Network


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

Hotspot


Dive into the research topics where Simon M. Petersen-Jones is active.

Publication


Featured researches published by Simon M. Petersen-Jones.


The New England Journal of Medicine | 2008

Effect of Gene Therapy on Visual Function in Leber's Congenital Amaurosis

James W. Bainbridge; Alexander J. Smith; Susie S. Barker; Scott J. Robbie; Robert H. Henderson; Kamaljit S. Balaggan; Ananth C. Viswanathan; Graham E. Holder; Andrew Stockman; Nick Tyler; Simon M. Petersen-Jones; Shomi S. Bhattacharya; Adrian J. Thrasher; Fred W. Fitzke; Barrie J. Carter; Gary S. Rubin; Anthony T. Moore; Robin R. Ali

Early-onset, severe retinal dystrophy caused by mutations in the gene encoding retinal pigment epithelium-specific 65-kD protein (RPE65) is associated with poor vision at birth and complete loss of vision in early adulthood. We administered to three young adult patients subretinal injections of recombinant adeno-associated virus vector 2/2 expressing RPE65 complementary DNA (cDNA) under the control of a human RPE65 promoter. There were no serious adverse events. There was no clinically significant change in visual acuity or in peripheral visual fields on Goldmann perimetry in any of the three patients. We detected no change in retinal responses on electroretinography. One patient had significant improvement in visual function on microperimetry and on dark-adapted perimetry. This patient also showed improvement in a subjective test of visual mobility. These findings provide support for further clinical studies of this experimental approach in other patients with mutant RPE65. (ClinicalTrials.gov number, NCT00643747 [ClinicalTrials.gov].).


The New England Journal of Medicine | 2015

Long-term effect of gene therapy on Leber's congenital amaurosis.

James W. Bainbridge; M. S. Mehat; Venki Sundaram; S. J. Robbie; Susie E. Barker; Caterina Ripamonti; A. Georgiadis; Freya M. Mowat; S. G. Beattie; Peter J. Gardner; Kecia L. Feathers; Vy Luong; Suzanne Yzer; Kamaljit S. Balaggan; Ananth C. Viswanathan; T. de Ravel; Ingele Casteels; Graham E. Holder; Nick Tyler; Frederick W. Fitzke; Richard G. Weleber; Marko Nardini; Anthony T. Moore; Debra A. Thompson; Simon M. Petersen-Jones; Michel Michaelides; L. I. Van Den Born; Andrew Stockman; Alexander J. Smith; Gary S. Rubin

BACKGROUND Mutations in RPE65 cause Lebers congenital amaurosis, a progressive retinal degenerative disease that severely impairs sight in children. Gene therapy can result in modest improvements in night vision, but knowledge of its efficacy in humans is limited. METHODS We performed a phase 1-2 open-label trial involving 12 participants to evaluate the safety and efficacy of gene therapy with a recombinant adeno-associated virus 2/2 (rAAV2/2) vector carrying the RPE65 complementary DNA, and measured visual function over the course of 3 years. Four participants were administered a lower dose of the vector, and 8 were administered a higher dose. In a parallel study in dogs, we investigated the relationship among vector dose, visual function, and electroretinography (ERG) findings. RESULTS Improvements in retinal sensitivity were evident, to varying extents, in six participants for up to 3 years, peaking at 6 to 12 months after treatment and then declining. No associated improvement in retinal function was detected by means of ERG. Three participants had intraocular inflammation, and two had clinically significant deterioration of visual acuity. The reduction in central retinal thickness varied among participants. In dogs, RPE65 gene therapy with the same vector at lower doses improved vision-guided behavior, but only higher doses resulted in improvements in retinal function that were detectable with the use of ERG. CONCLUSIONS Gene therapy with rAAV2/2 RPE65 vector improved retinal sensitivity, albeit modestly and temporarily. Comparison with the results obtained in the dog model indicates that there is a species difference in the amount of RPE65 required to drive the visual cycle and that the demand for RPE65 in affected persons was not met to the extent required for a durable, robust effect. (Funded by the National Institute for Health Research and others; ClinicalTrials.gov number, NCT00643747.).


