroblasts. Overall, our data position ACO2 as the third most frequently mutated gene in autosomal inherited optic neuropathies, after OPA1 and WFS1, and emphasize the crucial involvement of the first steps of the Krebs cycle in the maintenance and survival of retinal ganglion cells., Graphical Abstract Graphical Abstract, By screening European cohorts of individuals with genetically unsolved inherited optic neuropathies, Charif et al. report 61 new cases harbouring variants in ACO2, among whom 50 with dominant mutations, emphasizing for the first time the important contribution of ACO2 monoallelic pathogenic variants to dominant optic atrophy.
- Published
- 2021
34. Color Vision in Blue Cone Monochromacy: Outcome Measures for a Clinical Trial
- Author
-
Abraham A. Mascio, Alejandro J. Roman, Artur V. Cideciyan, Rebecca Sheplock, Vivian Wu, Alexandra V. Garafalo, Alexander Sumaroka, Sydney Pirkle, Susanne Kohl, Bernd Wissinger, Samuel G. Jacobson, and John L. Barbur
- Subjects
Ophthalmology ,Biomedical Engineering - Published
- 2023
- Full Text
- View/download PDF
35. Biallelic Loss-of-Function
- Author
-
Francesca, Magrinelli, Elisa, Cali, Vinícius Lopes, Braga, Uluç, Yis, Hoda, Tomoum, Hanan, Shamseldin, Julian, Raiman, Christoph, Kernstock, Flávio Moura, Rezende Filho, Orlando Graziani Povoas, Barsottini, Robert W, Taylor, Elsebet, Østergaard, Abdullah, Tamim, Karin, Schäferhoff, Juliana Maria Ferraz, Sallum, Maha S, Zaki, Fernando, Kok, Kailash P, Bhatia, Bernd, Wissinger, Kate, Sergeant, Tobias B, Haack, Rita, Horvath, Semra, Hiz, Fowzan S, Alkuraya, Henry, Houlden, José Luiz, Pedroso, and Reza, Maroofian
- Abstract
Biallelic loss-of-functionTo fully characterize, both phenotypically and genotypically,We collected data from cases identified by screening genetic databases of several laboratories worldwide and systematically reviewed the literature.Nine unreportedOur case series expands phenotype-genotype correlations in
- Published
- 2021
36. Oscillatory Potentials in Achromatopsia as a Tool for Understanding Cone Retinal Functions
- Author
-
Giulia Righetti, Melanie Kempf, Christoph Braun, Ronja Jung, Susanne Kohl, Bernd Wissinger, Eberhart Zrenner, Katarina Stingl, and Krunoslav Stingl
- Subjects
Adult ,Male ,genetic structures ,QH301-705.5 ,cone functions ,Morlet wavelet transform ,Action Potentials ,Color Vision Defects ,time-frequency analysis ,Article ,oscillatory potentials ,Chemistry ,Retinal Rod Photoreceptor Cells ,Case-Control Studies ,ERG ,Electroretinography ,Retinal Cone Photoreceptor Cells ,Humans ,Female ,sense organs ,Biology (General) ,achromatopsia ,QD1-999 ,Photic Stimulation - Abstract
Achromatopsia (ACHM) is an inherited autosomal recessive disease lacking cone photoreceptors functions. In this study, we characterize the time-frequency representation of the full-field electroretinogram (ffERG) component oscillatory potentials (OPs), to investigate the connections between photoreceptors and the inner retinal network using ACHM as a model. Time-frequency characterization of OPs was extracted from 52 controls and 41 achromat individuals. The stimulation via ffERG was delivered under dark-adaptation (DA, 3.0 and 10.0 cd·s·m−2) to assess mixed rod-cone responses. The ffERG signal was subsequently analyzed using a continuous complex Morlet transform. Time-frequency maps of both DA conditions show the characterization of OPs, disclosing in both groups two distinct time-frequency windows (~70–100 Hz and >100 Hz) within 50 ms. Our main result indicates a significant cluster (p < 0.05) in both conditions of reduced relative power (dB) in ACHM people compared to controls, mainly at the time-frequency window >100 Hz. These results suggest that the strongly reduced but not absent activity of OPs above 100 Hz is mostly driven by cones and only in small part by rods. Thus, the lack of cone modulation of OPs gives important insights into interactions between photoreceptors and the inner retinal network and can be used as a biomarker for monitoring cone connection to the inner retina.
