Network


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

Hotspot


Dive into the research topics where Jean M. Lawrence is active.

Publication


Featured researches published by Jean M. Lawrence.


PLOS ONE | 2009

Protective Effects of Human iPS-Derived Retinal Pigment Epithelium Cell Transplantation in the Retinal Dystrophic Rat

Amanda-Jayne F. Carr; Anthony Vugler; Sherry T. Hikita; Jean M. Lawrence; Carlos Gias; Li Li Chen; David E. Buchholz; Ahmad Ahmado; Ma’ayan Semo; Matthew J. K. Smart; Shazeen M. Hasan; Lyndon da Cruz; Lincoln V. Johnson; Dennis O. Clegg; Peter J. Coffey

Transformation of somatic cells with a set of embryonic transcription factors produces cells with the pluripotent properties of embryonic stem cells (ESCs). These induced pluripotent stem (iPS) cells have the potential to differentiate into any cell type, making them a potential source from which to produce cells as a therapeutic platform for the treatment of a wide range of diseases. In many forms of human retinal disease, including age-related macular degeneration (AMD), the underlying pathogenesis resides within the support cells of the retina, the retinal pigment epithelium (RPE). As a monolayer of cells critical to photoreceptor function and survival, the RPE is an ideally accessible target for cellular therapy. Here we report the differentiation of human iPS cells into RPE. We found that differentiated iPS-RPE cells were morphologically similar to, and expressed numerous markers of developing and mature RPE cells. iPS-RPE are capable of phagocytosing photoreceptor material, in vitro and in vivo following transplantation into the Royal College of Surgeons (RCS) dystrophic rat. Our results demonstrate that iPS cells can be differentiated into functional iPS-RPE and that transplantation of these cells can facilitate the short-term maintenance of photoreceptors through phagocytosis of photoreceptor outer segments. Long-term visual function is maintained in this model of retinal disease even though the xenografted cells are eventually lost, suggesting a secondary protective host cellular response. These findings have identified an alternative source of replacement tissue for use in human retinal cellular therapies, and provide a new in vitro cellular model system in which to study RPE diseases affecting human patients.


Experimental Neurology | 2008

Elucidating the phenomenon of HESC-derived RPE: Anatomy of cell genesis, expansion and retinal transplantation

Anthony Vugler; Amanda-Jayne F. Carr; Jean M. Lawrence; Li Li Chen; Kelly Burrell; Andrew Wright; Peter Lundh; Ma'ayan Semo; Ahmad Ahmado; Carlos Gias; Lyndon da Cruz; Harry Moore; Peter W. Andrews; James Walsh; Peter J. Coffey

Healthy Retinal Pigment Epithelium (RPE) cells are required for proper visual function and the phenomenon of RPE derivation from Human Embryonic Stem Cells (HESC) holds great potential for the treatment of retinal diseases. However, little is known about formation, expansion and expression profile of RPE-like cells derived from HESC (HESC-RPE). By studying the genesis of pigmented foci we identified OTX1/2-positive cell types as potential HESC-RPE precursors. When pigmented foci were excised from culture, HESC-RPE expanded to form extensive monolayers, with pigmented cells at the leading edge assuming a precursor role: de-pigmenting, proliferating, expressing keratin 8 and subsequently re-differentiating. As they expanded and differentiated in vitro, HESC-RPE expressed markers of both developing and mature RPE cells which included OTX1/2, Pax6, PMEL17 and at low levels, RPE65. In vitro, without signals from a developing retinal environment, HESC-RPE could produce regular, polarised monolayers with developmentally important apical and basal features. Following transplantation of HESC-RPE into the degenerating retinal environment of Royal College of Surgeons (RCS) dystrophic rats, the cells survived in the subretinal space, where they maintained low levels of RPE65 expression and remained out of the cell cycle. The HESC-RPE cells responded to the in vivo environment by downregulating Pax6, while maintaining expression of other markers. The presence of rhodopsin-positive material within grafted HESC-RPE indicates that in the future, homogenous transplants of this cell type may be capable of supporting visual function following retinal dystrophy.