Gene Therapy | 2014

Tyrosine capsid-mutant AAV vectors for gene delivery to the canine retina from a subretinal or intravitreal approach

Freya M. Mowat; Kara R. Gornik; Astra Dinculescu; Sanford L. Boye; William W. Hauswirth; Simon M. Petersen-Jones; Joshua T. Bartoe

Recombinant adeno-associated viruses are important vectors for retinal gene delivery. Currently utilized vectors have relatively slow onset, and for efficient transduction it is necessary to deliver treatment subretinally, with the potential for damage to the retina. Amino-acid substitutions in the viral capsid improve efficiency in rodent eyes by evading host responses. As dogs are important large animal models for human retinitis pigmentosa, we evaluated the speed and efficiency of retinal transduction using capsid-mutant vectors injected both subretinally and intravitreally. We evaluated AAV serotypes 2 and 8 with amino-acid substitutions of surface-exposed capsid tyrosine residues. The chicken beta-actin promoter was used to drive green fluorescent protein expression. Twelve normal adult beagles were injected; four dogs received intravitreal injections and eight dogs received subretinal injections. Capsid-mutant viruses tested included AAV2(quad Y-F) (intravitreal and subretinal) and self-complementary scAAV8(Y733F) (subretinal only). Contralateral control eyes received injections of scAAV5 (subretinal) or scAAV2 (intravitreal). Subretinally delivered vectors had a faster expression onset than intravitreally delivered vectors. Subretinally delivered scAAV8(Y733F) had a faster onset of expression than scAAV5. All subretinally injected vector types transduced the outer retina with high efficiency and the inner retina with moderate efficiency. Intravitreally delivered AAV2(quad Y-F) had a marginally higher efficiency of transduction of both outer retinal and inner retinal cells than scAAV2. Because of their rapid expression onset and efficient transduction, subretinally delivered capsid-mutant AAV8 vectors may increase the efficacy of gene therapy treatment for rapid photoreceptor degenerative diseases. With further refinement, capsid-mutant AAV2 vectors show promise for retinal gene delivery from an intravitreal approach.


Gene Therapy | 2011

Gene therapy in the second eye of RPE65-deficient dogs improves retinal function.

Matthew J. Annear; Joshua T. Bartoe; Susie E. Barker; Alexander J. Smith; Paul G. Curran; James W. Bainbridge; Robin R. Ali; Simon M. Petersen-Jones

The purpose of this study was to evaluate whether immune responses interfered with gene therapy rescue using subretinally delivered recombinant adeno-associated viral vector serotype 2 carrying the RPE65 cDNA gene driven by the human RPE65 promoter (rAAV2.hRPE65p.hRPE65) in the second eye of RPE65−/− dogs that had previously been treated in a similar manner in the other eye. Bilateral subretinal injection was performed in nine dogs with the second eye treated 85–180 days after the first. Electroretinography (ERG) and vision testing showed rescue in 16 of 18 treated eyes, with no significant difference between first and second treated eyes. A serum neutralizing antibody (NAb) response to rAAV2 was detected in all treated animals, but this did not prevent or reduce the effectiveness of rescue in the second treated eye. We conclude that successful rescue using subretinal rAAV2.hRPE65p.hRPE65 gene therapy in the second eye is not precluded by prior gene therapy in the contralateral eye of the RPE65−/− dog. This finding has important implications for the treatment of human LCA type II patients.


Gene Therapy | 2013

RPE65 gene therapy slows cone loss in Rpe65-deficient dogs

Freya M. Mowat; A R Breuwer; Joshua T. Bartoe; Matthew J. Annear; Z Zhang; A J Smith; James W. Bainbridge; Simon M. Petersen-Jones; Robin R. Ali

Recent clinical trials of retinal pigment epithelium gene (RPE65) supplementation therapy in Leber congenital amaurosis type 2 patients have demonstrated improvements in rod and cone function, but it may be some years before the effects of therapy on photoreceptor survival become apparent. The Rpe65-deficient dog is a very useful pre-clinical model in which to test efficacy of therapies, because the dog has a retina with a high degree of similarity to that of humans. In this study, we evaluated the effect of RPE65 gene therapy on photoreceptor survival in order to predict the potential benefit and limitations of therapy in patients. We examined the retinas of Rpe65-deficient dogs after RPE65 gene therapy to evaluate the preservation of rods and cone photoreceptor subtypes. We found that gene therapy preserves both rods and cones. While the moderate loss of rods in the Rpe65-deficient dog retina is slowed by gene therapy, S-cones are lost extensively and gene therapy can prevent that loss, although only within the treated area. Although LM-cones are not lost extensively, cone opsin mislocalization indicates that they are stressed, and this can be partially reversed by gene therapy. Our results suggest that gene therapy may be able to slow cone degeneration in patients if intervention is sufficiently early and also that it is probably important to treat the macula in order to preserve central function.