- Published
- 2021
37. Blue cone monochromacy: visual function and efficacy outcome measures for clinical trials.
- Author
-
Xunda Luo, Artur V Cideciyan, Alessandro Iannaccone, Alejandro J Roman, Lauren C Ditta, Barbara J Jennings, Svetlana A Yatsenko, Rebecca Sheplock, Alexander Sumaroka, Malgorzata Swider, Sharon B Schwartz, Bernd Wissinger, Susanne Kohl, and Samuel G Jacobson
- Subjects
Medicine ,Science - Abstract
BackgroundBlue Cone Monochromacy (BCM) is an X-linked retinopathy caused by mutations in the OPN1LW / OPN1MW gene cluster, encoding long (L)- and middle (M)-wavelength sensitive cone opsins. Recent evidence shows sufficient structural integrity of cone photoreceptors in BCM to warrant consideration of a gene therapy approach to the disease. In the present study, the vision in BCM is examined, specifically seeking clinically-feasible outcomes for a future clinical trial.MethodsBCM patients (n = 25, ages 5-72) were studied with kinetic and static chromatic perimetry, full-field sensitivity testing, and eye movement recordings. Vision at the fovea and parafovea was probed with chromatic microperimetry.ResultsKinetic fields with a Goldmann size V target were generally full. Short-wavelength (S-) sensitive cone function was normal or near normal in most patients. Light-adapted perimetry results on conventional background lights were abnormally reduced; 600-nm stimuli were seen by rods whereas white stimuli were seen by both rods and S-cones. Under dark-adapted conditions, 500-nm stimuli were seen by rods in both BCM and normals. Spectral sensitivity functions in the superior retina showed retained rod and S-cone functions in BCM under dark-adapted and light-adapted conditions. In the fovea, normal subjects showed L/M-cone mediation using a 650-nm stimulus under dark-adapted conditions, whereas BCM patients had reduced sensitivity driven by rod vision. Full-field red stimuli on bright blue backgrounds were seen by L/M-cones in normal subjects whereas BCM patients had abnormally reduced and rod-mediated sensitivities. Fixation location could vary from fovea to parafovea. Chromatic microperimetry demonstrated a large loss of sensitivity to red stimuli presented on a cyan adapting background at the anatomical fovea and surrounding parafovea.ConclusionsBCM rods continue to signal vision under conditions normally associated with daylight vision. Localized and retina-wide outcome measures were examined to evaluate possible improvement of L/M-cone-based vision in a clinical trial.
- Published
- 2015
- Full Text
- View/download PDF
38. Homozygous stop mutation in AHR causes autosomal recessive foveal hypoplasia and infantile nystagmus
- Author
-
Elfride De Baere, Gail Maconachie, Abdussalam Azem, Peter Bauer, Yuval Cohen, Martin Schulze, Birgit Lorenz, Bernd Wissinger, Rajech Sharkia, Basamat Almoallem, Irene Gottlob, Muhammad Mahajnah, Anja K. Mayer, Susanne Kohl, Elias I. Traboulsi, Adib Habib, and Mervyn G Thomas
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,genetic structures ,Consanguinity ,Nystagmus ,Nervous System Malformations ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Basic Helix-Loop-Helix Transcription Factors ,Electroretinography ,medicine ,Animals ,Humans ,Optic Nerve Hypoplasia ,Child ,Exome sequencing ,biology ,business.industry ,Homozygote ,Genetic disorder ,medicine.disease ,Aryl hydrocarbon receptor ,eye diseases ,Hypoplasia ,Pedigree ,030104 developmental biology ,Endocrinology ,Receptors, Aryl Hydrocarbon ,Mutation ,biology.protein ,Albinism ,Female ,Neurology (clinical) ,medicine.symptom ,business ,Nystagmus, Congenital ,030217 neurology & neurosurgery ,Horizontal pendular nystagmus - Abstract
Herein we present a consanguineous family with three children affected by foveal hypoplasia with infantile nystagmus, following an autosomal recessive mode of inheritance. The patients showed normal electroretinography responses, no signs of albinism, and no anterior segment or brain abnormalities. Upon whole exome sequencing (WES), we identified a homozygous mutation (c.1861C>T;p.Q621*) in the aryl hydrocarbon receptor (AHR) gene that perfectly co-segregated with the disease in the larger family. The aryl hydrocarbon receptor is a ligand-activated transcription factor that has been intensively studied in xenobiotic-induced toxicity. It was further shown to play a physiological role under normal cellular conditions, such as in immunity, inflammatory response and neurogenesis. Notably, knockout of the Ahr gene in the mouse impairs optic nerve myelin sheath formation and results in oculomotor deficits sharing many features with our patients: the eye movement disorder in the Ahr(-/-) mice appears early in development and presents as conjugate horizontal pendular nystagmus (Chevallier et al., 2013; Juricek et al., 2017). We therefore propose AHR to be a novel disease gene for a new, recessively inherited disorder in humans, characterized by infantile nystagmus and foveal hypoplasia.
