Network


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

Hotspot


Dive into the research topics where Robin R. Ali is active.

Publication


Featured researches published by Robin R. Ali.


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].).


Nature | 2006

Retinal repair by transplantation of photoreceptor precursors.

Robert E. MacLaren; Rachael A. Pearson; Angus MacNeil; R. H. Douglas; T.E. Salt; M Akimoto; Anand Swaroop; Jane C. Sowden; Robin R. Ali

Photoreceptor loss causes irreversible blindness in many retinal diseases. Repair of such damage by cell transplantation is one of the most feasible types of central nervous system repair; photoreceptor degeneration initially leaves the inner retinal circuitry intact and new photoreceptors need only make single, short synaptic connections to contribute to the retinotopic map. So far, brain- and retina-derived stem cells transplanted into adult retina have shown little evidence of being able to integrate into the outer nuclear layer and differentiate into new photoreceptors. Furthermore, there has been no demonstration that transplanted cells form functional synaptic connections with other neurons in the recipient retina or restore visual function. This might be because the mature mammalian retina lacks the ability to accept and incorporate stem cells or to promote photoreceptor differentiation. We hypothesized that committed progenitor or precursor cells at later ontogenetic stages might have a higher probability of success upon transplantation. Here we show that donor cells can integrate into the adult or degenerating retina if they are taken from the developing retina at a time coincident with the peak of rod genesis. These transplanted cells integrate, differentiate into rod photoreceptors, form synaptic connections and improve visual function. Furthermore, we use genetically tagged post-mitotic rod precursors expressing the transcription factor Nrl (ref. 6) (neural retina leucine zipper) to show that successfully integrated rod photoreceptors are derived only from immature post-mitotic rod precursors and not from proliferating progenitor or stem cells. These findings define the ontogenetic stage of donor cells for successful rod photoreceptor transplantation.


The Lancet | 2004

Gene therapy of X-linked severe combined immunodeficiency by use of a pseudotyped gammaretroviral vector

H. Bobby Gaspar; Kathryn L. Parsley; Steven J. Howe; Doug King; Kimberly Gilmour; Joanna Sinclair; Gaby Brouns; Manfred Schmidt; Christof von Kalle; Torben Barington; Marianne Antonius Jakobsen; Hans Ole Christensen; Abdulaziz Al Ghonaium; Harry White; J. L. Smith; Roland J. Levinsky; Robin R. Ali; Christine Kinnon; Adrian J. Thrasher

BACKGROUND X-linked severe combined immunodeficiency (SCID-X1) is caused by mutations in the common cytokine-receptor gamma chain (gamma(c)), resulting in disruption of development of T lymphocytes and natural-killer cells. B-lymphocyte function is also intrinsically compromised. Allogeneic bone-marrow transplantation is successful if HLA-matched family donors are available, but HLA-mismatched procedures are associated with substantial morbidity and mortality. We investigated the application of somatic gene therapy by use of a gibbon-ape-leukaemia-virus pseudotyped gammaretroviral vector. METHODS Four children with SCID-X1 were enrolled. Autologous CD34-positive haemopoietic bone-marrow stem cells were transduced ex vivo and returned to the patients without preceding cytoreductive chemotherapy. The patients were monitored for integration and expression of the gamma(c) vector and for functional immunological recovery. FINDINGS All patients have shown substantial improvements in clinical and immunological features, and prophylactic medication could be withdrawn in two. No serious adverse events have been recorded. T cells responded normally to mitogenic and antigenic stimuli, and the T-cell-receptor (TCR) repertoire was highly diverse. Where assessable, humoral immunity, in terms of antibody production, was also restored and associated with increasing rates of somatic mutation in immunoglobulin genes. INTERPRETATION Gene therapy for SCID-X1 is a highly effective strategy for restoration of functional cellular and humoral immunity.


Nature Medicine | 2006

Effective gene therapy with nonintegrating lentiviral vectors

Rafael J. Yáñez-Muñoz; Kamaljit S. Balaggan; Angus MacNeil; Steven J. Howe; Manfred Schmidt; Alexander J. Smith; Prateek K. Buch; Robert E. MacLaren; Patrick N. Anderson; Susie E. Barker; Yanai Duran; Cynthia C. Bartholomae; Christof von Kalle; John R. Heckenlively; Christine Kinnon; Robin R. Ali; Adrian J. Thrasher

Retroviral and lentiviral vector integration into host-cell chromosomes carries with it a finite chance of causing insertional mutagenesis. This risk has been highlighted by the induction of malignancy in mouse models, and development of lymphoproliferative disease in three individuals with severe combined immunodeficiency–X1 (refs. 2,3). Therefore, a key challenge for clinical therapies based on retroviral vectors is to achieve stable transgene expression while minimizing insertional mutagenesis. Recent in vitro studies have shown that integration-deficient lentiviral vectors can mediate stable transduction. With similar vectors, we now show efficient and sustained transgene expression in vivo in rodent ocular and brain tissues. We also show substantial rescue of clinically relevant rodent models of retinal degeneration. Therefore, the high efficiency of gene transfer and expression mediated by lentiviruses can be harnessed in vivo without a requirement for vector integration. For therapeutic application to postmitotic tissues, this system substantially reduces the risk of insertional mutagenesis.