Stem Cells | 2007

MIO‐M1 Cells and Similar Müller Glial Cell Lines Derived from Adult Human Retina Exhibit Neural Stem Cell Characteristics

Jean M. Lawrence; Shweta Singhal; Bhairavi Bhatia; David J. Keegan; Thomas A. Reh; Philip J. Luthert; Peng T. Khaw; G. A. Limb

Growing evidence suggests that glial cells may have a role as neural precursors in the adult central nervous system. Although it has been shown that Müller cells exhibit progenitor characteristics in the postnatal chick and rat retinae, their progenitor‐like role in developed human retina is unknown. We first reported the Müller glial characteristics of the spontaneously immortalized human cell line MIO‐M1, but recently we have derived similar cell lines from the neural retina of several adult eye donors. Since immortalization is one of the main properties of stem cells, we investigated whether these cells expressed stem cell markers. Cells were grown as adherent monolayers, responded to epidermal growth factor, and could be expanded indefinitely without growth factors under normal culture conditions. They could be frozen and thawed without losing their characteristics. In the presence of extracellular matrix and fibroblast growth factor‐2 or retinoic acid, they acquired neural morphology, formed neurospheres, and expressed neural stem cell markers including βIII tubulin, Sox2, Pax6, Chx10, and Notch 1. They also expressed markers of postmitotic retinal neurons, including peripherin, recoverin, calretinin, S‐opsin, and Brn3. When grafted into the subretinal space of dystrophic Royal College of Surgeons rats or neonatal Lister hooded rats, immortalized cells migrated into the retina, where they expressed various markers of retinal neurons. These observations indicate that adult human neural retina harbors a population of cells that express both Müller glial and stem cell markers and suggest that these cells may have potential use for cell‐based therapies to restore retinal function.


Progress in Retinal and Eye Research | 2001

Cell transplantation as a treatment for retinal disease.

Raymond D. Lund; Anthony Kwan; David Keegan; Yves Sauvé; Peter J. Coffey; Jean M. Lawrence

It has been shown that photoreceptor degeneration can be limited in experimental animals by transplantation of fresh RPE to the subretinal space. There is also evidence that retinal cell transplants can be used to reconstruct retinal circuitry in dystrophic animals. Here we describe and review recent developments that highlight the necessary steps that should be taken prior to embarking on clinical trials in humans.


Journal of Leukocyte Biology | 2003

Retinal transplantation: progress and problems in clinical application

Raymond D. Lund; Santa Jeremy Ono; David Keegan; Jean M. Lawrence

There is currently no real treatment for blinding disorders that stem from the degeneration of cells in the retina and affect at least 50 million individuals worldwide. The excitement that accompanied the first studies showing the potential of retinal cell transplantation to alleviate the progress of blindness in such diseases as retinitis pigmentosa and age‐related macular degeneration has lost some of its momentum, as attempts to apply research to the clinic have failed so far to provide effective treatments. What these studies have shown, however, is not that the approach is flawed but rather that the steps that need to be taken to achieve a viable, clinical treatment are many. This review summarizes the course of retinal transplant studies and points to obstacles that still need to be overcome to improve graft survival and efficacy and to develop a protocol that is effective in a clinical setting. Emphasis is given particularly to the consequences of introducing transplants to sites that have been considered immunologically privileged and to the role of the major histocompatibility complex classes I and II molecules in graft survival and rejection.


Stem Cells | 2008

Chondroitin Sulfate Proteoglycans and Microglia Prevent Migration and Integration of Grafted Müller Stem Cells into Degenerating Retina

Shweta Singhal; Jean M. Lawrence; Bhairavi Bhatia; J. S. Ellis; Anthony Kwan; Angus MacNeil; Philip J. Luthert; James W. Fawcett; Maria-Thereza R. Perez; Peng T. Khaw; G. Astrid Limb

At present, there are severe limitations to the successful migration and integration of stem cells transplanted into the degenerated retina to restore visual function. This study investigated the potential role of chondroitin sulfate proteoglycans (CSPGs) and microglia in the migration of human Müller glia with neural stem cell characteristics following subretinal injection into the Lister hooded (LH) and Royal College of Surgeons (RCS) rat retinae. Neonate LH rat retina showed minimal baseline microglial accumulation (CD68‐positive cells) that increased significantly 2 weeks after transplantation (p < .001), particularly in the ganglion cell layer (GCL) and inner plexiform layer. In contrast, nontransplanted 5‐week‐old RCS rat retina showed considerable baseline microglial accumulation in the outer nuclear layer (ONL) and photoreceptor outer segment debris zone (DZ) that further increased (p < .05) throughout the retina 2 weeks after transplantation. Marked deposition of the N‐terminal fragment of CSPGs, as well as neurocan and versican, was observed in the DZ of 5‐week‐old RCS rat retinae, which contrasted with the limited expression of these proteins in the GCL of the adult and neonate LH rat retinae. Staining for CSPGs and CD68 revealed colocalization of these two molecules in cells infiltrating the ONL and DZ of the degenerating RCS rat retina. Enhanced immune suppression with oral prednisolone and intraperitoneal injections of indomethacin caused a reduction in the number of microglia but did not facilitate Müller stem cell migration. However, injection of cells with chondroitinase ABC combined with enhanced immune suppression caused a dramatic increase in the migration of Müller stem cells into all the retinal cell layers. These observations suggest that both microglia and CSPGs constitute a barrier for stem cell migration following transplantation into experimental models of retinal degeneration and that control of matrix deposition and the innate microglial response to neural retina degeneration may need to be addressed when translating cell‐based therapies to treat human retinal disease.


Mechanisms of Development | 2007

Embryonic stem cells and retinal repair

Anthony Vugler; Jean M. Lawrence; James Walsh; Amanda Carr; Carlos Gias; Ma’ayan Semo; Ahmad Ahmado; Lyndon da Cruz; Peter W. Andrews; Peter J. Coffey

In this review we examine the potential of embryonic stem cells (ESCs) for use in the treatment of retinal diseases involving photoreceptors and retinal pigment epithelium (RPE). We outline the ontogenesis of target retinal cell types (RPE, rods and cones) and discuss how an understanding of developmental processes can inform our manipulation of ESCs in vitro. Due to their potential for cellular therapy, special emphasis is placed upon the derivation and culture of human embryonic stem cells (HESCs) and their differentiation towards a retinal phenotype. In terms of achieving this goal, we suggest that much of the success to date reflects permissive in vitro environments provided by established protocols for HESC derivation, propagation and neural differentiation. In addition, we summarise key factors that may be important for enhancing efficiency of retinal cell-type derivation from HESCs. The retina is an amenable component of the central nervous system (CNS) and as such, diseases of this structure provide a realistic target for the application of HESC-derived cellular therapy to the CNS. In order to further this goal, the second component of our review focuses on the cellular and molecular cues within retinal environments that may influence the survival and behaviour of transplanted cells. Our analysis considers both the potential barriers to transplant integration in the retina itself together with the remodelling in host visual centres that is known to accompany retinal dystrophy.


Experimental Eye Research | 2009

Distribution of Müller stem cells within the neural retina: evidence for the existence of a ciliary margin-like zone in the adult human eye.

Bhairavi Bhatia; Shweta Singhal; Jean M. Lawrence; Peng T. Khaw; G. Astrid Limb

Much interest has been generated by the identification of neural stem cells in the human neural retina and ciliary body. However, it is not clear whether stem cells identified in these ocular compartments are of the same origin or whether they ontogenically derive from different cell populations. This study examined the in situ anatomical distribution of these cells within the neural retina and ciliary body, as well as their ability to proliferate in response to EGF. Human retinae and ciliary body were examined for co-expression of Nestin, cellular retinaldehyde binding (CRALBP) or Vimentin, and the stem cell markers SOX2, CHX10, NOTCH1 and SHH. Retinal explants were cultured with epidermal growth factor (EGF) to assess retinal cell proliferation. Intense Nestin and CRALBP staining was observed in the neural retinal margin, where cells formed bundles of spindle cells (resembling glial cells) that lacked lamination and co-stained for SOX2, CHX10 and SHH. This staining differentiated the neural retina from the ciliary epithelium, which expressed SOX2, CHX10 and NOTCH1 but not Nestin or CRALBP. Nestin and CRALBP expression decreased towards the posterior retina, where it anatomically identified a population of Müller glia. All Vimentin positive Müller glia co-stained for SOX2, but only few Vimentin positive cells expressed Nestin and SOX2. Cells of the retinal margin and the inner nuclear layer (INL), where the soma of Müller glia predominate, re-entered the cell cycle upon retinal explant culture with EGF. Lack of lamination and abundance of Müller glia expressing stem cell markers in the marginal region of the adult human retina resemble the ciliary marginal zone (CMZ) of fish and amphibians. The findings that cells in this CM-like zone, as well in the inner nuclear layer proliferate in response to EGF suggest that the adult human retina has regenerative potential. Identification of factors that may promote retinal regeneration in the adult human eye would provide efficient treatments for retinal degenerative conditions for which treatments are not yet available.