- Published
- 2019
- Full Text
- View/download PDF
39. Relatively mild blue cone monochromacy phenotype caused by various haplotypes in the L- and M-cone opsin genes
- Author
-
Samer, Khateb, Aya, Shemesh, Ashly, Offenheim, Ruth, Sheffer, Tamar, Ben-Yosef, Itay, Chowers, Rina, Leibu, Britta, Baumann, Bernd, Wissinger, Susanne, Kohl, Eyal, Banin, and Dror, Sharon
- Subjects
Phenotype ,Haplotypes ,Electroretinography ,Myopia ,Humans ,Color Vision Defects ,Cone Opsins ,Pedigree - Abstract
Blue cone monochromacy (BCM) is an X-linked retinopathy caused by mutations in the red and green cone opsin genes. The aim of this study was to establish the clinical, genetic, and electrophysiological characteristics of a specific form of BCM.Patients harboring mutations in theTwenty-five patients harboring various haplotypes in exon 3 of theThe present study included genetic and clinical data from the largest cohort of patients with exon 3 haplotypes that were previously shown to cause missplicing of the
- Published
- 2021
40. CNGB1 ‐related rod‐cone dystrophy: A mutation review and update
- Author
-
Brooke Saffren, Bernd Wissinger, Eberhart Zrenner, Fadi Nasser, José-Alain Sahel, Christel Condroyer, Claire Marie Dhaenens, Stephen H. Tsang, Vivienne C. Greenstein, Rola Ba-Abbad, Isabelle Audo, Melanie Kempf, Susanne Kohl, Omar A. Mahroo, Cyntia Solis Hernandez, Andrew R. Webster, Nan-Kai Wang, Janet R. Sparrow, Saddek Mohand-Said, Vasily M. Smirnov, Simon M. Petersen-Jones, Sabine Defoort-Dhellemmes, Alex V. Levin, Laura Kühlewein, Sara D. Ragi, William W. Hauswirth, Jenina E. Capasso, Marco Nassisi, Michel Michaelides, Christina Zeitz, Stylianos Michalakis, Simona Degli Esposti, Aline Antonio, Institut de la Vision, Centre National de la Recherche Scientifique (CNRS)-Sorbonne Université (SU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts (CHNO), University of Milan, CHU Lille, University of Tübingen, Columbia University [New York], Ludwig Maximilian University [Munich] (LMU), University College of London [London] (UCL), University of Rochester [USA], University of Florida [Gainesville] (UF), Lille Neurosciences & Cognition - U 1172 (LilNCog (ex-JPARC)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), University of Pittsburgh (PITT), Pennsylvania Commonwealth System of Higher Education (PCSHE), Fondation Ophtalmologique Adolphe de Rothschild [Paris], Michigan State University [East Lansing], Michigan State University System, Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Università degli Studi di Milano = University of Milan (UNIMI), Lille Neurosciences & Cognition - U 1172 (LilNCog), and Gestionnaire, Hal Sorbonne Université
- Subjects
CNGB1 ,medicine.medical_specialty ,rod‐cone dystrophy ,[SDV]Life Sciences [q-bio] ,DNA Mutational Analysis ,cyclic nucleotide‐gated channel ,Cyclic Nucleotide-Gated Cation Channels ,Genomics ,Biology ,genotype-phenotype correlation ,medicine.disease_cause ,Cohort Studies ,03 medical and health sciences ,cyclic nucleotide-gated channel ,retinitis pigmentosa ,Retinitis pigmentosa ,Genetics ,medicine ,Rod-cone dystrophy ,Missense mutation ,Humans ,Genetics (clinical) ,genotype‐phenotype correlation ,Genetic Association Studies ,030304 developmental biology ,0303 health sciences ,Mutation ,Mutation Update ,030305 genetics & heredity ,Dystrophy ,medicine.disease ,3. Good health ,[SDV] Life Sciences [q-bio] ,inherited retinal disease ,RNA splicing ,Medical genetics ,rod-cone dystrophy ,Cone-Rod Dystrophies - Abstract
International audience; Cyclic nucleotide-gated channel β1 (CNGB1) encodes the 240-kDa β subunit of the rod photoreceptor cyclic nucleotide-gated ion channel. Disease-causing sequence variants in CNGB1 lead to autosomal recessive rod-cone dystrophy/retinitis pigmentosa (RP). We herein present a comprehensive review and analysis of all previously reported CNGB1 sequence variants, and add 22 novel variants, thereby enlarging the spectrum to 84 variants in total, including 24 missense variants (two of which may also affect splicing), 21 nonsense, 19 splicing defects (7 at noncanonical positions), 10 small deletions, 1 small insertion, 1 small insertion-deletion, 7 small duplications, and 1 gross deletion. According to the American College of Medical Genetics and Genomics classification criteria, 59 variants were considered pathogenic or likely pathogenic and 25 were variants of uncertain significance. In addition, we provide further phenotypic data from 34 CNGB1-related RP cases, which, overall, are in line with previous findings suggesting that this form of RP has long-term retention of useful central vision despite the early onset of night blindness, which is valuable for patient counseling, but also has implications for it being considered a priority target for gene therapy trials.
- Published
- 2021
- Full Text
- View/download PDF
41. Dominant ACO2 mutations are a frequent cause of isolated optic atrophy
- Author
-
Chiara LaMorgia, Pascal Reynier, Céline Wetterwald, Vincent Procaccio, Simone Schimpf-Linzenbold, Bernd Wissinger, Valérie Desquiret-Dumas, Stéphanie Chupin, Felix Tonagel, Leonardo Caporali, Selma Kane, Valerio Carelli, Magalie Barth, Naïg Gueguen, Xavier Zanlonghi, Majida Charif, Patrick Yu-Wai-Man, Neringa Jurkute, Morgane LeMao, Francesca Tagliavini, David Goudenège, Zouhair Elkarhat, Céline Bris, Marc Ferré, Jennifer Alban, Isabelle Meunier, Guy Lenaers, Arnaud Chevrollier, Abdelhamid Barakat, Ulrich Kellner, Patrizia Amati-Bonneau, Christophe Verny, Salim Khiati, Nicole Weisschuh, Philippe Gohier, Michele Carbonelli, Dominique Bonneau, Charif, Majida, Gueguen, Naïg, Ferré, Marc, Elkarhat, Zouhair, Khiati, Salim, LeMao, Morgane, Chevrollier, Arnaud, Desquiret-Dumas, Valerie, Goudenège, David, Bris, Céline, Kane, Selma, Alban, Jennifer, Chupin, Stéphanie, Wetterwald, Céline, Caporali, Leonardo, Tagliavini, Francesca, LaMorgia, Chiara, Carbonelli, Michele, Jurkute, Neringa, Barakat, Abdelhamid, Gohier, Philippe, Verny, Christophe, Barth, Magalie, Procaccio, Vincent, Bonneau, Dominique, Zanlonghi, Xavier, Meunier, Isabelle, Weisschuh, Nicole, Schimpf-Linzenbold, Simone, Tonagel, Felix, Kellner, Ulrich, Yu-Wai-Man, Patrick, Carelli, Valerio, Wissinger, Bernd, Amati-Bonneau, Patrizia, Reynier, Pascal, Lenaers, Guy, Université