Archives of Ophthalmology | 2010

Improvement of Visual Performance With Intravitreal Administration of 9-cis-Retinal in Rpe65-Mutant Dogs

Patricia M. Gearhart; Chris Gearhart; Debra A. Thompson; Simon M. Petersen-Jones

OBJECTIVE To determine the efficacy of intravitreal administration of 9-cis-retinal in restoring visual function in Rpe65-mutant dogs. METHODS Intravitreal injection of 9-cis-retinal was administered in 1 eye of 7 Rpe65-/- dogs at a range of ages. Electroretinogram analysis and testing of visual performance was used to evaluate outcomes after a single injection and in 2 dogs after a second injection in the same eye. RESULTS In 5 of 7 injected dogs, 9-cis-retinal injection resulted in increased rod electroretinogram responses and improved functional vision. Three injected dogs exhibited increased 33-Hz flicker amplitudes characteristic of cone-mediated responses. Electroretinogram improvement was no longer evident by week 10 postinjection in 1 dog monitored over time. A second injection of 9-cis-retinal was performed in the same eye of 2 of the 7 dogs and also resulted in rescue of visual function. CONCLUSION Our findings establish that 9-cis-retinoid therapy can restore visual function in a canine model of human disease resulting from RPE65 mutations. CLINICAL RELEVANCE These positive proof-of-principle results provide support for the development of intravitreal devices for sustained delivery of 9-cis-retinal as a therapy for conditions resulting from failure of the visual cycle.


Investigative Ophthalmology & Visual Science | 2009

Characterization of a Canine Model of Autosomal Recessive Retinitis Pigmentosa due to a PDE6A Mutation

N. Tuntivanich; Steven J. Pittler; Andy J. Fischer; Ghezal Omar; Matti Kiupel; Arthur J. Weber; Suxia Yao; Juan P. Steibel; Naheed W. Khan; Simon M. Petersen-Jones

PURPOSE To characterize a canine model of autosomal recessive RP due to a PDE6A gene mutation. METHODS Affected and breed- and age-matched control puppies were studied by electroretinography (ERG), light and electron microscopy, immunohistochemistry, and assay for retinal PDE6 levels and enzymatic activity. RESULTS The mutant puppies failed to develop normal rod-mediated ERG responses and had reduced light-adapted a-wave amplitudes from an early age. The residual ERG waveforms originated primarily from cone-driven responses. Development of photoreceptor outer segments stopped, and rod cells were lost by apoptosis. Immunohistochemistry demonstrated a marked reduction in rod opsin immunostaining outer segments and relative preservation of cones early in the disease process. With exception of rod bipolar cells, which appeared to be reduced in number relatively early in the disease process, other inner retinal cells were preserved in the early stages of the disease, although there was marked and early activation of Müller glia. Western blot analysis showed that the PDE6A mutation not only resulted in a lack of PDE6A protein but the affected retinas also lacked the other PDE6 subunits, suggesting expression of PDE6A is essential for normal expression of PDE6B and PDE6G. Affected retinas lacked PDE6 enzymatic activity. CONCLUSIONS This represents the first characterization of a PDE6A model of autosomal recessive retinitis pigmentosa, and the PDE6A mutant dog shows promise as a large animal model for investigation of therapies to rescue mutant rod photoreceptors and to preserve cone photoreceptors in the face of a rapid loss of rod cells.


Documenta Ophthalmologica | 2013

Guidelines for clinical electroretinography in the dog: 2012 update

Björn Ekesten; András M. Komáromy; Ron Ofri; Simon M. Petersen-Jones; Kristina Narfström

The full-field, flash electroretinogram (ERG) is now a widely used test of canine retinal function for the clinical diagnosis of hereditary retinal dystrophies and other causes of retinal degeneration, assessment of retinal function in patients with opaque media, ruling out of generalized retinal diseases in patients with sudden loss of vision and in ophthalmological research, as well as in pharmaceutical and toxicological screening for deleterious side effects of drugs and other chemical compounds. In 2002, the first guidelines for clinical ERGs in this species adopted by the European College of Veterinary Ophthalmologists were published. This work provides an update of these guidelines.