Experimental Neurology | 2007

Dopamine neurones form a discrete plexus with melanopsin cells in normal and degenerating retina.

Anthony Vugler; Peter Redgrave; Ma'ayan Semo; Jean M. Lawrence; John Greenwood; Peter J. Coffey

In addition to rods and cones of the outer retina, a third class of photoreceptive cell has recently been described in the inner retina of mammals. These intrinsically photosensitive retinal ganglion cells (ipRGCs) have been shown to relay luminance information to the mammalian brain. In addition to their intrinsic photosensitivity, the function of ipRGCs may also be modulated by signals from within the retina itself. Such signals may emanate from classical photoreceptors in the outer retina or from the circadian activity of adjacent inner retinal neurones. Prime candidates for the latter are the retinal dopamine neurones which ramify at the border of the inner plexiform and inner nuclear layers. In order to investigate the nature of any interaction between dopamine and ipRGC populations in normal retina and to assess the impact of outer retinal degeneration on this interrelationship, we examined the retinae of normal and RCS dystrophic rats. We report a direct interaction between the dendrites of ipRGCs and dopaminergic neurones which is conserved across species. Triple immunolabelling using synaptic markers provides evidence for the unidirectionality of information transfer between the two cell types, with processes of ipRGCs being directly adjacent to sites of dopamine release. This fundamental architectural feature of the mammalian retina appears resistant to degeneration of classical photoreceptors and may provide the anatomical substrate by which dopamine cells influence the physiology of central circadian targets in the brain.


PLOS ONE | 2010

Dissecting a role for melanopsin in behavioural light aversion reveals a response independent of conventional photoreception.

Ma'ayan Semo; Carlos Gias; Ahmad Ahmado; Eriko Sugano; Annette E. Allen; Jean M. Lawrence; Hiroshi Tomita; Peter J. Coffey; Anthony Vugler

Melanopsin photoreception plays a vital role in irradiance detection for non-image forming responses to light. However, little is known about the involvement of melanopsin in emotional processing of luminance. When confronted with a gradient in light, organisms exhibit spatial movements relative to this stimulus. In rodents, behavioural light aversion (BLA) is a well-documented but poorly understood phenomenon during which animals attribute salience to light and remove themselves from it. Here, using genetically modified mice and an open field behavioural paradigm, we investigate the role of melanopsin in BLA. While wildtype (WT), melanopsin knockout (Opn4−/−) and rd/rd cl (melanopsin only (MO)) mice all exhibit BLA, our novel methodology reveals that isolated melanopsin photoreception produces a slow, potentiating response to light. In order to control for the involvement of pupillary constriction in BLA we eliminated this variable with topical atropine application. This manipulation enhanced BLA in WT and MO mice, but most remarkably, revealed light aversion in triple knockout (TKO) mice, lacking three elements deemed essential for conventional photoreception (Opn4−/− Gnat1−/− Cnga3−/−). Using a number of complementary strategies, we determined this response to be generated at the level of the retina. Our findings have significant implications for the understanding of how melanopsin signalling may modulate aversive responses to light in mice and humans. In addition, we also reveal a clear potential for light perception in TKO mice.

Collaboration


Dive into the Jean M. Lawrence's collaboration.

Top Co-Authors

Avatar

Peter J. Coffey

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Anthony Vugler

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar

Ahmad Ahmado

University College London

View shared research outputs
Top Co-Authors

Avatar

Li Li Chen

University College London

View shared research outputs
Top Co-Authors

Avatar

Ma'ayan Semo

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carlos Gias

UCL Institute of Ophthalmology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Keegan

University College London

View shared research outputs
Top Co-Authors

Avatar

James Walsh

University of Sheffield

View shared research outputs
Researchain Logo
Decentralizing Knowledge