Mohamed 1 Oujda MAROC, MitoVasc - Physiopathologie Cardiovasculaire et Mitochondriale (MITOVASC), Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), SFR UA 4208 Interactions Cellulaires et Applications Thérapeutiques (ICAT), Université d'Angers (UA), Centre Hospitalier Universitaire d'Angers (CHU Angers), PRES Université Nantes Angers Le Mans (UNAM), Institut Pasteur du Maroc, Réseau International des Instituts Pasteur (RIIP), University of Bologna/Università di Bologna, Maggiore-Bellaria Hospital [Bologna], University College of London [London] (UCL), Moorfields Eye Hospital [London], Cambridge University Hospitals - NHS (CUH), University of Cambridge [UK] (CAM), Clinique Jules-Vernes [Nantes], Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), University of Tübingen, Molecular Genetics Laboratory [Tuebingen, Germany] (Centre for Ophthalmology), Institute for Ophthalmic Research [Tuebingen, Germany]-University Clinics Tuebingen [Germany], Rare Retinal Disease Center [Siegburg, Germany], AugenZentrum Siegburg-MVZ ADTC Siegburg GmbH [Germany], and LENAERS, Guy
- Subjects
0301 basic medicine ,Retinal degeneration ,Mitochondrial DNA ,[SDV]Life Sciences [q-bio] ,Biology ,Mitochondrion ,medicine.disease_cause ,Retinal ganglion ,Optic neuropathy ,03 medical and health sciences ,0302 clinical medicine ,Atrophy ,medicine ,ACO2 ,Genetics ,Mutation ,General Engineering ,medicine.disease ,eye diseases ,aconitase 2 ,optic neuropathy ,[SDV] Life Sciences [q-bio] ,mitochondria ,030104 developmental biology ,sense organs ,Krebs cycle ,030217 neurology & neurosurgery ,Optic nerve disorder - Abstract
Biallelic mutations in ACO2, encoding the mitochondrial aconitase 2, have been identified in individuals with neurodegenerative syndromes, including infantile cerebellar retinal degeneration and recessive optic neuropathies (locus OPA9). By screening European cohorts of individuals with genetically unsolved inherited optic neuropathies, we identified 61 cases harbouring variants in ACO2, among whom 50 carried dominant mutations, emphasizing for the first time the important contribution of ACO2 monoallelic pathogenic variants to dominant optic atrophy. Analysis of the ophthalmological and clinical data revealed that recessive cases are affected more severely than dominant cases, while not significantly earlier. In addition, 27% of the recessive cases and 11% of the dominant cases manifested with extraocular features in addition to optic atrophy. In silico analyses of ACO2 variants predicted their deleterious impacts on ACO2 biophysical properties. Skin derived fibroblasts from patients harbouring dominant and recessive ACO2 mutations revealed a reduction of ACO2 abundance and enzymatic activity, and the impairment of the mitochondrial respiration using citrate and pyruvate as substrates, while the addition of other Krebs cycle intermediates restored a normal respiration, suggesting a possible short-cut adaptation of the tricarboxylic citric acid cycle. Analysis of the mitochondrial genome abundance disclosed a significant reduction of the mitochondrial DNA amount in all ACO2 fibroblasts. Overall, our data position ACO2 as the third most frequently mutated gene in autosomal inherited optic neuropathies, after OPA1 and WFS1, and emphasize the crucial involvement of the first steps of the Krebs cycle in the maintenance and survival of retinal ganglion cells.
- Published
- 2021
- Full Text
- View/download PDF
42. A duplication on chromosome 16q12 affecting the IRXB gene cluster is associated with autosomal dominant cone dystrophy with early tritanopic color vision defect
- Author
-
Susanne Kohl, Ieva Sliesoraityte, Laura Kühlewein, Mary J. van Schooneveld, Bernd Wissinger, Eberhart Zrenner, Alexandra Sauer, Günther Rudolph, Pablo Llavona, Melanie Kempf, Florian A. Dehmelt, Peggy Reuter, Aristides B. Arrenberg, and Nicole Weisschuh
- Subjects
0301 basic medicine ,Male ,AcademicSubjects/SCI01140 ,genetic structures ,Color vision ,Locus (genetics) ,Color Vision Defects ,03 medical and health sciences ,0302 clinical medicine ,Cone dystrophy ,Gene duplication ,Gene cluster ,Chromosome Duplication ,Genetics ,medicine ,Animals ,Humans ,Cone Dystrophy ,Molecular Biology ,Zebrafish ,Genetics (clinical) ,Genes, Dominant ,Family Health ,Homeodomain Proteins ,Comparative Genomic Hybridization ,biology ,General Medicine ,Sequence Analysis, DNA ,medicine.disease ,biology.organism_classification ,eye diseases ,Pedigree ,030104 developmental biology ,Gene Expression Regulation ,Multigene Family ,030221 ophthalmology & optometry ,Female ,General Article ,Retinal Dystrophies ,Chromosomes, Human, Pair 16 ,Transcription Factors - Abstract
Cone dystrophies are a rare subgroup of inherited retinal dystrophies and hallmarked by color vision defects, low or decreasing visual acuity and central vision loss, nystagmus and photophobia. Applying genome-wide linkage analysis and array comparative genome hybridization, we identified a locus for autosomal dominant cone dystrophy on chromosome 16q12 in four independent multigeneration families. The locus is defined by duplications of variable size with a smallest region of overlap of 608 kb affecting the IRXB gene cluster and encompasses the genes IRX5 and IRX6. IRX5 and IRX6 belong to the Iroquois (Iro) protein family of homeodomain-containing transcription factors involved in patterning and regionalization of embryonic tissue in vertebrates, including the eye and the retina. All patients presented with a unique progressive cone dystrophy phenotype hallmarked by early tritanopic color vision defects. We propose that the disease underlies a misregulation of the IRXB gene cluster on chromosome 16q12 and demonstrate that overexpression of Irx5a and Irx6a, the two orthologous genes in zebrafish, results in visual impairment in 5-day-old zebrafish larvae.
