Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nele Schwarz is active.

Publication


Featured researches published by Nele Schwarz.


Human Molecular Genetics | 2010

The retinitis pigmentosa protein RP2 links pericentriolar vesicle transport between the Golgi and the primary cilium

R. Jane Evans; Nele Schwarz; Kerstin Nagel-Wolfrum; Uwe Wolfrum; Alison J. Hardcastle; Michael E. Cheetham

Photoreceptors are complex ciliated sensory neurons. The basal body and periciliary ridge of photoreceptors function in association with the Golgi complex to regulate the export of proteins from the inner segment to the outer segment sensory axoneme. Here, we show that the retinitis pigmentosa protein RP2, which is a GTPase activating protein (GAP) for Arl3, localizes to the ciliary apparatus, namely the basal body and the associated centriole at the base of the photoreceptor cilium. Targeting to the ciliary base was dependent on N-terminal myristoylation. RP2 also localized to the Golgi and periciliary ridge of photoreceptors, which suggested a role for RP2 in regulating vesicle traffic and docking. To explore this hypothesis, we investigated the effect of RP2 depletion and the expression of a constitutively active form of Arl3 (Q71L) on pericentriolar vesicle transport. Kif3a, a component of intraflagellar transport (IFT), is important in cilia maintenance and transport of proteins through the connecting cilium in photoreceptors. Similar to Kif3a and Arl3 depletion, loss of RP2 led to fragmentation of the Golgi network. Depletion of RP2 and dysregulation of Arl3 resulted in dispersal of vesicles cycling cargo from the Golgi complex to the cilium, including the IFT protein IFT20. We propose that RP2 regulation of Arl3 is important for maintaining Golgi cohesion, facilitating the transport and docking of vesicles and thereby carrying proteins to the base of the photoreceptor connecting cilium for transport to the outer segment.


Human Molecular Genetics | 2012

Deep intronic mutation in OFD1, identified by targeted genomic next-generation sequencing, causes a severe form of X-linked retinitis pigmentosa (RP23)

Tom R. Webb; David A. Parfitt; Jessica C. Gardner; Ariadna Martinez; Dalila Bevilacqua; Alice E. Davidson; Ilaria Zito; Jacob Ressa; Marina Apergi; Nele Schwarz; Naheed Kanuga; Michel Michaelides; Michael E. Cheetham; Michael B. Gorin; Alison J. Hardcastle

X-linked retinitis pigmentosa (XLRP) is genetically heterogeneous with two causative genes identified, RPGR and RP2. We previously mapped a locus for a severe form of XLRP, RP23, to a 10.71 Mb interval on Xp22.31-22.13 containing 62 genes. Candidate gene screening failed to identify a causative mutation, so we adopted targeted genomic next-generation sequencing of the disease interval to determine the molecular cause of RP23. No coding variants or variants within or near splice sites were identified. In contrast, a variant deep within intron 9 of OFD1 increased the splice site prediction score 4 bp upstream of the variant. Mutations in OFD1 cause the syndromic ciliopathies orofaciodigital syndrome-1, which is male lethal, Simpson-Golabi-Behmel syndrome type 2 and Joubert syndrome. We tested the effect of the IVS9+706A>G variant on OFD1 splicing in vivo. In RP23 patient-derived RNA, we detected an OFD1 transcript with the insertion of a cryptic exon spliced between exons 9 and 10 causing a frameshift, p.N313fs.X330. Correctly spliced OFD1 was also detected in patient-derived RNA, although at reduced levels (39%), hence the mutation is not male lethal. Our data suggest that photoreceptors are uniquely susceptible to reduced expression of OFD1 and that an alternative disease mechanism can cause XLRP. This disease mechanism of reduced expression for a syndromic ciliopathy gene causing isolated retinal degeneration is reminiscent of CEP290 intronic mutations that cause Leber congenital amaurosis, and we speculate that reduced dosage of correctly spliced ciliopathy genes may be a common disease mechanism in retinal degenerations.


Cell Stem Cell | 2016

Identification and Correction of Mechanisms Underlying Inherited Blindness in Human iPSC-Derived Optic Cups

David A. Parfitt; Amelia Lane; Conor Ramsden; Amanda Jayne F Carr; Peter M.G. Munro; Katarina Jovanovic; Nele Schwarz; Naheed Kanuga; Manickam N. Muthiah; Sarah Hull; Jean-Marc Gallo; Lyndon da Cruz; Anthony T. Moore; Alison J. Hardcastle; Peter J. Coffey; Michael E. Cheetham

Summary Leber congenital amaurosis (LCA) is an inherited retinal dystrophy that causes childhood blindness. Photoreceptors are especially sensitive to an intronic mutation in the cilia-related gene CEP290, which causes missplicing and premature termination, but the basis of this sensitivity is unclear. Here, we generated differentiated photoreceptors in three-dimensional optic cups and retinal pigment epithelium (RPE) from iPSCs with this common CEP290 mutation to investigate disease mechanisms and evaluate candidate therapies. iPSCs differentiated normally into RPE and optic cups, despite abnormal CEP290 splicing and cilia defects. The highest levels of aberrant splicing and cilia defects were observed in optic cups, explaining the retinal-specific manifestation of this CEP290 mutation. Treating optic cups with an antisense morpholino effectively blocked aberrant splicing and restored expression of full-length CEP290, restoring normal cilia-based protein trafficking. These results provide a mechanistic understanding of the retina-specific phenotypes in CEP290 LCA patients and potential strategies for therapeutic intervention.


Human Molecular Genetics | 2015

Translational read-through of the RP2 Arg120stop mutation in patient iPSC-derived retinal pigment epithelium cells

Nele Schwarz; Amanda-Jayne F. Carr; Amelia Lane; Fabian Moeller; Li Li Chen; Mònica Aguilà; Britta Nommiste; Manickam N. Muthiah; Naheed Kanuga; Uwe Wolfrum; Kerstin Nagel-Wolfrum; Lyndon da Cruz; Peter J. Coffey; Michael E. Cheetham; Alison J. Hardcastle

Mutations in the RP2 gene lead to a severe form of X-linked retinitis pigmentosa. RP2 patients frequently present with nonsense mutations and no treatments are currently available to restore RP2 function. In this study, we reprogrammed fibroblasts from an RP2 patient carrying the nonsense mutation c.519C>T (p.R120X) into induced pluripotent stem cells (iPSC), and differentiated these cells into retinal pigment epithelial cells (RPE) to study the mechanisms of disease and test potential therapies. RP2 protein was undetectable in the RP2 R120X patient cells, suggesting a disease mechanism caused by complete lack of RP2 protein. The RP2 patient fibroblasts and iPSC-derived RPE cells showed phenotypic defects in IFT20 localization, Golgi cohesion and Gβ1 trafficking. These phenotypes were corrected by over-expressing GFP-tagged RP2. Using the translational read-through inducing drugs (TRIDs) G418 and PTC124 (Ataluren), we were able to restore up to 20% of endogenous, full-length RP2 protein in R120X cells. This level of restored RP2 was sufficient to reverse the cellular phenotypic defects observed in both the R120X patient fibroblasts and iPSC-RPE cells. This is the first proof-of-concept study to demonstrate successful read-through and restoration of RP2 function for the R120X nonsense mutation. The ability of the restored RP2 protein level to reverse the observed cellular phenotypes in cells lacking RP2 indicates that translational read-through could be clinically beneficial for patients.


Progress in Retinal and Eye Research | 2008

Molecular chaperones and photoreceptor function

Maria Kosmaoglou; Nele Schwarz; John S. Bett; Michael E. Cheetham

Molecular chaperones facilitate and regulate protein conformational change within cells. This encompasses many fundamental cellular processes: including the correct folding of nascent chains; protein transport and translocation; signal transduction and protein quality control. Chaperones are, therefore, important in several forms of human disease, including neurodegeneration. Within the retina, the highly specialized photoreceptor cell presents a fascinating paradigm to investigate the specialization of molecular chaperone function and reveals unique chaperone requirements essential to photoreceptor function. Mutations in several photoreceptor proteins lead to protein misfolding mediated neurodegeneration. The best characterized of these are mutations in the molecular light sensor, rhodopsin, which cause autosomal dominant retinitis pigmentosa. Rhodopsin biogenesis is likely to require chaperones, while rhodopsin misfolding involves molecular chaperones in quality control and the cellular response to protein aggregation. Furthermore, the specialization of components of the chaperone machinery to photoreceptor specific roles has been revealed by the identification of mutations in molecular chaperones that cause inherited retinal dysfunction and degeneration. These chaperones are involved in several important cellular pathways and further illuminate the essential and diverse roles of molecular chaperones.


Human Molecular Genetics | 2014

Hsp90 inhibition protects against inherited retinal degeneration

Mònica Aguilà; Dalila Bevilacqua; Caroline McCulley; Nele Schwarz; Dimitra Athanasiou; Naheed Kanuga; Sergey S. Novoselov; Clemens A. Lange; Robin R. Ali; James W. Bainbridge; Carlos Gias; Peter J. Coffey; Pere Garriga; Michael E. Cheetham

The molecular chaperone Hsp90 is important for the functional maturation of many client proteins, and inhibitors are in clinical trials for multiple indications in cancer. Hsp90 inhibition activates the heat shock response and can improve viability in a cell model of the P23H misfolding mutation in rhodopsin that causes autosomal dominant retinitis pigmentosa (adRP). Here, we show that a single low dose of the Hsp90 inhibitor HSP990 enhanced visual function and delayed photoreceptor degeneration in a P23H transgenic rat model. This was associated with the induction of heat shock protein expression and reduced rhodopsin aggregation. We then investigated the effect of Hsp90 inhibition on a different type of rod opsin mutant, R135L, which is hyperphosphorylated, binds arrestin and disrupts vesicular traffic. Hsp90 inhibition with 17-AAG reduced the intracellular accumulation of R135L and abolished arrestin binding in cells. Hsf-1−/− cells revealed that the effect of 17-AAG on P23H aggregation was dependent on HSF-1, whereas the effect on R135L was HSF-1 independent. Instead, the effect on R135L was mediated by a requirement of Hsp90 for rhodopsin kinase (GRK1) maturation and function. Importantly, Hsp90 inhibition restored R135L rod opsin localization to wild-type (WT) phenotype in vivo in rat retina. Prolonged Hsp90 inhibition with HSP990 in vivo led to a posttranslational reduction in GRK1 and phosphodiesterase (PDE6) protein levels, identifying them as Hsp90 clients. These data suggest that Hsp90 represents a potential therapeutic target for different types of rhodopsin adRP through distinct mechanisms, but also indicate that sustained Hsp90 inhibition might adversely affect visual function.


American Journal of Human Genetics | 2013

Mutations in ARL2BP, Encoding ADP-Ribosylation-Factor-Like 2 Binding Protein, Cause Autosomal-Recessive Retinitis Pigmentosa

Alice E. Davidson; Nele Schwarz; Lina Zelinger; Gabriele Stern-Schneider; Amelia Shoemark; Benjamin Spitzbarth; Menachem Gross; Uri Laxer; Jacob Sosna; Panagiotis I. Sergouniotis; Naushin Waseem; Rob Wilson; Richard A. Kahn; Vincent Plagnol; Uwe Wolfrum; Eyal Banin; Alison J. Hardcastle; Michael E. Cheetham; Dror Sharon; Andrew R. Webster

Retinitis pigmentosa (RP) is a genetically heterogeneous retinal degeneration characterized by photoreceptor death, which results in visual failure. Here, we used a combination of homozygosity mapping and exome sequencing to identify mutations in ARL2BP, which encodes an effector protein of the small GTPases ARL2 and ARL3, as causative for autosomal-recessive RP (RP66). In a family affected by RP and situs inversus, a homozygous, splice-acceptor mutation, c.101-1G>C, which alters pre-mRNA splicing of ARLBP2 in blood RNA, was identified. In another family, a homozygous c.134T>G (p.Met45Arg) mutation was identified. In the mouse retina, ARL2BP localized to the basal body and cilium-associated centriole of photoreceptors and the periciliary extension of the inner segment. Depletion of ARL2BP caused cilia shortening. Moreover, depletion of ARL2, but not ARL3, caused displacement of ARL2BP from the basal body, suggesting that ARL2 is vital for recruiting or anchoring ARL2BP at the base of the cilium. This hypothesis is supported by the finding that the p.Met45Arg amino acid substitution reduced binding to ARL2 and caused the loss of ARL2BP localization at the basal body in ciliated nasal epithelial cells. These data demonstrate a role for ARL2BP and ARL2 in primary cilia function and that this role is essential for normal photoreceptor maintenance and function.


Vision Research | 2012

Arl3 and RP2 mediated assembly and traffic of membrane associated cilia proteins

Nele Schwarz; Alison J. Hardcastle; Michael E. Cheetham

The traffic of proteins to the outer segment of photoreceptors is a fundamentally important process, which when perturbed results in photoreceptor cell death. Recent reports have revealed a novel pathway for the traffic of lipid-modified proteins involving the small GTPase Arl3 and its effectors PDEδ and Unc119. The retinitis pigmentosa protein RP2 is a GTPase activating protein (GAP) for Arl3 and also appears to regulate the assembly and traffic of membrane associated protein complexes. We recently identified the Gβ subunit of transducin (Gβ1) as a novel RP2 interacting protein. Our data support a role for RP2 in facilitating membrane association and traffic of Gβ1, potentially prior to the formation of the obligate Gβ:Gγ heterodimer. Here, we review the recent evidence that suggests that RP2 co-operates with Arl3 and its effectors in protein complex assembly and membrane specification for lipid-modified proteins. This is exemplified by the co-ordination of cilia associated traffic for heterotrimeric G proteins and we propose a model for the role of Arl3 and RP2 in this process.


Human Molecular Genetics | 2012

The X-linked retinitis pigmentosa protein RP2 facilitates G protein traffic

Nele Schwarz; Tatiana V. Novoselova; Robin Wait; Alison J. Hardcastle; Michael E. Cheetham

The X-linked retinitis pigmentosa protein RP2 is a GTPase activating protein (GAP) for the small GTPase Arl3 and both proteins are implicated in the traffic of proteins to the primary cilia. Here, we show that RP2 can facilitate the traffic of the Gβ subunit of transducin (Gβ1). Glutathione S-transferase (GST)-RP2 pulled down Gβ from retinal lysates and the interaction was specific to Gβ1, as Gβ3 or Gβ5L did not bind RP2. RP2 did not appear to interact with the Gβ:Gγ heterodimer, in contrast Gγ1 competed with RP2 for Gβ binding. Overexpression of Gβ1 in SK-N-SH cells led to a cytoplasmic accumulation of Gβ1, while co-expression of RP2 or Gγ1 with Gβ1 restored membrane association of Gβ1. Furthermore, RP2 small interfering RNA in ARPE19 cells resulted in a reduction in Gβ1 membrane association that was rescued by Gγ1 overexpression. The interaction of RP2 with Gβ1 required RP2 N-terminal myristolyation and the co-factor C (TBCC) homology domain. The interaction was also disrupted by the pathogenic mutation R118H, which blocks Arl3 GAP activity. Interestingly, Arl3-Q71L competed with Gβ1 for RP2 binding, suggesting that Arl3-GTP binding by RP2 would release Gβ1. RP2 also stimulated the association of Gβ1 with Rab11 vesicles. Collectively, the data support a role for RP2 in facilitating the membrane association and traffic of Gβ1, potentially prior to the formation of the obligate Gβ:Gγ heterodimer. Combined with other recent evidence, this suggests that RP2 may co-operate with Arl3 and its effectors in the cilia-associated traffic of G proteins.


Human Mutation | 2014

Three Different Cone Opsin Gene Array Mutational Mechanisms with Genotype–Phenotype Correlation and Functional Investigation of Cone Opsin Variants

Jessica C. Gardner; Gerald Liew; Yinghua Quan; Burcu Ermetal; Hisao Ueyama; Alice E. Davidson; Nele Schwarz; Naheed Kanuga; Ravinder Chana; Eamonn R. Maher; Andrew R. Webster; Graham E. Holder; Anthony G. Robson; Michael E. Cheetham; Jan Liebelt; Jonathan B Ruddle; Anthony T. Moore; Michel Michaelides; Alison J. Hardcastle

Mutations in the OPN1LW (L‐) and OPN1MW (M‐)cone opsin genes underlie a spectrum of cone photoreceptor defects from stationary loss of color vision to progressive retinal degeneration. Genotypes of 22 families with a range of cone disorders were grouped into three classes: deletions of the locus control region (LCR); missense mutation (p.Cys203Arg) in an L‐/M‐hybrid gene; and exon 3 single‐nucleotide polymorphism (SNP) interchange haplotypes in an otherwise normal gene array. Moderate‐to‐high myopia was observed in all mutation categories. Individuals with LCR deletions or p.Cys203Arg mutations were more likely to have nystagmus and poor vision, with disease progression in some p.Cys203Arg patients. Three disease‐associated exon 3 SNP haplotypes encoding LIAVA, LVAVA, or MIAVA were identified in our cohort. These patients were less likely to have nystagmus but more likely to show progression, with all patients over the age of 40 years having marked macular abnormalities. Previously, the haplotype LIAVA has been shown to result in exon 3 skipping. Here, we show that haplotypes LVAVA and MIAVA also result in aberrant splicing, with a residual low level of correctly spliced cone opsin. The OPN1LW/OPN1MW:c.532A>G SNP, common to all three disease‐associated haplotypes, appears to be principally responsible for this mutational mechanism.

Collaboration


Dive into the Nele Schwarz's collaboration.

Top Co-Authors

Avatar

Michael E. Cheetham

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Alison J. Hardcastle

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

David A. Parfitt

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Mònica Aguilà

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Naheed Kanuga

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Dalila Bevilacqua

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Alice E. Davidson

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Peter J. Coffey

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amelia Lane

UCL Institute of Ophthalmology

View shared research outputs
Researchain Logo
Decentralizing Knowledge