Nature Genetics | 2000

Restoration of photoreceptor ultrastructure and function in retinal degeneration slow mice by gene therapy

Robin R. Ali; Gian-Marco Sarra; C Stephens; M de Alwis; James W. Bainbridge; Peter M.G. Munro; Sascha Fauser; M. B. Reichell; Christine Kinnon; David M. Hunt; Shomi S. Bhattacharya; Adrian J. Thrasher

The gene Prph2 encodes a photoreceptor-specific membrane glycoprotein, peripherin-2 (also known as peripherin/rds), which is inserted into the rims of photoreceptor outer segment discs in a complex with rom-1 (ref. 2). The complex is necessary for the stabilization of the discs, which are renewed constantly throughout life, and which contain the visual pigments necessary for photon capture. Mutations in Prph2 have been shown to result in a variety of photoreceptor dystrophies, including autosomal dominant retinitis pigmentosa and macular dystrophy. A common feature of these diseases is the loss of photoreceptor function, also seen in the retinal degeneration slow (rds or Prph2 Rd2/Rd2) mouse, which is homozygous for a null mutation in Prph2. It is characterized by a complete failure to develop photoreceptor discs and outer segments, downregulation of rhodopsin and apoptotic loss of photoreceptor cells. The electroretinograms (ERGs) of Prph2Rd2/Rd2 mice have greatly diminished a-wave and b-wave amplitudes, which decline to virtually undetectable concentrations by two months. Subretinal injection of recombinant adeno-associated virus (AAV) encoding a Prph2 transgene results in stable generation of outer segment structures and formation of new stacks of discs containing both perpherin-2 and rhodopsin, which in many cases are morphologically similar to normal outer segments. Moreover, the re-establishment of the structural integrity of the photoreceptor layer also results in electrophysiological correction. These studies demonstrate for the first time that a complex ultrastructural cell defect can be corrected both morphologically and functionally by in vivo gene transfer.


Nature | 2012

Restoration of vision after transplantation of photoreceptors

Rachael A. Pearson; Amanda C. Barber; Matteo Rizzi; Claire Hippert; Tian Xue; Emma L. West; Yanai Duran; Anthony J. Smith; J. Z. Chuang; S A Sultana Azam; Ulrich F.O. Luhmann; Andrea Benucci; Choon Ho Sung; James W. Bainbridge; Matteo Carandini; King Wai Yau; Jane C. Sowden; Robin R. Ali

Cell transplantation is a potential strategy for treating blindness caused by the loss of photoreceptors. Although transplanted rod-precursor cells are able to migrate into the adult retina and differentiate to acquire the specialized morphological features of mature photoreceptor cells, the fundamental question remains whether transplantation of photoreceptor cells can actually improve vision. Here we provide evidence of functional rod-mediated vision after photoreceptor transplantation in adult Gnat1−/− mice, which lack rod function and are a model of congenital stationary night blindness. We show that transplanted rod precursors form classic triad synaptic connections with second-order bipolar and horizontal cells in the recipient retina. The newly integrated photoreceptor cells are light-responsive with dim-flash kinetics similar to adult wild-type photoreceptors. By using intrinsic imaging under scotopic conditions we demonstrate that visual signals generated by transplanted rods are projected to higher visual areas, including V1. Moreover, these cells are capable of driving optokinetic head tracking and visually guided behaviour in the Gnat1−/− mouse under scotopic conditions. Together, these results demonstrate the feasibility of photoreceptor transplantation as a therapeutic strategy for restoring vision after retinal degeneration.


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.).


Nature Biotechnology | 2013

Photoreceptor precursors derived from three-dimensional embryonic stem cell cultures integrate and mature within adult degenerate retina

Anai Gonzalez-Cordero; Emma L. West; Rachael A. Pearson; Yanai Duran; Livia S. Carvalho; Colin Chu; Arifa Naeem; Samuel J.I. Blackford; Anastasios Georgiadis; Jorn Lakowski; Mike Hubank; Alexander J. Smith; James W. Bainbridge; Jane C. Sowden; Robin R. Ali

Irreversible blindness caused by loss of photoreceptors may be amenable to cell therapy. We previously demonstrated retinal repair1 and restoration of vision through transplantation of photoreceptor precursors obtained from post-natal retinas into visually impaired adult mice2,3. Considerable progress has been made in differentiating embryonic stem cells (ESCs) in vitro toward photoreceptor lineages4-6. However, the capability of ESC-derived photoreceptors to integrate after transplantation has not been demonstrated unequivocally. Here, to isolate photoreceptor precursors fit for transplantation, we adapted a recently reported three-dimensional (3D) differentiation protocol that generates neuroretina from mouse ESCs6. We show that Rhop.GFP-selected rod precursors derived by this protocol integrate within degenerate retinae of adult mice and mature into outer segment–bearing photoreceptors. Notably, ESC-derived precursors at a developmental stage similar to postnatal days 4-8 integrate more efficiently than cells at other stages. This study shows conclusively that ESCs can provide a source of photoreceptors for retinal cell transplantation.


Gene Therapy | 2001

In vivo gene transfer to the mouse eye using an HIV-based lentiviral vector; efficient long-term transduction of corneal endothelium and retinal pigment epithelium

James W. Bainbridge; C Stephens; K. Parsley; C. Demaison; A. Halfyard; Aj Thrasher; Robin R. Ali

We have evaluated the transduction profiles of an HIV-based lentiviral vector delivered regionally to ocular tissues in vivo. Following subretinal injection, a green fluorescent protein (GFP) reporter gene was efficiently and stably expressed in retinal pigment epithelial (RPE) cells. Limited transduction of adjacent photoreceptors occurred in newborn mice, but was inefficient in adult animals. Injection of the vector into the anterior chamber resulted in efficient and stable transduction of corneal endothelial cells. Efficient in vivo gene transfer into cells of the corneal endothelium and retinal pigment epithelium by lentiviral vectors may therefore offer a valuable approach to the treatment of disorders of the cornea and outer retina.


Gene Therapy | 2007

Restoration of vision in RPE65-deficient Briard dogs using an AAV serotype 4 vector that specifically targets the retinal pigmented epithelium.

G Le Meur; Knut Stieger; Alexander J. Smith; M. Weber; Jack-Yves Deschamps; D. Nivard; Alexandra Mendes-Madeira; Nathalie Provost; Yann Péréon; Yan Cherel; Robin R. Ali; Christian P. Hamel; Philippe Moullier; Fabienne Rolling

Previous studies have tested gene replacement therapy in RPE65-deficient dogs using recombinant adeno-associated virus 2/2 (rAAV2/2), -2/1 or -2/5 mediated delivery of the RPE65 gene. They all documented restoration of dark- and light-adapted electroretinography responses and improved psychophysical outcomes. Use of a specific RPE65 promoter and a rAAV vector that targets transgene expression specifically to the RPE may, however, provide a safer setting for the long-term therapeutic expression of RPE65. Subretinal injection of rAAV2 pseudotyped with serotype 4 (rAAV2/4) specifically targets the RPE. The purpose of our study was to evaluate a rAAV2/4 vector carrying a human RPE65cDNA driven by a human RPE65 promoter, for the ability to restore vision in RPE65−/− purebred Briard dogs and to assess the safety of gene transfer with respect to retinal morphology and function. rAAV2/4 and rAAV2/2 vectors containing similar human RPE65 promoter and cDNA cassettes were generated and administered subretinally in eight affected dogs, ages 8–30 months (n=6 with rAAV2/4, n=2 with rAAV2/2). Although fluorescein angiography and optical coherence tomography examinations displayed retinal abnormalities in treated retinas, electrophysiological analysis demonstrated that restoration of rod and cone photoreceptor function started as soon as 15 days post-injection, reaching maximal function at 3 months post-injection, and remaining stable thereafter in all animals treated at 8–11 months of age. As assessed by the ability of these animals to avoid obstacles in both dim and normal light, functional vision was restored in the treated eye, whereas the untreated contralateral eye served as an internal control. The dog treated at a later age (30 months) did not recover retinal function or vision, suggesting that there might be a therapeutic window for the successful treatment of RPE65−/− dogs by gene replacement therapy.

Collaboration


Dive into the Robin R. Ali's collaboration.

Top Co-Authors

Avatar

James W. Bainbridge

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Alexander J. Smith

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Jane C. Sowden

UCL Institute of Child Health

View shared research outputs
Top Co-Authors

Avatar

Rachael A. Pearson

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Yanai Duran

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Ulrich F.O. Luhmann

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aj Thrasher

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Emma L. West

UCL Institute of Ophthalmology

View shared research outputs
Researchain Logo
Decentralizing Knowledge