- Published
- 2021
43. Simultaneous Detection of Multiple Point Mutations Using Fluorescence-Coupled Competitive Primer Extension
- Author
-
Sascha Fauser and Bernd Wissinger
- Subjects
Biology (General) ,QH301-705.5 - Abstract
We report the development of a method for the simultaneous genotyping of several distinct nucleotide positions by means of fluorescence-coupled competitive primer extension. We demonstrate the application of this method for the simultaneous detection of three point mutations in the human mitochondrial genome, at nucleotide positions 3460, 11778 and 14484, which account for about 90% of cases with Leber’s hereditary optic neuropathy. mtDNA fragments encompassing these nucleotide positions are initially amplified in a multiplex PCR assay. Genotyping is then carried out by a simultaneous primer extension assay using wild-type-specific (FAM-labeled) and mutant-specific (JOE-labeled) oligonucleotides. Primer extension products are separated on a 6% polyacrylamide/8 M urea gel on a fluorescence DNA sequencer. Patients’ genotypes can be derived from the peak color of the different-sized extension products. As little as 10% mutant DNA can be detected in heteroplasmic mixtures of wild-type and mutant mtDNA, a degree that is sufficient for routine clinical practice.
- Published
- 1997
- Full Text
- View/download PDF
44. Paternal uniparental isodisomy of chromosome 2 in a patient with CNGA3-associated autosomal recessive achromatopsia
- Author
-
Susanne Kohl, Britta Baumann, Francesca Dassie, Anja K. Mayer, Maria Solaki, Peggy Reuter, Laura Kühlewein, Bernd Wissinger, and Pietro Maffei
- Subjects
ACHM ,Male ,genetic structures ,Adolescent ,QH301-705.5 ,Cyclic Nucleotide-Gated Cation Channels ,Genes, Recessive ,Color Vision Defects ,Achromatopsia ,Chromosome 2 ,CNGA3 ,Uniparental isodisomy ,Chromosomes, Human, Pair 2 ,Female ,Humans ,Pedigree ,Phenotype ,Fathers ,Mutation ,Uniparental Disomy ,Article ,Chromosomes ,Recessive ,Biology (General) ,QD1-999 ,eye diseases ,Chemistry ,Genes ,Pair 2 ,Human - Abstract
Achromatopsia (ACHM) is a rare autosomal recessively inherited retinal disease characterized by congenital photophobia, nystagmus, low visual acuity, and absence of color vision. ACHM is genetically heterogeneous and can be caused by biallelic mutations in the genes CNGA3, CNGB3, GNAT2, PDE6C, PDE6H, or ATF6. We undertook molecular genetic analysis in a single female patient with a clinical diagnosis of ACHM and identified the homozygous variant c.778G>, C, p.(D260H) in the CNGA3 gene. While segregation analysis in the father, as expected, identified the CNGA3 variant in a heterozygous state, it could not be displayed in the mother. Microsatellite marker analysis provided evidence that the homozygosity of the CNGA3 variant is due to partial or complete paternal uniparental isodisomy (UPD) of chromosome 2 in the patient. Apart from the ACHM phenotype, the patient was clinically unsuspicious and healthy. This is one of few examples proving UPD as the underlying mechanism for the clinical manifestation of a recessive mutation in a patient with inherited retinal disease. It also highlights the importance of segregation analysis in both parents of a given patient or especially in cases of homozygous recessive mutations, as UPD has significant implications for genetic counseling with a very low recurrence risk assessment in such families.
- Published
- 2021
45. Union makes strength: a worldwide collaborative genetic and clinical study to provide a comprehensive survey of RD3 mutations and delineate the associated phenotype.
- Author
-
Isabelle Perrault, Alejandro Estrada-Cuzcano, Irma Lopez, Susanne Kohl, Shiqiang Li, Francesco Testa, Renate Zekveld-Vroon, Xia Wang, Esther Pomares, Jean Andorf, Nisrine Aboussair, Sandro Banfi, Nathalie Delphin, Anneke I den Hollander, Catherine Edelson, Ralph Florijn, Marc Jean-Pierre, Corinne Leowski, Andre Megarbane, Cristina Villanueva, Blanca Flores, Arnold Munnich, Huanan Ren, Ditta Zobor, Arthur Bergen, Rui Chen, Frans P M Cremers, Roser Gonzalez-Duarte, Robert K Koenekoop, Francesca Simonelli, Edwin Stone, Bernd Wissinger, Qingjiong Zhang, Josseline Kaplan, and Jean-Michel Rozet
- Subjects
Medicine ,Science - Abstract
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
- Full Text
- View/download PDF
46. Autosomal dominant optic atrophy: A novel treatment for
- Author
-
Christoph, Jüschke, Thomas, Klopstock, Claudia B, Catarino, Marta, Owczarek-Lipska, Bernd, Wissinger, and John, Neidhardt
- Subjects
endocrine system ,splicing ,ExSpeU1 ,U1 snRNA ,Optic Atrophy Type 1 ,Original Article ,DOA ,Dominant Optic Atrophy ,gene therapy ,OPA1 ,eye diseases ,ADOA ,Autosomal Dominant Optic Atrophy - Abstract
Autosomal dominant optic atrophy (ADOA) is frequently caused by mutations in the optic atrophy 1 (OPA1) gene, with haploinsufficiency being the major genetic pathomechanism. Almost 30% of the OPA1-associated cases suffer from splice defects. We identified a novel OPA1 mutation, c.1065+5G>A, in patients with ADOA. In patient-derived fibroblasts, the mutation led to skipping of OPA1 exon 10, reducing the OPA1 protein expression by approximately 50%. We developed a molecular treatment to correct the splice defect in OPA1 using engineered U1 splice factors retargeted to different locations in OPA1 exon 10 or intron 10. The strongest therapeutic effect was detected when U1 binding was engineered to bind to intron 10 at position +18, a position predicted by bioinformatics to be a promising binding site. We were able to significantly silence the effect of the mutation (skipping of exon 10) and simultaneously increase the expression level of normal transcripts. Retargeting U1 to the canonical splice donor site did not lead to a detectable splice correction. This proof-of-concept study indicates for the first time the feasibility of splice mutation correction as a treatment option for ADOA. Increasing the amount of correctly spliced OPA1 transcripts may suffice to overcome the haploinsufficiency., Graphical abstract, Optic atrophy is one of the most common inherited optic neuropathies, with progressive loss of vision starting in childhood. In patient-derived fibroblasts, the authors identified a mutation in OPA1 causing aberrant precursor mRNA splicing concomitant with reduced protein expression. Using engineered small nuclear RNAs, they could partially restore normal splicing.
- Published
- 2020
47. Clinical phenotype and course of PDE6A-associated retinitis pigmentosa disease, characterized in preparation for a gene supplementation trial
- Author
-
Günther Rudolph, Susanne Kohl, Alberta A H J Thiadens, Fadi Nasser, Carmen Ayuso, Thomas Rosenberg, Bart P. Leroy, Ulrich Kellner, Bernd Wissinger, Camiel J. F. Boon, Eberhart Zrenner, Sten Andréasson, Susan M Downes, Isabelle Meunier, Laura Kuehlewein, Béatrice Bocquet, M. Dominik Fischer, Saskia Biskup, Nicole Weisschuh, Frank G. Holz, Ditta Zobor, Katarina Stingl, Sandro Banfi, Barbara Wilhelm, Antje S Bernd, Ophthalmology, Kuehlewein, Laura, Zobor, Ditta, Andreasson, Sten Olof, Ayuso, Carmen, Banfi, Sandro, Bocquet, Beatrice, Bernd, Antje S, Biskup, Saskia, Boon, Camiel J F, Downes, Susan M, Fischer, M Dominik, Holz, Frank G, Kellner, Ulrich, Leroy, Bart P, Meunier, Isabelle, Nasser, Fadi, Rosenberg, Thoma, Rudolph, Günther, Stingl, Katarina, Thiadens, Alberta A H J, Wilhelm, Barbara, Wissinger, Bernd, Zrenner, Eberhart, Kohl, Susanne, Weisschuh, Nicole, and Amsterdam Neuroscience - Complex Trait Genetics
- Subjects
Adult ,Male ,medicine.medical_specialty ,Visual acuity ,genetic structures ,Adolescent ,Visual impairment ,Dark Adaptation ,Single Center ,Compound heterozygosity ,Article ,Internal medicine ,Retinitis pigmentosa ,medicine ,Electroretinography ,Humans ,Prospective Studies ,Child ,Eye Proteins ,Macular edema ,Original Investigation ,Aged ,Cyclic Nucleotide Phosphodiesterases, Type 6 ,business.industry ,Genetic Variation ,Genetic Therapy ,Middle Aged ,medicine.disease ,Ophthalmology ,Phenotype ,Dietary Supplements ,Cohort ,Female ,medicine.symptom ,Visual Fields ,business ,Retinitis Pigmentosa ,Tomography, Optical Coherence ,Cohort study - Abstract
Importance Treatment trials require sound knowledge on the natural course of disease. Objective To assess clinical features, genetic findings, and genotype-phenotype correlations in patients with retinitis pigmentosa (RP) associated with biallelic sequence variations in the PDE6A gene in preparation for a gene supplementation trial. Design, Setting, and Participants This prospective, longitudinal, observational cohort study was conducted from January 2001 to December 2019 in a single center (Centre for Ophthalmology of the University of Tubingen, Germany) with patients recruited multinationally from 12 collaborating European tertiary referral centers. Patients with retinitis pigmentosa, sequence variants in PDE6A, and the ability to provide informed consent were included. Exposures Comprehensive ophthalmological examinations; validation of compound heterozygosity and biallelism by familial segregation analysis, allelic cloning, or assessment of next-generation sequencing-read data, where possible. Main Outcomes and Measures Genetic findings and clinical features describing the entire cohort and comparing patients harboring the 2 most common disease-causing variants in a homozygous state (c.304C>A;p.(R102S) and c.998 + 1G>A;p.?). Results Fifty-seven patients (32 female patients [56%]; mean [SD], 40 [14] years) from 44 families were included. All patients completed the study. Thirty patients were homozygous for disease-causing alleles. Twenty-seven patients were heterozygous for 2 different PDE6A variants each. The most frequently observed alleles were c.304C>A;p.(R102S), c.998 + 1G>A;p.?, and c.2053G>A;p.(V685M). The mean (SD) best-corrected visual acuity was 0.43 (0.48) logMAR (Snellen equivalent, 20/50). The median visual field area with object III4e was 660 square degrees (5th and 95th percentiles, 76 and 11 019 square degrees; 25th and 75th percentiles, 255 and 3923 square degrees). Dark-adapted and light-adapted full-field electroretinography showed no responses in 88 of 108 eyes (81.5%). Sixty-nine of 108 eyes (62.9%) showed additional findings on optical coherence tomography imaging (eg, cystoid macular edema or macular atrophy). The variant c.998 + 1G>A;p.? led to a more severe phenotype when compared with the variant c.304C>A;p.(R102S). Conclusions and Relevance Seventeen of the PDE6A variants found in these patients appeared to be novel. Regarding the clinical findings, disease was highly symmetrical between the right and left eyes and visual impairment was mild or moderate in 90% of patients, providing a window of opportunity for gene therapy.Question What are the clinical features and course of retinitis pigmentosa associated with biallelic sequence variations in the PDE6A gene? Findings In this longitudinal cohort study of 57 adults, 17 of the PDE6A variants appeared to be novel. Disease was highly symmetrical between right and left eyes, and visual impairment was mild or moderate in 90% of patients. Meaning These data suggest that PDE6A-retinitis pigmentosa may be amenable to gene therapy.In this cohort study, 57 patients with biallelic sequence variations in the PDE6A gene and retinitis pigmentosa were followed up to assess clinical features, genetic findings, and genotype-phenotype correlations of the disease.
- Published
- 2020
48. Identification of Chemical and Pharmacological Chaperones for Correction of Trafficking-Deficient Mutant Cyclic Nucleotide-Gated A3 Channels
- Author
-
Susanne Kohl, Bernd Wissinger, Joachim Täger, and Peggy Reuter
- Subjects
0301 basic medicine ,Dihydropyridines ,Achromatopsia ,Cell Survival ,Mutant ,Aequorin ,Photoprotein ,Mutation, Missense ,Cyclic Nucleotide-Gated Cation Channels ,Color Vision Defects ,Genes, Recessive ,Protein degradation ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Missense mutation ,Humans ,Pharmacology ,biology ,Chemistry ,Endoplasmic reticulum ,medicine.disease ,Cell biology ,Protein Transport ,030104 developmental biology ,HEK293 Cells ,biology.protein ,Molecular Medicine ,Protein folding ,Calcium ,030217 neurology & neurosurgery - Abstract
Trafficking deficiency caused by missense mutations is a well known phenomenon that occurs for mutant, misfolded proteins. Typically, the misfolded protein is retained by the protein quality-control system and degraded by the endoplasmic reticulum-associated protein degradation pathway and thus does not reach its destination, although residual function of the protein may be preserved. Chemical and pharmacological chaperones can improve the targeting of trafficking-deficient proteins and thus may be promising candidates for therapeutic applications. Here, we report the application of a cellular bioassay based on the bioluminescent calcium reporter aequorin to quantify surface expression of mutant CNGA3 channels associated with the autosomal recessively inherited retinal disease achromatopsia. A screening of 77 compounds enabled the identification of effective chemical and pharmacological chaperones that result in a 1.5- to 4.8-fold increase of surface expression of mutant CNGA3. Using selected compounds, we confirmed that the rescue of the defective trafficking is not limited to a single mutation in CNGA3. Active compounds and our structure-activity correlated data for the dihydropyridine compound class may provide valuable information for developing a treatment of the trafficking defect in achromatopsia. SIGNIFICANCE STATEMENT This study describes a novel luminescence-based assay to detect the surface expression of mutant trafficking-deficient CNGA3 channels based on the calcium-sensitive photoprotein aequorin. Using this assay for a compound screening, this study identifies novel chemical and pharmacological chaperones that restore the surface localization of mutant trafficking-deficient CNGA3 channels. The results from this work may serve as starting point for the development of potent compounds that rescue trafficking deficiencies in the autosomal recessively inherited retinal disease achromatopsia.
- Published
- 2020
49. Foveal Therapy in Blue Cone Monochromacy: Predictions of Visual Potential From Artificial Intelligence
- Author
-
Bernd Wissinger, Samuel G. Jacobson, Susanne Kohl, Alexander Sumaroka, Rebecca Sheplock, Artur V. Cideciyan, and Vivian Wu
- Subjects
0301 basic medicine ,Retinal degeneration ,medicine.medical_specialty ,Visual acuity ,visual acuity ,genetic structures ,Computer science ,lcsh:RC321-571 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,BLUE CONE MONOCHROMACY ,cones ,rods ,Optical coherence tomography ,Foveal ,Ophthalmology ,medicine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Original Research ,chromatic perimetry ,optical coherence tomography ,medicine.diagnostic_test ,General Neuroscience ,Retinal ,medicine.disease ,eye diseases ,030104 developmental biology ,machine learning ,chemistry ,OPN1LW ,030221 ophthalmology & optometry ,Curve fitting ,retinal degeneration ,sense organs ,medicine.symptom ,random forest ,Neuroscience - Abstract
Novel therapeutic approaches for treating inherited retinal degenerations (IRDs) prompt a need to understand which patients with impaired vision have the anatomical potential to gain from participation in a clinical trial. We used supervised machine learning to predict foveal function from foveal structure in blue cone monochromacy (BCM), an X-linked congenital cone photoreceptor dysfunction secondary to mutations in the OPN1LW/OPN1MW gene cluster. BCM patients with either disease-associated large deletion or missense mutations were studied and results compared with those from subjects with other forms of IRD and various degrees of preserved central structure and function. A machine learning technique was used to associate foveal sensitivities and best-corrected visual acuities to foveal structure in IRD patients. Two random forest (RF) models trained on IRD data were applied to predict foveal function in BCM. A curve fitting method was also used and results compared with those of the RF models. The BCM and IRD patients had a comparable range of foveal structure. IRD patients had peak sensitivity at the fovea. Machine learning could successfully predict foveal sensitivity (FS) results from segmented or un-segmented optical coherence tomography (OCT) input. Application of machine learning predictions to BCM at the fovea showed differences between predicted and measured sensitivities, thereby defining treatment potential. The curve fitting method provided similar results. Given a measure of visual acuity (VA) and foveal outer nuclear layer thickness, the question of how many lines of acuity would represent the best efficacious result for each BCM patient could be answered. We propose that foveal vision improvement potential in BCM is predictable from retinal structure using machine learning and curve fitting approaches. This should allow estimates of maximal efficacy in patients being considered for clinical trials and also guide decisions about dosing.
- Published
- 2020
- Full Text
- View/download PDF
50. Dominant optic atrophy: Culprit mitochondria in the optic nerve
- Author
-
Christoph Jüschke, Pascal Reynier, Guy Lenaers, Yannick Le Dantec, Sarah Decembrini, Sebastian Swirski, Sinja Kieninger, Ungsoo Samuel Kim, Cavit Agca, Bernd Wissinger, Ting Xiao, Albert Neutzner, John Neidhardt, Patrick Yu-Wai-Man, MitoVasc - Physiopathologie Cardiovasculaire et Mitochondriale (MITOVASC), Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of Basel (Unibas), University Hospital Basel [Basel], University of Oldenburg, University of Tübingen, Sabanci University [Istanbul], University of Cambridge [UK] (CAM), Cambridge University Hospitals - NHS (CUH), Centre Hospitalier Universitaire d'Angers (CHU Angers), PRES Université Nantes Angers Le Mans (UNAM), University College of London [London] (UCL), and LENAERS, Guy
- Subjects
Retinal Ganglion Cells ,Mitochondrial DNA ,Dominant optic atrophy ,genetic structures ,[SDV]Life Sciences [q-bio] ,Mitochondrial disease ,Mitochondrion ,Biology ,OPA1 ,Retinal ganglion ,GTP Phosphohydrolases ,Mice ,Atrophy ,Optic Atrophy, Autosomal Dominant ,medicine ,Animals ,Humans ,Retina ,Optic Nerve ,medicine.disease ,eye diseases ,Sensory Systems ,Mitochondria ,[SDV] Life Sciences [q-bio] ,Ophthalmology ,medicine.anatomical_structure ,Optic nerve ,Intermembrane space ,Neuroscience - Abstract
International audience; Dominant optic atrophy (DOA) is an inherited mitochondrial disease leading to specific degeneration of retinal ganglion cells (RGCs), thus compromising transmission of visual information from the retina to the brain. Usually, DOA starts during childhood and evolves to poor vision or legal blindness, affecting the central vision, whilst sparing the peripheral visual field. In 20% of cases, DOA presents as syndromic disorder, with secondary symptoms affecting neuronal and muscular functions. Twenty years ago, we demonstrated that heterozygous mutations in OPA1 are the most frequent molecular cause of DOA. Since then, variants in additional genes, whose functions in many instances converge with those of OPA1, have been identified by next generation sequencing. OPA1 encodes a dynamin-related GTPase imported into mitochondria and located to the inner membrane and intermembrane space. The many OPA1 isoforms, resulting from alternative splicing of three exons, form complex homopolymers that structure mitochondrial cristae, and contribute to fusion of the outer membrane, thus shaping the whole mitochondrial network. Moreover, OPA1 is required for oxidative phosphorylation, maintenance of mitochondrial genome, calcium homeostasis and regulation of apoptosis, thus making OPA1 the Swiss army-knife of mitochondria. Understanding DOA pathophysiology requires the understanding of RGC peculiarities with respect to OPA1 functions. Besides the tremendous energy requirements of RGCs to relay visual information from the eye to the brain, these neurons present unique features related to their differential environments in the retina, and to the anatomical transition occurring at the lamina cribrosa, which parallel major adaptations of mitochondrial physiology and shape, in the pre- and post-laminar segments of the optic nerve. Three DOA mouse models, with different Opa1 mutations, have been generated to study intrinsic mechanisms responsible for RGC degeneration, and these have further revealed secondary symptoms related to mitochondrial dysfunctions, mirroring the more severe syndromic phenotypes seen in a subgroup of patients. Metabolomics analyses of cells, mouse organs and patient plasma mutated for OPA1 revealed new unexpected pathophysiological mechanisms related to mitochondrial dysfunction, and biomarkers correlated quantitatively to the severity of the disease. Here, we review and synthesize these data, and propose different approaches for embracing possible therapies to fulfil the unmet clinical needs of this disease, and provide hope to affected DOA patients.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.