Neuroscience | 2010

The pattern of expression of guanine nucleotide-binding protein β3 in the retina is conserved across vertebrate species

Eric R. Ritchey; Rachel Bongini; Kimberly A. Code; Christopher Zelinka; Simon M. Petersen-Jones; Andy J. Fischer

Guanine nucleotide-binding protein beta3 (GNB3) is an isoform of the beta subunit of the heterotrimeric G protein second messenger complex that is commonly associated with transmembrane receptors. The presence of GNB3 in photoreceptors, and possibly bipolar cells, has been confirmed in murine, bovine and primate retinas [Lee RH, Lieberman BS, Yamane HK, Bok D, Fung BK (1992) J Biol Chem 267:24776-24781; Peng YW, Robishaw JD, Levine MA, Yau KW (1992) Proc Natl Acad Sci U S A 89:10882-10886; Huang L, Max M, Margolskee RF, Su H, Masland RH, Euler T (2003) J Comp Neurol 455:1-10]. Studies have indicated that a mutation in the GNB3 gene causes progressive retinopathy and globe enlargement (RGE) in chickens. The goals of this study were to (1) examine the expression pattern of GNB3 in wild-type and RGE mutant chickens, (2) characterize the types of bipolar cells that express GNB3 and (3) examine whether the expression of GNB3 in the retina is conserved across vertebrate species. We find that chickens homozygous for the RGE allele completely lack GNB3 protein. We find that the pattern of expression of GNB3 in the retina is highly conserved across vertebrate species, including teleost fish (Carassius auratus), frogs (Xenopus laevis), chickens (Gallus domesticus), mice (Mus musculata), guinea-pigs (Cavia porcellus), dogs (Canis familiaris) and non-human primates (Macaca fasicularis). Regardless of the species, we find that GNB3 is expressed by Islet1-positive cone ON-bipolar cells and by cone photoreceptors. In some vertebrates, GNB3-immunoreactivity was observed in both rod and cone photoreceptors. A protein-protein alignment of GNB3 across different vertebrates, from fish to humans, indicates a high degree (>92%) of sequence conservation. Given that analogous types of retinal neurons express GNB3 in different species, we propose that the functions and the mechanisms that regulate the expression of GNB3 are highly conserved.


Vision Research | 2003

Clinical features of the retinopathy, globe enlarged (rge) chick phenotype

Fabiano Montiani-Ferreira; Tong Li; Matti Kiupel; Howard C. Howland; Paul Hocking; Roger Curtis; Simon M. Petersen-Jones

The purpose of the study reported here was to characterize the clinical aspects of the autosomal recessive retinopathy, globe enlarged (rge) phenotype in chicks (Gallus gallus). Rge/rge, rge/+ and +/+ chicks were studied from hatch to 336 days of age by general clinical examination, post-mortem examination, vision testing with an optokinetic device, ophthalmoscopy, biomicroscopy, tonometry, central corneal pachymetry, a-mode ultrasonography, infrared photoretinoscopy and photokeratometry. Additionally, preliminary electroretinographic and histopathologic investigations were performed. There is a variable degree of vision loss in rge/rge chicks at 1 day of age with further chicks losing vision over the next few weeks until all chicks become functionally blind by 30 days of age (although some optokinetic responses remain in some of the rge/rge chicks). Over the first few weeks of life rge/rge chicks develop thicker corneas with a larger radius, hyperopia, shallower anterior chambers and enlarged globes both radially and axially, compared to controls. A preliminary ERG study showed that 1 day old rge/rge chicks have an elevated response threshold, a lower amplitude a-wave with a markedly shallow leading slope, a lack of both oscillatory responses and c-waves and, at brighter flashes, an increased b-wave amplitude. Light microscopy revealed no gross retinal abnormalities in young chicks to account for the blindness. A thinning of all retinal layers developed in parallel with globe enlargement. The rge defect is a unique progressive retinal dystrophy that results in a severe visual deficit, abnormal electroretinographic waveforms, and secondary globe enlargement.

Collaboration


Dive into the Simon M. Petersen-Jones's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Freya M. Mowat

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James W. Bainbridge

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robin R. Ali

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge