retinitis pigmentosa case

16
OPTOMETRY I INVITED REVIEW I Retinitis pigmentosa: understanding the clinical presentation, mechanisms and treatment options Michael Kalloniatis' PhD Erica L Fletcher' PhD * Department of Optometry and Vision Science, University of Auckland, New Zealand The University of Melbourne, Australia Department ofAnatomy and Cell Biology, Submitted: 19 January 2004 Revised: 9 February 2004 Accepted for publication: 9 February 2004 Clin Ex$ Optom 2004; 87: 2: 65-80 Retinitis pigmentosa (RP) is a leading cause of human blindness due to degeneration of retinal photoreceptor cells. Causes of retinal degeneration include defects in the visual pigment, defects in the proteins important for photoreceptor function or in enzymes involved in initiating visual transduction. Despite the diversity of genetic muta- tions identified in inherited forms of retinal dystrophy, there is a common end result of photoreceptor death and functional blindness. In this review, pertinent anatomical and physiological pathways involved in RP and the underlying genetic mutations are out- lined, including a discussion on the inheritance patterns revealed by advances in molecular biological techniques. Characteristics of progression rates of visual field loss and current management options will provide useful clinical guidelines for the man- agement of patients with RP. Key words: cGMP, degeneration, gene mutations, retina, retinal transplants, retinitis pigmentosa, visual field, vitamin A RETINAL STRUCTURE/FUNCTION AS IT RELATES TO IDENTIFIED GENE MUTATIONS IN RETINITIS PIGMENTOM Retinitis pigmentosa (RP) reflects a heter- ogenous group of inherited ocular diseases representing the most recurrent retinal dystrophies, with a worldwide prevalence of 1:3000 to 1:5000.' Inherited forms of reti- nal degeneration are largely focused on gene mutations within photoreceptors or WE cells leading to devastatingloss ofvisual function, often progressing to functional blindness. The chief gene mutations lead- ing to the RP phenotype24relate to defects in the activation/de-activation of the visual pigment or pathways involved in the visual phototransduction cascade. To better man- age patients with RP, it is important to review the basic structure of the retina and the key pathways involved in the visual response affected in the disease process. The visual process begins with light absorption in the photoreceptor outer seg- ment with subsequent encoding occurring through interactions between the five other major classes of retinal ne~lrons.~.~ Photoreceptor cells (rods and cones), bi- polar cells and ganglion cells form the 'through' pathway, while horizontal cells, amacrine cells and interplexiform cells form the 'lateral' or 'modulatory' pathways (Figure 1). The complex interplay be- tween the different retinal neurons leads to the encoding of spatial, temporal and chromatic information that is subse- quently relayed by ganglion cells to the central visual pathway~.~.'-'I In addition to neural cells, several glial cells are localised in the retina, with the major retinal glial cell being the Miiller cell" (Figure 1). Mtller cells are intimately involved in a range of activities essential for normal reti- nal functions that include neurotransmit- ter and metabolite metabolism, and chromophore recycling. The retinal pig- ment epithelial (RPE) cells are considered part of the retina and are involved in a multitude of functions essential for neu- ral retinal function. These include the transport and storage of vitamin A deriva- tives (retinoids), recycling of the visual pig- ment, outer photoreceptor segment phagocytosis and anti-oxidant functions.'J3 Activation and deactivation of the visual pigment The visual pigment within the photorecep tor outer segment is composed of an in- Clinical and Experimental Optometry 87.2 March 2004 65

Upload: envad

Post on 16-Dec-2015

243 views

Category:

Documents


0 download

DESCRIPTION

makalah retinitis

TRANSCRIPT

  • OPTOMETRY

    I INVITED REVIEW I

    Retinitis pigmentosa: understanding the clinical presentation,

    mechanisms and treatment options

    Michael Kalloniatis' PhD Erica L Fletcher' PhD * Department of Optometry and Vision Science, University of Auckland, New Zealand

    The University of Melbourne, Australia Department ofAnatomy and Cell Biology,

    Submitted: 19 January 2004 Revised: 9 February 2004 Accepted for publication: 9 February 2004

    Clin Ex$ Optom 2004; 87: 2: 65-80

    Retinitis pigmentosa (RP) is a leading cause of human blindness due to degeneration of retinal photoreceptor cells. Causes of retinal degeneration include defects in the visual pigment, defects in the proteins important for photoreceptor function or in enzymes involved in initiating visual transduction. Despite the diversity of genetic muta- tions identified in inherited forms of retinal dystrophy, there is a common end result of photoreceptor death and functional blindness. In this review, pertinent anatomical and physiological pathways involved in RP and the underlying genetic mutations are out- lined, including a discussion on the inheritance patterns revealed by advances in molecular biological techniques. Characteristics of progression rates of visual field loss and current management options will provide useful clinical guidelines for the man- agement of patients with RP.

    Key words: cGMP, degeneration, gene mutations, retina, retinal transplants, retinitis pigmentosa, visual field, vitamin A

    RETINAL STRUCTURE/FUNCTION AS IT RELATES TO IDENTIFIED GENE MUTATIONS IN RETINITIS PIGMENTOM

    Retinitis pigmentosa (RP) reflects a heter- ogenous group of inherited ocular diseases representing the most recurrent retinal dystrophies, with a worldwide prevalence of 1:3000 to 1:5000.' Inherited forms of reti- nal degeneration are largely focused on gene mutations within photoreceptors or W E cells leading to devastating loss ofvisual function, often progressing to functional blindness. The chief gene mutations lead- ing to the RP phenotype24 relate to defects in the activation/de-activation of the visual pigment or pathways involved in the visual phototransduction cascade. To better man- age patients with RP, it is important to review

    the basic structure of the retina and the key pathways involved in the visual response affected in the disease process.

    The visual process begins with light absorption in the photoreceptor outer seg- ment with subsequent encoding occurring through interactions between the five other major classes of retinal n e ~ l r o n s . ~ . ~ Photoreceptor cells (rods and cones), bi- polar cells and ganglion cells form the 'through' pathway, while horizontal cells, amacrine cells and interplexiform cells form the 'lateral' or 'modulatory' pathways (Figure 1). The complex interplay be- tween the different retinal neurons leads to the encoding of spatial, temporal and chromatic information that is subse- quently relayed by ganglion cells to the central visual pathway~.~.'-'I In addition to neural cells, several glial cells are localised

    in the retina, with the major retinal glial cell being the Miiller cell" (Figure 1). Mtller cells are intimately involved in a range of activities essential for normal reti- nal functions that include neurotransmit- ter and metabolite metabolism, and chromophore recycling. The retinal pig- ment epithelial (RPE) cells are considered part of the retina and are involved in a multitude of functions essential for neu- ral retinal function. These include the transport and storage of vitamin A deriva- tives (retinoids), recycling of the visual pig- ment , outer photoreceptor segment phagocytosis and anti-oxidant functions.'J3

    Activation and deactivation of the visual pigment The visual pigment within the photorecep tor outer segment is composed of an in-

    Clinical and Experimental Optometry 87.2 March 2004

    65

  • Retinitis pigmentosa Kulloniutis and Fletcher

    ing of Mata and colleagues14 that all trans retinol to 11-cis retinol conversion can occur in Miiller cells, explains the reason why MUer cells have long been known to contain retinoid binding protein^.'^.'^ With the 11-cis retinol available to cone photoreceptors, pigment regeneration can occur after cones convert 1 1 4 s retinol to 1 l-cis retinal: a reaction known to occur in isolated cone photoreceptors but not in rod photo receptor^.'^

    Figure 1. Schematic of the mammalian retina showing the different retinal layers and the rod and cone circuitry. The cone pathway involves a unique set of cone bipolar cells that synapse onto ganglion cells. The signal in both the rod and cone pathway is modified by horizontal cells in the OPL and amacrine cells in the IPL. The rod pathway has one type of bipolar cell that synapses with a unique amacrine cell (MI amacrine cell), which subsequently passes the information to the cone pathway via conventional synapses or via gap junctions (gi) with cone bipolar cells. These latter cells synapse onto ganglion cells and hence, ganglion cells carry both the rod and cone signals. Miiller cells span the entire retina. The blood supply in most mammalian retinas involves the outer blood supply from the choriocapillaris and the inner blood supply through the central retinal artery. The inner blood supply has capillary beds in both the inner and outer plexiform layers (for simplicity they are not shown in this diagram). The locations of the outer and inner retinal blood vessels imply that the mid-retina has poor vascular coverage. Abbreviations: ORBV = outer retinal blood vessels, RL = receptor layer, OPL = outer plexiform layer, INL = inner nuclear layer, IPL = inner plexiform layer, GCL = ganglion cell layer, NFL = nerve fibre layer, IRBV = inner retinal blood vessels, HC = horizontal cell, BC = bipolar cell, AC = amacrine cell, HCCB = horizontal cell cell body, HC-AT = horizontal cell axon terminal, GC = ganglion cell

    trinsic membrane protein (opsin) and a chromophore (vitamin-A derivative). Light activation leads to a conformational change in the chromophore (1 l-cis to all- trans conformational change), that initi- ates a series of protein changes within the opsin closing the ion channels in the outer segment (Figure 2). The activated form of the visual pigment (Rh* for rhodopsin) , must be deactivated to ensure that subse- quent light can be detected. The rho- dopsin deactivation process begins by

    multiple phosphorylations catalysed by rhodopsin kinase, followed by the binding of a protein (arrestin) that terminates the activation properties of rhodopsin.

    Regeneration of the photopigment Subsequent to the phosphorylation and arrestin binding, the chromophore (all- trans retinal) detaches from the opsin and begins its regeneration via the RPE or Miiller cells'4 (Figures 3 and 4). The find-

    Phototransduction cascade Activation of the visual pigment (Rh* in rod photoreceptors or the long- middle- or short-wavelength photopigment for cone photoreceptors), allows for the bind- ing of a series of membrane-associated proteins involved in the visual transduc- tion cascade. The first protein to bind, transducin, is in the Gfamily of proteins, which are often associated with processes linked with signal amplification. The bind- ing and activation of transducin subse- quently leads to activation of phosphodi- esterase (PDE): the enzyme hydrolysing 3' 5' cyclic guanyl monophosphate (cGMP), the intermediary compound controlling cation ion entry in the photoreceptor outer segment. The hydrolysis of cGMP leads to closure of cation channels with a subsequent photoreceptor hyperpolarisa- tion to a light The produc- tion of cGMP is dependent on the enzyme guanylate cyclase, the activity of which is modulated by a calcium-sensitive enzyme, guanylate cyclase activation protein (GCAP: also referred to as recoverin). Guanylate cyclase maintains a steady state level of cGMP with the activity of this en- zyme markedly increasing secondary to GCAP activation (subsequent to calcium concentration changes in the photorecep tor outer segment2u-22 (Figure 4).

    Rhodopsin gene mutations cause the majority of RP, although gene mutations in different catalytic units of PDE, peripherin and the cyclic nucleotide gated channel proteins are also known to cause RP. Gene mutations alter proteins such as transducin, rhodopsin kinase, arrestin, and proteins forming the voltage gated calcium channel. A few rhodopsin gene mutations cause congenital stationary

    Clinical and Experimental Optometry 87.2 March 2004

    66

  • Retinitis pigmentosa Kalloniatis and Fletcher

    night blindness; gene mutations in guanylate cyclase may cause cone dystro- phy or a form of Lebers congenital amau- rosis; whereas a well-defined gene muta- tion in the RPE (RPE65) causes another form of Lebers congenital amaurosis; the gene encoding the alpha subunit of the cone cGMP gated channel is mutated in rod monochromacy. Other gene muta- tions may cause syndromes associated with RP that include Laurence-Moon/Bardet- Biedl and Ushers syndrome as well as other RP-like conditions involving meta- bolic defects such as Refsums disease. Recent reviews on the molecular biology of RP and other inherited retinal dystrophies are provided by several arti- cless.2328 and Daiger and co-worker? main- tain an up-to-date website at the address http://www.sph.uth.tmc.edu/Retnet/, which outlines genes causing inherited reti- nal dystrophies. In addition, clinical trial information can he obtained from the National Institutes of Health (USA) address http://clinicaltrials.gov/ct/gui.

    RP INHERITANCE PAITERNS AND CLINICAL CHARACTERISTICS

    RP inheritance patterns The advent of molecular techniques has lead to the identification of a number of genes that are thought to he causally linked to the onset of RP. This suggests that there is a number of possible genetic origins, all of which result in the same degenerative process. As a general rule, retinal conditions that demonstrate a bilateral involvement, loss of peripheral vision, predominantly rod dysfunction (elevated rod threshold shown by electo- physiological methods or psychophysi- cally) and progression of photoreceptor dysfunction are classified as RP. Clini- cal presentation may he varied hut include symptoms of night blindness (nyctalopia) with elevated dark adaptation thresholds (predominantly rod hut also cone thresh- olds), abnormal electroretinographic a- and h-wave, difficulty with mobility sec- ondary to visual field constriction, ac- quired blue-yellow (tritan) colour vision defect, abnormal retinal pigmentation,

    Figure 2. Simplified photoreceptor outer segment under dark and light adaptation. The non- specific cationic channel is maintained in the open state by cGMP, allowing sodium, calcium and magnesium (not shown) ions to enter the outer segment. Light leads to activation of rhodopsin (R), tranducin (T) and phosphodiesterase (the different subunits are shown with the alpha and beta identified), causing hydrolysis of cCMP to GMP, closing the cationic channels and driving the photoreceptor away for the equilibrium potential of these ions (hyperpolarising the photoreceptor).

    including mid-peripheral hone spicules, arterial narrowing; optic nerve pallor, pre- disposition to myopia, posterior subcap- sular cataract and vitreous changes.30-3z Visual disturbances occur when visual acu- ity is relatively unaffected and careful choice of colour vision tests may reveal the acquired tritan colour vision defect early in the disease pro~ess.~~~ M o st of the ocular signs noted earlier are evident in more advanced stages of the condition.

    Massof and associate^^^ reported on the existence of two general categories of RP patients dependen t on the relative involvement of rod and cone photo- receptors. Patients with Type 1 RP display an early diffuse and preferential loss of rod sensitivity (hence early nyctalopia) and later progression and regionalised loss of visual field. Patients with Type 2 RP have a regionalised and progressive combined loss of rod and cone sensitivity with late difficulty with night vision, typically in adulthood. The inheritance patterns in RP are classified into autosomal dominant, X-linked, simplex and multiple^.'^^^^^^ Heckenlively, Boughman and Friedmansfi

    averaged data from five studies (n = 2,406) and showed that RP could he classified as autosomal recessive in about 31 per cent of cases, autosomal dominant in approxi- mately 16 per cent, X-linked in around nine per cent and simplex/multiplex in approximately 44 per cent. Calculations from the Danish RP register showed that the prevalence and distribwion were not significantly different from those pub- lished from around the world, with auto- soma1 recessive (18.8 per cent), autosomal dominant (8.4per cent), X-linked (14.4 per cent), simplex (43 per cent), multiplex (11.2 per cent), with 4.2 per cent being unclassified. The prevalence rate for all forms of RP in Denmark was 1:3,333 to 4,000 for females and 1:2,500 to 2,857 for males. It is likely that the prevalence of the various forms of RP within the Australian and New Zealand populations are similar.

    Autosomal dominant classification is primarily based on an established parent- to-child transmission. Autosomal recessive classification is based on established inher- itance within different family groups consistent with Mendelian characteristics.

    Clinical and Experimental Optometry 87.2 March 2004

    67

  • Retinitis pigmentosa Kalloniatis and Fletcher

    Figure 3. A schematic of the rhodopsin activation, deactivation and the recycling of the visual pigment. The recycling of the chromophore for cone photoreceptors involving retinoid transport and recycling through Miiller cells (Mata and colleaguees") is not shown. Rhodopsin is an integral membrane protein with an amino terminal (N-) and a carboxyl terminal (C), where the binding of tranducin occurs. Activated rhodopsin (RW) is deactivated via the action of rhodopsin kinase (Rh-kinase) causing multiple phosphorylations (P(n)) and the binding of arrestin leads to separation of the chromophore from the opsin. The chromophore, in the form of all-trans retinal is converted to all-trans-retinol before leaving the retina to be converted to 1 l-cis-retinol and 1 14s-retinal, before reentering the photoreceptor outer segment to bind to the opsin. The retinoids in the RPE also arrive via the complex containing retinoid binding protein (RBP), thransthyretin (Tr) and all-trans retinol (AT-retinol). Retinoids can also be stored in the form of retinyl esters. The scheme of the RPE and rod photoreceptor has the cyclic nucleotide channels identified by cG and the voltage gated calcium channels in the axon terminal by vCa. Other abbreviations: RPE = retinal pigment epithelium, 0s = outer segment, IS = inner segment, N = nucleus, AT = axon terminal

    X-linked is also based on established fam- ily inheritance based on Mendelian char- acteristics. Simplex reflects isolated cases with one affected member and multiplex where at least two family members are affected. Often, simplex and multiplex cases were thought to reflect autosomal recessive inheritance patterns, however, when simplex and multiplex cases are taken together, there are too many sim- plex cases compared with predictions by Mendelian genetic^.^^^^^^'^^* FishmanSg

    reported 60 per cent with a known Men- delian inheritance pattern, whereas in a recent Danish study,' 40 per cent of the patients were characterised by a known inheritance pattern. Considering that it is estimated that one in 50 carries a gene for the recessive form of RP,40 a key consid- eration in this classification is the exclu- sion of consanguinity. Consanguinity would significantly increase the odds of carrying the same mutated gene.'

    A new evolving concept in the inherit-

    ance of many conditions, including some forms of RP or syndromes associated with RP (Usher type 1; RDS/ROMl; Bardet- Biedl), is referred to as ~ l i g o g e n i c . ~ ~ Poly- genic traits are disorders that occur through poorly understood interactions between many genes and the environ- ment. Such an environmental interaction could explain the higher correlation be- tween perinatal stress and some simplex forms of RF' reported by Stone and col- l e a g u e ~ . ~ * , ~ ~ The authors suggest the existence of a non-hereditary explanation for photoreceptor death (such as peri- natal stress), for some simplex RP. This suggestion awaits further experimental evi- dence and the exclusion of polygenic and oligogenic inheritance. Oligogenic traits are primarily genetic in nature but require synergistic action of mutant alleles at a small number of loci to exhibit the phe- n ~ t y p e . ~ ' For example, if heterozygous mutations in either the RDS gene or the ROMl gene were present, individuals were asymptomatic, whereas if gene mutations were evident in both the RDS and ROMl gene, patients would exhibit the RP phe- notype despite the fact that the mutations were in two distinct genes.49 In addition, it may be possible to modify the expres- sion of an autosomal dominant gene by the effect of a mutation in a second or third gene.41

    In the classical analysis of inheritance patterns in RP, when considering the high number of simplex and multiplex cases, variously the typical RP phenotype does not follow classical Mendelian inheritance patterns, the expressivity or penetrance is altered,',35 or other non-genetic mecha- nisms are in p l a ~ e . ~ ~ * ~ ' For example, we now know that human subjects with spe- cific gene mutations such as that affecting a photoreceptor structural protein peripherin (peripherin/RDS gene muta- tion), display marked variations in the age of onset and progression of the disease and exhibit oligogenic inheritance patterns, offering one explanation for the high prevalence of simplex and multiplex cases in RP.23*43*44 Wang and associates45 outlined the following clinical implications of gene mutations. Different gene mutations of the same gene result in different clinical pres-

    Clinical and Experimental Optometry 87.2 March 2004

    68

  • Retinitis pigmentosa Kalloniatis and Fletcher

    entations, (for example, different muta- tions in the rhodopsin gene give rise to different clinical presentations: allelic het- erogeneity); mutations in different genes lead to the same disease phenotype, (for example, mutations in peripherin/RDS and rhodopsin gene both lead to typical RP: non-allelic heterogeneity); mutations in different genes at different loci lead to the same phenotype (for example, in Ush- er's syndrome: locus heterogeneity); the expression is different for the same ge- netic defect (expressivity) ; the defective gene is not expressing leading to varia- tions in visual function (penetrance).45 The evolution of our understanding of polygenic and ohgogenic disorders and further work on potential environmental factors will allow us to better understand the high number of simplex/multiplex cases of RP.

    The examination of family members is essential to assist in the identification of the hereditary nature of the condition. In particular, patients with possible X-linked recessive RP require careful assessment of the mother. Dr Mary Lyon proposed that in every somatic cell of a female, only one X-chromosome is functioning, that is, one X-chromosome in every cell is randomly inactivated during development. The Lyon hypothesis predicts that every female carrier of X-linked RP will have two cell populations: one with normal activity and one with mutant a~tivity.~"~' Thus, in X- linked RP, female carriers should display signs and symptoms of RP; a common find- ing, although there is considerable vari- a b i l i t ~ . ~ " . ~ ~ Female carriers may display a tapetal reflex, irregular pigmentation in the posterior pole, WE atrophy and pig- ment stippling and may express a pheno- type typical of more advanced RP.4R-50

    Visual function in Rp Traditionally it has been thought that the progression of visual dysfunction is slow- est in those with autosomal dominant RP, followed by recessive (including simplex/ multiplex) RP and the fastest are those that are X-linked.35 The advent of molecu- lar biological techniques to classify RP pa- tients has provided useful insights to com- plement studies supporting this clinical

    Figure 4. Schematic of the key pathways controlling cGMP concentration. Activated rhodopsin (Rh*) activates transducin (T*), which binds the two gamma inhibitory subunits of phospodiesterase to activate the enzyme (PDE*) and hydrolyse cGMP. Steady state production of cGMP from guanyl tri-phosphate (GTP) is maintained by guanylate cyclase, the activity of which is controlled by the calcium-sensitive guanylate cyclase activation protein (GCAP).

    view5' and those that found no significant difference in progression between the dif- ferent genetic subgroups.52 In a major study of 172 typical RP patients followed from 2.5 to 10 years, Massof and col- l e a g u e ~ " ~ reported some key features of visual field changes measured with Goldmann perimetry. Two key parameters will be reviewed here: the time constant (tc) , reflecting the time taken to lose one half of the remaining visual field, and the critical age (Ac: extrapolated age of on- set). These parameters reflect photopic visual fields that represent cone loss sec- ondary to rod loss or concurrent rod and cone loss. I t follows that such measure- ments overestimate the age of onset of sco- topic visual field loss by an unknown amount because we do not know when rod photoreceptor degeneration begins in classical RP.

    When using bright small targets [Goldmann II/4e (0.22 degrees)], the time constant was found to be tc = 7.4 f 4.7 years (ISD range of 2.7 to 12.1 years); whereas the use of a bright larger target [Goldmann V/4e (1.72 degrees)] resulted

    in a time constant = 8.4 f 4.9 years ( E D range of 3.5 to 13.3 years) ..y5 There was no significant difference in the time constant for visual field progression across the dif- ferent genetic subtypes, providing support for the proposition that, on average, visual field progression is consistent for RP subtypes that are broadly classified. The time constants and ranges noted above provide useful guidelines for the likely pro- gression of patients with RP. The second parameter investigated by Massof and col- leaguess5 was the critical age value (that is, age of onset ofvisual field loss). For the Goldmann II/4e target, Ac was 22 f 13.8 years ( E D range 8.2 to 35.8 years) and for the Goldmann V/4e target, 28 f 13.9 years (1SD range of 14.1 to 41.9 years).35 Massof and c011eagues~~ report close to significant difference (p = 0.07) for differ- ences in type 1 versus type 2 RF', implying that the age of onset demonstrates some variability. The study of Haim' has a younger mean age of onset (six to18 years of age) but includes atypical RP cases. The data of Massof and colleaguess5 more accurately reflect the patient pool of RP

    Clinical and Experimental Optometry 87.2 March 2004

    69

  • Retinitis pigmentosa Kalloniatis and Fletcher

    patients likely to be encountered in opto- metric practice.

    Berson and associates53 followed 94 patients over three years and found visual field loss (approximately five per cent of field per year), which is half that predicted by Massof and colleague^.^^ The key find- ing was that a minimum of two years was required to follow visual fields. Visual acu- ity and dark adaptation ( 1 1 degrees cen- tral) remained unchanged over three years and 77 per cent showed significant reduc- tion in the electroretinogram (16 per cent amplitude loss per year). There was no sig- nificant difference in loss of electroretino- gram amplitude across genetic subtypes, supporting the view that broadly classified RP cases follow a similar time course in changes of visual function.

    With the advent of molecular biologi- cal genotyping, several studies have focused on visual function measurements in patients with specific gene mutations causing RP. Berson and c o - ~ o r k e r s ~ ~ stud- ied 17 cases with an autosomal dominant form of RP (P23H: proline to histidine sub- stitution at position 23 of the rhodopsin molecule) and found that median onset of night blindness was at 13 to 14 years. As a group, these patients displayed better visual acuity and larger ERG amplitudes compared with 131 subjects with other autosomal dominant rhodopsin gene mu- tations. However, there was considerable heterogeneity within families. At 37 years, visual fields ranged from greater than120 degrees to less than 21 degrees and Berson and co-~orkers '~ proposed that some pa- tients may have functional vision up to 70 years of age. In another form of rhodopsin autosomal dominant gene mutation (P347L: proline to leucine amino acid sub stitution at location 347), Berson and co- w o r k e r ~ ~ ~ reported considerable he teroge- neity within families with onset of night blindness often at early childhood (n = 8 cases). This group of patients with the P347L gene mutation displayed smaller ERG amplitudes and visual fields com- pared with 140 unrelated subjects with autosomal dominant rhodopsin gene mu- tations. Mobility was impaired between 12 and 27 years of age.55 Although the number of subjects in these original stud-

    ies was small, the characteristic of gene mutations involving the C-terminus (for example, P347L) of rhodopsin being more severe than that involving the N-terminus (for example, P23H) has held up in a sub- sequent study encompassing a spectrum of rhodopsin gene mutations at these two terminals.56 The G and N-terminus are il- lustrated in Figure 3. The same amino acid switch (proline to alanine) at two differ- ent locations (P23A versus P347A), re- sulted in a greater loss in all visual func- tions measured for the Gterminus located gene mutation (P347A).56 Gene mutations at loci encoding the C-terminal may im- pact function, for example, by slowing the deactivation of activated rhodopsin, whereas gene mutations at loci encoding the N-terminal impact on the delivery of rhodopsin to the outer segment (see be- low for further comments). The use of genetic phenotyping invariably will lead to a better understanding of the RP pheno- type including visual function progression characteristics.

    The following two case studies illustrate the usefulness of appreciating the inher- itance pattern and the use of appropriate visual function testing in routine clinical practice.

    years earlier with a 2/1000 white target were reported to be five degrees (R and L) . There was no detectable visual field in the mid-to-far periphery and automated visual fields were conducted to obtain the extent of the remaining visual fields (Figure 5h). The visual fields were largely confined to the central five degrees, although some more peripheral locations were detected. The visual fields have remained stable over a three-year follow up.

    Although this is an example of advanced RP, it illustrates several points. This patient displayed the characteristics of Type 2 RP with what appears to be regionalised and progressive combined loss of rod and cone sensitivity with late difficulty with night vision in adulthood. The age of presenta- tion secondary to functional mobility dif- ficulties was within the critical age predic- tion from the results of Massof and colleague^.^^ The fact that a male cousin was affected with a similar condition indi- cates that autosomal recessive may be a mode of inheritance, although the possi- bility of oligogenic inheritance should also be con~idered.~' On further questioning, it was ascertained that the parents of both affected members were related and there- fore consanguinity of the parents may be a factor in the inheritance pattern.

    CASE I

    A 44year-old female with typical RP was diagnosed at age 29 years after seeking an ophthalmic examination because of 'trip- ping over her children's toys'. Nyctalopia was not a problem at the time of diagno- sis. A first cousin (male) was also affected but there was no other family history re- ported (her parents and the parents of her affected cousin had normal vision). Her siblings were examined and were unaf- fected (Figure 5a). Visual acuity with spec- tacle correction was RE 6/18, LE 6/21 but corneal topography indicated a kerato- conus-like appearance and the patient was referred for a hard contact lens assessment and fitting. Visual acuity of 6/ 15 (R and L) was achieved with her new hard contact lenses and the patient gained a marked improvement in confidence in mobility and a perceptual improvement in contrast levels. Bjerrum visual fields conducted two

    CASE 2

    A 54year-old male with typical RP had been diagnosed at age 44 years. Nyctalo- pia was first noticed about 10 years before diagnosis. There was no history of consan- guinity and the patient had no siblings. The mother may have had abnormal vision (but was deceased), with one sister and one brother being normal. The patient complained of nyctalopia but avoided go- ing out at night. There was a detectable 30 Hz flicker ERG signal indicating cone function and mobility was excellent under photopic conditions. A previous report had noted that after automated perimetry, 'the patient has only 10 to 15 degrees of central vision'. The patient was referred for evaluation of dark adaptation and re- assessment of visual fields using Goldmann perimetry. Visual acuities were 6/6 R and L and dark adaptation thresholds were

    Clinical and Experimental Optometry 87.2 March 2004

    70

  • Retinitis pigmentosa Kalloniatis and Fletcher

    Patient#l A w

    Cousin

    Figure 5. Pedigree showing the affected members in two families (a). Automated visual field results of Patient 1 showing severe restriction to within the central five degrees (b).

    elevated approximately 0.8 log units for the cone component with no detectable rod function (that is, the rod dark adapta- tion thresholds were elevated by about four log units and the dark adaptation curve was monophasic). Goldmann W/4e (Figure 6 has the right eye results) showed an annular loss located only around the region of maximal rod density. Visual fields were normal under photopic conditions outside this range for both eyes.

    This patient also fits into the Type 2 RP with a critical age still within one standard deviation of the upper limit predictions5 of 42 years. With the mother potentially affected, the possible modes of inherit- ance include X-linked or autosomal domi- nant. Because the patient has no siblings and did not intend to procreate, genetic counselling was not required. The extent of the visual field should be evident from observing the mobility of the patient, with Goldmann perimetry providing quantita- tive data on the extensive remaining (photopic) visual field in this typical RP patient.

    Figure 6. Visual fields of Patient 2 using W/4e target on the Goldmann perimeter. The only area of absolute visual field loss under photopic conditions was an annular scotoma centring around the 15-degree meridian.

    Clinical and Experimental Optometry 87.2 March 2004

    71

  • Retinitis pigmentosa Kalloniatis and Fletcher

    HOW DO YOU MANAGE THE PATIENT WITH RETINITIS PIGMENTOSA?

    Most patients with RP are likely to present for an eye examination when night vision and/or mobility are affected and therefore the critical age estimate of Massof and col- leagueP is a useful guide. Early adulthood is the most likely presentation time for an optometric examination.

    The diagnosis should be confirmed by electrophysiology and genetic counselling should be undertaken, as well as low vision service delivery. Other family members should be examined, particularly the mother of a male RP sufferer who may have an X-linked inheritance pattern.

    Goldmann visual field assessment should be used (loss occurs in the mid- to far-periphery) and visual fields should be followed for a minimum of two years to estimate the time constant for visual field loss. An estimate of the visual field pro- gression rate is given by the time constant estimate of tc = 8.4 f 4.9 years when using the V/4e target.

    Time of onset of visual field loss can be combined with the time constant to ob- tain a reasonable predictor of remaining duration of functional photopic vision.

    When using broad classifications such as autosomal dominant or X-linked, genetic subtype is not a reasonable pre- dictor of severity and progression.

    MECHANISMS OF RETINAL DEGENERATION

    The location of the gene mutation leads to different functional deficits Mutations on the rhodopsin gene lead to autosomal dominant forms of retinitis pigmentosa in humans and a canine model, and introducing this human gene in the rat or mouse genome leads to transgenic rodent models of retinitis p i g r n e n t o ~ a ~ ~ . ~ ~ - ~ ~ (for example, P23H rat). Why a rhodopsin gene mutation causes retinal degeneration is unclear. Rhodopsin is an integral membrane protein that com- prises the bulk of the outer segment pro-

    teins. The gene mutation may change the tertiary structure of the protein and alter the structural role of rhodopsin.23 Using the P23H rat model of RP, Illing and asso- ciatesM showed that rhodopsin was prone to form high molecular weight oligomeric species in the cytoplasm of transfected cells, accumulating in aggresomes (pericentiolar inclusion bodies). These cellular changes make rhodopsin suscep- tible to degradation by the ubiquitin- dependent proteosome system. In effect, mutated P23H rhodopsin is a potential cytotoxin mimicking neurodegenerative diseases of the central nervous system.

    On one hand, gene mutations that en- code structural proteins, such as peripherin (peripherin/RDS) and rod outer segment protein 1 (ROM-1) lead to RP characterised by 'slower' degeneration in rodent models (hence the term retinal degeneration slow or rds mouse) and may account for about five per cent of auto- soma1 dominant RP in human^.^^^^^^ On the other hand, the mutant rhodopsin may have an alteration to protein structure that subsequently modifies the phototransduc- tion role of this protein. Based on the modelling of the a-wave in RP, Birch and associates6* proposed that the reduced maximum response, low gain and slower activation are due to an abnormality within the activation stages of the transduction cascade. They suggest a prolonged lifetime of activated rhodopsin (Rh*) and may re- flect a mechanism similar to light-induced retinal damage. Other findings suggesting abnormal photopigment kinetics include reduced rod bwave amplitude and delayed implicit time54 and delayed time course of dark adaptation after large bleach.63 In s u p port of this concept of abnormal activa- tion, in a model of congenital night blind- ness due to rhodopsin gene mutations, the mutant rhodopsin remained constitutively activated and thus would cause desensiti- sation of photoreceptors.M

    Photoreceptor degenerations such as a human form of autosomal recessive retini- tis pigmentosa, the rd mouse and Irish Setter dog model are caused by defects in photoreceptor cCMP-phosphodiesterase a ~ t i v i t y ~ - ~ ~ . ~ ~ (Figure 7). In the rd mouse model, a substantial increase in retinal

    cGMP appears to precede both photore- ceptor and inner retinal In vitro experiments have demonstrated a causal link between increases in cCMP levels and retinal degenerati~n.~~.~.~~~~".~' Incubation of isolated human retina, in the presence of PDE inhibitors or cCMP analogues, leads to degenerative changes that are proportional to the concentration of the drug used and the period of expo- sure. Over an eight-hour period, the presence of high concentrations of PDE inhibitor or cGMP analogue induced vesiculation of rod inner segments, and cone morphological changes. A combina- tion of PDE inhibitor and cGMP analogue in the incubation medium virtually de- stroyed every rod in the specimen over the same incubation p e r i ~ d . ~ ' The use of iodoacetate, a known inhibitor of glycolysis, which also inhibits sulfhydryl- containing enzymes such as PDE, results in elevated cGMP levels (within 35 min- utes) and visible photoreceptor degenera- tion within a day.72

    Despite such studies and the established elevation of cGMP in animal models of RP,67*68 the mechanism by which cCMP leads to photoreceptor degeneration re- mains unresolved. Elevated cyclic nucle- otide (cGMP) levels in the outer segment of photoreceptors would place photo- receptors effectively in a state of high activity or 'constant darkness'. The func- tional significance of 'constant darkness' is that photoreceptors must maintain their dark current at a considerable energy cost.79 It has been established by S te i r~be rg~~ that photoreceptors are in a state of physiologi- cal hypoxia in the dark: a hypoxia that does not lead to functional deficits, presumably due to the lowered energy demands dur- ing the light cycle.

    Anomalies in the delivery or recycling of the chromophore leads to a spectrum of RP conditions including a well charac- terised form of Leber's congenital amau- rosis involving RPE65, an RPE protein associated with ch romophore recy- ling.^.^^ The Royal College of Surgeons (RCS) rat, is an established model for retinal degeneration with the primary defect being a failure of phagocytosis of photoreceptor outer segments by the

    Clinical and Experimental Optometry 87.2 March 2004

    72

  • Retinitis pigmentosa Kalloniatis and Fletcher

    RPE.76R" In both humans and the RCS rat, a mutation in a tyrosine kinase receptor gene (Mertk"'."') has been identified as the genetic defect.

    In a chimeric model, where both nor- mal rhodopsin and mutated rhodopsin were present in the same retina, uniform retinal degeneration was evident, although the progression was slower than in the cor- responding transgenic mice.Rg The finding of Huang and associatesHS has important implications for the autosomal dominant form of RP. I t implies that the gene muta- tion per se does not lead to photoreceptor degeneration but that the mutant prod- uct triggers other not yet fully understood events (see discussion earlier), leading to degeneration of photoreceptors that do not have defective rhodopsin. These find- ings for the rhodopsin gene mutation dif- fer from previous work using chimeras, for the RCS rat rds mouse6' and the rd mouse.a In these three chimeric mod- els (RCS, rd, rds), the retinal degeneration occurs in a patchy manner, implying that the genetic defect is largely confined to the anomalous cells, unlike the rhodopsin gene mutation model of Huang and asso- c i a t e ~ . ~ ~ However, it was not clear whether the chimeric models (RCS, rd, rds) were followed to completion considering the different time courses in the diverse ani- mal models of RE4

    From a clinical viewpoint, variations in rod and cone sensitivity profiles suggest that the diversity in photoreceptor degenera- tion is evident in human forms of the con- dition. Massof and Finkelsteins4 assessed rod and cone sensitivity and reported the existence of three groups of patients: 1. subjects with only cone mediated re-

    sponses throughout their visual field 2. subjects with both rod and cone thresh-

    old elevation with the rod/cone thresh- old differences being similar to those expected in normal subjects

    3. subjects with cone mediated central vision, rod and cone mediated mid- peripheral vision and far-peripheral function mediated by rods.R4

    When genetic classification was used in three family groups displaying different gene mutations of the peripherin/RDS gene, all displayed similar psychophysical

    Anatomy Electroretinogram

    1 +a-wave

    Figure 7. Anatomical and physiological changes in degenerating (rd/rd and control C57 mouse retina). Nissl stained sections of the trf mouse retina at post-natal age 49 shows an absent photoreceptor layer with a few cone photoreceptors (asterisk). The inner nuclear layer (arrow) is also reduced in size compared to the control adult retina (C57 mouse). The electro- retinogram shows a normal appearance for the control (C57) animal with clear a- and bwaves, whereas the degenerating mouse (rd/rd) at the same age had effectively no detectable signal. Scale bar is 50 microns and stimulus intensity was 2.50 log cd.s.m-* (Gibson, Kalloniatis and Vigrys, unpublished data).

    and electrophysiological characteristics with rod and cone threshold elevations for the different gene mutations.44 Although there was variability in the degree of visual dysfunction within each group, the peripherin/RDS gene mutations appear to fall within category two of Massof and Finkel~tein.~~

    Despite differences in the underlying genetics causing the RP phenotype and other proposed environmental mecha- n i s m ~ , ~ ~ * ~ ' the established pathway of cell death is through apoptosis, a form of programmed cell death.7R.R5 Although the mechanism(s) leading to apoptosis in RP is not fully understood, an area of interest has been retinal metabolism.

    Retinal metabolism The major glial cell in the retina is the Miiller cell, which spans the entire length of the retina and is intricately related to

    retinal function and development.86.RR Normal retinal function is dependent on tight metabolic coupling between neurons and glia and the provision of oxygen and g l u c ~ s e . ~ ~ ~ ~ Glucose is the major substrate for retinal and brain metabolism but in mammalian retina, the major product of glucose utilisation is lactate, even under aerobic ~ o n d i t i o n s . ~ ' ~ ~ ~ The dense mito- chondria] packing within photoreceptor inner segments accounts for the high rate of metabolism of the retina,92.94 with both glycolytic and aerobic pathways used by photore~eptors.9~ Photoreceptor cells have a high energy demand because of: 1. the maintenance of an ionic gradient

    via the Na/KATPase pump (maintains the dark current)

    2. the large turnover of cyclic nucleotides in the outer segment including hydroly- sis of guanyl tri-phosphate to form cCMP

    3. multiple phosphorylations of photo-

    Clinical and Experimental Optometry 87.2 March 2004

    73

  • Retinitis pigmentosa Kulloniutis and Fletcher

    pigment (an essential step in deactiva- tion of opsins) .

    One role of glial cells appears to involve initial glucose catabolism with the carbon skeleton provided to neurons predomi- nantly in the form of lactate (with the last step pyruvate to lactate catalysed by the enzyme lactate dehydrogenase [LDHlYo). Consequently, the major enzymes of glycolysis and glycogen synthesis a r e localised in Miiller cells and studies have demonstrated Miiller cell metabolism of glucose in mammalian retina,w,gfi Lactate is an essential metabolic substrate of reti- nal metabolism involving the trafficking of the carbon skeleton of glucose between glial cells and neurons, the 'lactate traf- ficking hypothesis' of Tsacopoulos and Magi~tretti.9~ The hypothesis centres on the contention that glial cells in the retina and brain metabolise glucose and provide the carbon skeleton (lactate) to neurons for conversion to pyruvate, which fuels aero- bic metabolism. In the retina, Poitry- Yamate, Poitry and Tsacopoulossg have demonstrated tracking of lactate from neurons to glia. Disruption of this shuttle using a range of lactate transport inhibi- tors severely impairs retinal functiongs however, the fact that photoreceptors can use both glycolytic and aerobic metabolic pathwdysy5 suggests that lactate trafficking reflects only part of the metabolic path- ways available to retinal neurons.

    A stable source of metabolic substrate is required to maintain a high metabolic ac- tivity of the retina.w.y2.95,9H One example of high energy demand is the requirement to maintain the cyclic nucleotide gated channel in the open state in the dark. The high metabolic demand is due to the high activity of the sodium/potassium ATPase in the inner segment.73 In theory, the con- tinuously high metabolic demand in cases of elevated cGMP provides a parsimoni- ous explanation for a mechanism induc- ing photoreceptor degeneration. In con- trast, retinal degeneration caused by light damage, or that due to continuously acti- vated rhodopsin, would have a different underlying cause because the cyclic nucle- otide channel would not be makinga high contribution to energy demand.

    The interconversion of pyruvate/lactate

    is catalysed by LDH, which comes in vari- ous isoenzymes. Graymoreg9 first reported the metabolic alteration in LDH isoenzyme distribution in the RCS rat com- pared to control rat retina. He showed a dramatic alteration in the distribution of LDH isoenzymes with a marked reduction of one of the LDH isoenzymes (LDH5) at P7, an age when photoreceptors have not differentiated in the rat retina and well before anatomical alterations are evident in the RCS rat.7g*'w~Lo' The RCS rat also dis- played little change in total LDH activity before degenerationIo2 but the LDH isoenzyme distribution was altered early (at P7), before photoreceptor degenera- t i ~ n . ~ ~ However, Bonavita, Ponte and AmoreIo2 demonstrated a major loss of LDH activity after degeneration in the RCS rat retina indicating that LDH activity is a sensitive indicator of photoreceptor reti- nal function. In addition, the RCS rat shows altered glucose utilisation and trans- port 9s*105-105 and neurochemical studies have demonstrated alterations before, during and after the degenera t i~n . '~ . '~ ' Miiller cells display abnormal metabolism of glutamate and altered neurochemistry before degeneration that continue during the degenerative phase involving the amino acids glutamate, GABA, aspartate, glutamine and arginine. The latter change involves neurochemical changes during/ after degeneration, involving the amino acids glycine and taurine, as well as u p take of glutamate, GABA and g ly~ ine .~~ , " '~ More recently, Kalloniatis and associates Io6 showed that cation channels are constitu- tively open in many photoreceptors des- tined to degenerate, before apoptotic markers are evident. Open cation chan- nels would place photoreceptors in a state of constant depolarisation increasing the energy demand'* or may initiate apoptotic mechanisms.

    In the rd mouse model, metabolic anomalies have also been demonstrated before degeneration. NoellIo7 showed an increase in oxygen uptake, glucose utili- sation and lactic acid production (aerobic) detectable at P8 in the rd mouse retina, followed by a rapid decrease from P12. Metabolic substrate concentrations and high energy phosphate compounds do not

    show major differences between normal and rd mice,108,Lo9 especially at P15 to P20. The changes in oxygen consumption, glucose utilisation and lactic acid produc- tionlo7 identify an early metabolic dysfunc- tion in the rd mouse, well before photore- ceptor cells die through apoptosis at around P10 in the rd mouse."n The chief conclusion that can be drawn from such work is that in two disparate models of retinal degeneration, the RCS rat (caused by a mutation of the tyrosine kinase gene (MERTK8i*82) and the rd mouse (caused by a mutation of the PDE genem), both display metabolic anomalies before pho- toreceptor degeneration.

    Adult onset retinal degeneration mimicking RP There are forms of adult onset photore- ceptor degeneration mimicking RP asso- ciated with malignancies, including melanoma associated retinopathy (MAR) a n d cancer associated retinopathy (CAR).1"-114 In these conditions, individu- als suffering from small-cell carcinoma of the lung/breast or melanomas produce antibodies as part of the body's response to cancer. These antibodies are produced against epitopes on the cancer cells that also recognise a range of proteins in the retina.'"."2."5 Circulating serum antibod- ies must enter the retina, presumably through breaks in the blood retinal bar- rier and consequently destroy retinal neu- rons leading to RP-like signs and symptoms in these individuals. '"J"J~~ Although pro- posed, an immunological aetiology in typical RP has not been supported by experimental data.llfi Heckenlively and co- workerstJ7 found eight instances of serum anti-recoverin immunoglobulins in a sam- ple of 521 typical RP cases, implying that an immunological aetiology is not a likely mechanism in most cases of RP.

    TREATMENT OPTIONS: WHAT DO YOU TEU YOUR PATIENT WITH INHERITED RETINAL DYSTROPHY?

    In certain inherited forms of retinal dys- trophy, where biochemical pathway anomalies lead to the degeneration, diet control of foods containing ornithine or

    Clinical and Experimental Optometry 87.2 March 2004

    74

  • Retinitis pigmentosa Kalloniatis and Fletcher

    increasing residual enzyme activity with vitamin B6 has been beneficial in gyrate atrophy or a diet to decrease phytanic acid aids in Refsum disease.116."" The methods undertaken to modify the progression of retinal degeneration in RP include medi- cal intervention, for example, vitamin A, cardizem (diltiazem); activation of inher- ent protection systems, for example, growth factors may reduce apoptosis; re- placement of mutated gene; replacement of affected cells, for example, RPE/neural retinal transplants; and retinal prosthesis (Sharma and Ehinger116 and Lund and co- workers'lg provide comprehensive recent reviews).

    Modifying retinal degeneration in animal models of RP Photoreceptor degeneration in rodent models can be affected through a variety of measures, including modified oxygen levels,'20-'22 retinal pigment epithelial trans- plant,IZ2 creating chimera^,'^ growth fac- tors or apoptosis inhibitors123,124 or differ- ent light levels.'25 Frasson and associates reported an exciting development in the treatment of RP using a calcium channel blocker (D-cis-diltiazem) in the rd mouse retina (PDE gene mutation). They re- ported that an L-type calcium channel blocker slowed the progression of retinal degeneration in the rd mouse. L-type cal- cium channels are present on photo- r e c e p t o r ~ , ~ ~ ~ although the physiology of these channels in cones does not reflect responses elicited by classic L-type calcium channels.IZ8 The fact that D-cis-diltiazem modulated cone calcium channels is im- portant because it means that pharmaco- logical manipulation may be a viable treat- ment option (see Hart and colleagues12* for discussion) but the exact mechanism of action of calcium channel blockers is unclear.12'i~lm Frasson and associates pro- posed that elevated cGMP levels lead to photoreceptor depolarisation, with cal- cium channel blockers preventing further depolarisation via calcium entry into the cell. Several other studies have not found a beneficial effect of D-cis-diltiazem in the rd mouse,"" in the P23H rat model"' or the PDE6B mutant rcdl canine model.1s2 There are several explanations for these

    negative findings, which include differ- ences in experimental conditions, dosing regime and animal strain.12"

    Although at the experimental stage with many problems to overcome, gene therapy has led to some exciting results in animal models. The benefits of gene therapy have been hindered by problems including tar- geting the gene to the right cells, getting the new gene integrated into the genome and expressed, controlling the new gene and suppressing expression of deleterious genes in autosomal dominant RI?'33 In autosomal recessive RP, introduction of a healthy gene into dividing cells (in both the rd mouse and rds mouse models) led to modified photoreceptor degeneration, although the technique was restricted to in utero use.134,135 In a large animal model of retinal degeneration, a canine with a form of Leber's congenital amaurosis (RPE65 gene mutation), Acland and co-

    successfully applied gene therapy in congenital amaurosis. They re- ported ERG and other parameters consist- ent with improved visual function over 100 days post-treatment, providing the first strong evidence of gene therapy for a con- dition causing a devastating early loss of vision.'% This model also requires perina- tal intervention.

    Growth factors or apoptosis inhibitors, for example, bFGF, are involved in nerve cell differentiation and growth and have proved useful in animal models such as the rd mouse and rds mouse.'" Growth fac- tors may work by reducing apoptosis but other apoptosis retarding methods are in experimental phases. They require re- peated intra-vitreal injections and growth factors may have other undesirable ac- tions."' Even when rod death is delayed by survival factors, photoreceptors still experience deconstruction of their phe- notypes and fail to function.

    Attempts have been made to transplant both RPE and neural retina but there is little evidence that this can be made viable, as the transplants must precede rod death."9,13R Surgery is still a challenge with cell survival and the formation of mean- ingful contacts for the transplanted neu- ral retina in doubt. More importantly, for both retinal transplants, the degenerating

    retina shows modified neuronal and glial s t r u c t ~ r e , ' ~ ~ J ~ ~ implying that new, rogue circuits are being formed. In retinal re- modelling secondary to rod-cone retinal degeneration, there is neurite remodel- ling, followed by global remodelling (see animation at the following address: http:/ /prometheus.med.utah.edu/-marclab/ PER-full-remodeling.htm1). One excit- ing outcome of this research139 is that the neural retina is 'plastic' secondary to de- generation. If the mechanism(s) for this plasticity were understood, it would provide a means to control the formation of retinal circuitry secondary to retinal transplant.

    TREATMENT OPTIONS AVAILABLE TO HUMAN SUFFERERS OF RP

    Treatments that do not work include: 1. the British therapy, where patients

    undertook bee stings on the nape of the neck

    2. the Cuban therapy, which included electric shock therapy, ozonation of the blood, surgical implant of retrobulbar fat, vasodilators and multivitamins

    3. the Russian therapy, involving intra- muscular and periorbital injection of extracted RNA from y e a ~ t . ~ ~ . ' ~ ~

    Treatment options with untested prom- ise include the use of sunglasses and hyperbaric oxygen. Although sunglasses, particularly those with short-wavelength filters, are useful to minimise discomfort glare, there is conflicting evidence that they modify progression rate in human sufferers of RP.28,30 Hyperbaric oxygen lev- els, although modifying the ERG during the treatment period, may not alter the natural course of the disease.I4' In addi- tion, high oxygen levels have been shown to be detrimental in more advanced stages of the disease.28

    Two types of retinal implants have been proposed, epiretinal and subretinal. Direct retinal stimulation using epiretinal im- plants in RP patients'43 resulted in patients seeing spots of light that were usually coloured (yellow/blue/yellow-green) . Humayun and de Juan143 proposed that the resolution was up to about 1.8 degrees. This group'44 also reported that a patient could detect spots of light and possibly the

    Clinical and Experimental Optometry 87.2 March 2004

    75

  • Retinitis pigmentosa Kalloniatis and Fktcher

    direction of movement using a 16-elec- trode array. In a study that included both RP patients and a normal volunteer, a reso- lution limit of 2.25 degrees to 4.50 degrees was achieved with an electrode array in contact with the retina.'45z'46 Often the patients did not report percepts that matched the stimulation pattern and fre- quently described multiple percepts when one electrode was driven. The differing results obtained from the normal-sighted volunteer indicate that retinal degenera- tion alone does not explain the limited results in blind p a t i e n t ~ , I ~ ~ . I ~ ~ providing functional evidence supporting the reti- nal remodeling concerns of Marc and co- w o r k e r ~ . ' " ~ ~ ~ ~ Subretinal electrodes have been attempted in animal rn~dels,~~' . '*~ with results indicating that cortical activity can be induced by subcortical electrode stimu- lation. The long-term effect of such im- plants has not been assessed nor the effect of electrodes placed between the neural retina and the retinal pigment epithelium on retinal metabolic function. In addition, unlike the ear, there is considerable encod- ing of visual information before it is trans- mitted via ganglion cells and such sensory encoding would be required by any pros- thetic device. Technological development is still required.

    A large double-masked treatment trial of vitamin A and/or vitamin E by Berson and associates 14g demonstrated a small but significant slowing in the progression of RP secondary to vitamin A (a daily dose of 15,000 IU). The use of 400 IU of vitamin E was detrimental and the authors suggest that the use of vitamin A may prolong use- ful vision by up to seven years. The lack of data on functional improvement, such as visual field progression, is a weakness of the study and care should be used in ad- vising patients on vitamin A considering the known toxicity of this retinoid.l16 This mode of treatment has been shown to be beneficial in other forms of retinal dystro- phy and is currently recommended by the National Institute of Health (USA)'I6 (http://www.nei.nih.gov/news/ clinicalalerts/alert-rp.htm: http:/ / www.nei.nih.gov/news/pressreleases/ rppressrelease.htm). The National Insti- tutes of Health is also currently recruiting

    subjects with RP for a further trial on the benefits of vitamin A (ht tp: / / clinicaltrials.gov/ct/gui) .

    What do you advise your RP patient regarding current treatment options? The complexity underlying the different RP phenotypes may necessitate expert genetic counselling. Particularly for pa- tients with known autosomal dominant mutations of RP, it may be possible to eliminate the transfer of the mutated gene. The use of pre-implantation genetic diag- nosis (PGD) , where the zygome that does not contain the mutated gene is chosen for implantation, ensures that the identi- fied mutation is not transferred. This tech- nique has been applied extensively to in- herited conditions such as blood disorders and neurodegenerative diseases, provid- ing an additional 'treatment' option in cases where the mutation is k n ~ w n . ' ~ " J ~ '

    Vitamin A at 15,000 IU daily is the rec- ommended dose from the National Eye Institute, National Institutes of Health (USA). This therapeutic option should be undertaken after the patient consults medical and ophthalmic practitioners. The normal recommended daily intake is 2,500 IU, with three medium-sized carrots providing around 6,000 IU, just under one half the dose used in the study of Berson and associate^.'^^ Even though Sibulesky and co-workers152 found that up to 25,000 IU per day was not detrimental to visual function over five- to 12-year follow-up, consideration should be given to the following: vitamin A is contraindicated in incipient, active or recent alcoholism; liver disease, for example, history of hepatitis A or C; in children; during pregnancy (vitamin A is a teratogenic agent); during breast feeding or in malnutrition.

    Anti-oxidants such as vitamin E may be detrimental. 149

    Other pharmacological agents may be- come available but other potential treat- ment options are at the laboratory experi- mental phase.

    ACKNOWLEDGEMENTS We thank Riki Gibson for compiling Fig- ure 7 and contribution to the schematics

    in Figures 2 and 3. We are also grateful for helpful comments provided by Dr Robert E Marc a n d two anonymous reviewers.

    GRANTS AND FINANCIAL SUPPORT Some of the work outlined in this review was conducted with grants from Retina Australia and the National Health and Medical Research Council of Australia. Michael Kalloniatis holds a professorship funded by the Robert G Leitl estate.

    REFERENCES 1 .

    2.

    3.

    4.

    5.

    6.

    7.

    8.

    9.

    Haim M. Epidemiology o f retinitis pigmentosa in Denmark. Acta Ophthalmol Scand Suppl2002; 233: 1-34. Cibis GW, Abdel-Latif AA, Bron AJ, Chalam KV, Tripathi BJ, Tripathi RC, Wiggs J, Aaby AA. Basic and Clinical Science Course- Section 2: Fundamental and Principles of Ophthalmology. The foundation of the American Academy of Ophthalmology.

    Bessant DAR, Kaushal S, Bhattacharya SS. Genetics and biology of the inherited reti- nal dystrophies. In: Kaufman PL, Alm A, eds. Adler's Physiology of the Eye, 10th ed. St Louis: Mosby; 2003. ~358-381. Pacione LR, Szego MJ, Ikeda S, Nishina PM, McInnes RR. Progress toward understand- ing the genetic and biochemical mecha- nisms of inherited photoreceptor degenera- tions. Annu Rev Neumsci 2003; 26: 657-700. Dowling JE. The Retina: An Approachable Part of the Brain. Cambridge MA The Belknap Press; 1987. Wu SM, Maple BR. Amino acid neurotrans- mitters in the retina: a functional overview. Vision Res 1998; 38: 1371-1384. Werblin FS, Dowling JE. Organization of the retina of the mudpuppy, Necturus maculosus. 11. Intracellular recording. J

    Kaneko A. Receptive field organization of bipolar and amacrine cells in the goldfish retina. JPhysiol (Lond) 1973; 235, 133-153. Stell WK, Lightfoot DO, Wheeler TG, Leeper HF. Goldfish retina: functional po- larization of cone horizontal cell dendrites and synapses. Science 1975; 190: 989-990.

    2001. ~355-386.

    N ~ ~ h y s 1969; 32: 339-355.

    10. Naka.KI.'The cells horizontal cells talk to. Vision Res 1982; 22: 653660.

    1 1 . Sakuranaga M, Naka K. Signal transmission in the catfish retina. I. Transmission in the outer retina. JNeurOphysiol1985; 53: 373389.

    12. Ripps H, Witkovsky P. Neuroglia interaction in the brain and retina. Pmg Retinal Res

    13. La Cour M. The retinal pigment epithe- lium. In: Kaufman PL, Alm A, eds. Adler's

    1985; 4: 181-219.

    Clinical and Experimental Optometry 87.2 March 2004

    76

  • Retinitis pigmentosa Kalloniatis and Fletcher

    Physiology of the Eye, 10th ed. St Louis:

    14. Mata NL, Radu RA, Clemmons RC, Travis GH. Isomerization and oxidation of vitamin A in conedominant retinas: a novel path- way for visual-pigment regeneration in day- light. Neuron 2002; 36: 69-80.

    15. Bunt-Milam AH, Saari JC. Immunocyto- chemical localization of two retinoid- binding proteins in vertebrate retina. J Cell Bioll983; 97: 703-712.

    16. Bok D, Ong DE, Chytil F. Immunocyto- chemical localization of cellular retinol binding protein in the rat retina. Invest Ophthalmol Vis Sri 1984; 25: 877-883.

    17. Jones GJ, Crouch RK, Wiggert B, Cornwall MC, Chader GJ. Retinoid requirements for recovery of sensitivity after visual-pigment bleaching in isolated photoreceptors. Proc NatlAcad Sci USA 1989; 86: 96069610.

    18. Fesenko EE, Kolesnikov SS, Lyubarsky AL. Induction by cyclic GMP of cationic conduct- ance in plasma membrane of retinal rod outer segment. Nature 1985; 313: 310-313.

    19. Picaud S. Retinal biochemistry. In: Kaufman PL, Alm A, eds. Adler's Physiol- ogy of the Eye, 10th ed. St Louis: Mosby;

    20. Stryer L. Visual excitation and recovery. J Biol Chem 1991; 266: 10711-10714.

    21. McBee JK, Palczewski K, Baehr W, Pepperberg DR. Confronting complexity: the interlink of phototransduction and retinoid metabolism in the vertebrate retina. Prog Retinal Eye Res 2001; 20: 469- 529.

    22. Baehr W, Liebman P. Visual cascade. In: Nature-Encyclopedia of Life Sciences. London: Macmillan Reference Ltd; 2001. pl-8. Available from: URL: h t tp : / / www.els.net.

    23. Van Soest S, Westerveld A, de Jong PTVM, Bleeker-Wagemakers E, Bergen AAB. Retinitis pigmentosa: defined from a mo- lecular point of view. Surv Ophthalmoll999;

    24. Phelan JK, Bok D. A brief review of retini- tis pigmentosa and the identified retinitis pigmentosa genes. Mol Vis2000; 6: 116124. Available from: URL: www.molvis.org/ molvis/.

    25. Baehr W, Frederick JM. Inherited retinal diseases: vertebrate animal models. In: Nature-Encyclopedia of Life Sciences. London: Macmillan Reference Ltd; 2001. pl-9. Available from: URL: ht tp: / / www.els.net.

    26. Koenig R. Bardet-Biedl syndrome and Usher syndrome. Dm Ophthalmol2003; 37: 126140.

    27. Daiger SP, Rossiter BJF, Greenberg J , Christoffels A, Hide W. Data services and software for identifying genes and muta- tions causing retinal degeneration. Invest

    Mosby; 2003. p 348-357.

    2003. ~382-408.

    43: 321-334.

    Ophthalmol Vis Sci 1998; 39: S295. Available from: URL: (http://www.sph.uth.tmc.edu/ Retnet/).

    28. Stone J, Maslim J, Valter-Kocsi K, Mervin K, Bowers F, Chu Y, Barnett N, Provis J, Lewis G, Fisher SK, Bisti S, Gargini C, Cervetto L, Merin S, Pe'er J. Mechanisms of photoreceptor death and survival in mammalian retina. Prog Ret Eye Res 1999;

    29. Marmor MF, Aguirre G, Arden G, Berson E, Birch DG, Boughman JA, Car r R, Chatrianb GE, Del Monte M, Dowling J, Enoch J, Fishman GA, Fulton AB, Garcia CA, Gouras P, Heckenlively J, Hu D, Lewis RA, Niemeyer G, ParkerJA, Perlman I, Ripps H, Sandberg MA, Siege1 1, Weleber RG, Wolf ML, Wu L,Young RSL. Retinitis pigmentosa. Ophthalmology 1983; 90: 126131.

    30. Pagon RA. Retinitis pigmentosa. Surv Ophthalmoll988; 33: 137-177.

    31. Kanski JJ. Clinical Ophthalmology. Edin- burgh: Butterworth Heinemann: 2003.

    32. Birch J. Diagnosis of Defective Colour Vision. Oxford: Butterworth Heinemann; 1993. ~ 1 5 6 1 5 7 .

    33. Sandberg MA, Berson EL. Blue and green cone mechanisms in retinitis pigmentosa. Invest Ophthalmol Vis Sci 1977; 16: 149-157.

    34. Massof RW, Finkelstein D, Starr SJ, Kenyon KR, Fleischman JA, Maumenee IH. Bilat- eral symmetry of vision disorders in typi- cal retinitis pigmentosa. Br J Ophthalmol

    35. Massof RW, Dagnelie G, Benzschawel T, Palmer RW, Finkelstein D. First order dy- namics of visual field loss in retinitis pigmentosa. Clin Vision Sci 1990; 5: 1-26.

    36. Heckenlively JR, Boughman JA, Friedman LH. Pedigree Analysis. In: Heckenlively JR. Retinitis Pigmentosa. Philadelphia, PA: Lippincott; 1988. p6-24.

    37. Jay M. O n the heredity of retinitis pigmentosa. Br J Ophthalmol 1982; 66: 405- 416.

    38. Boughman JA, Fishman GA. A genetic analysis of retinit is pigmentosa. Br J Ophthalmoll983; 67: 449-454.

    39. Fishman GA. Genetic percentages. Arch Ophthalmoll978; 9 6 822-826.

    40. Boughman JA, Conneally PM, Nance WE. Population genetic studies of retinitis pigmentosa. Am ]Hum Genet 1980; 3 2 223-235.

    41. Badano JL, Katsanis N. Beyond Mendel: an evolving view of human genetic disease transmission. Nut Reu Genet 2002; 3: 779-789.

    42. Stone J, Malsim J, Fawsi A, Lancaster P, Heckenlively J. The role of perinatal stress in retinitis pigmentosa: evidence from sur- veys in Australia and the USA. Can J Ophthalmol2001; 36: 315-322.

    43. Kajiwara K, Berson EL, Dryja TP. Digenic retinitis pigmentosa due to mutations at the

    18: 689-735.

    ~491-496.

    1979; 63: 90-96.

    unlinked peripherin/RDS and ROMl loci. Science 1994; 264: 16041608.

    44. Jacobson SG, Cideciyan AV, Kemp CM, Sheffield VC, Stone EM. Photoreceptor function in heterozygotes with insertion or deletion mutations in the RDS gene. Invest Ophthalmol Vis Sci 1996; 37: 1662-1674.

    45. Wang MX, Sando RS Jr, Crandall AS, Donoso LA. Recent advances in the mo- lecular genetics of retinitis pigmentosa. Curr Opin Ophthalmoll995; 6: 1-7.

    46. Reese BE, Harvey AR, Tan SS. Radial and tangential dispersion patterns in the mouse retina are cell-class specific. Proc Nut1 Acad Sci USA 1995; 92: 24942498.

    47. Tan SS, Godinho L, Tam PP. Cell lineage analysis. X-inactivation mosaics. Methods Mol Biol2000; 135: 289-295.

    48. Bird AC. Management of retinitis pigmen- tosa-summary. Trans Ophthalmol Soc UK 1983; 103: 494-495.

    49. Fishman GA, Weinberg AB, McMahon TT. X-linked recessive retinitis pigmentosa: clinical characteristics of carriers. Arch Ophthalmoll986; 104: 1329-1335.

    50. Koenekoop RK, Loyer M, Hand CK, Al Mahdi H, Dembinska 0, Beneish R, Racine J, Rouleau GA. Novel RPGR mutations with distinct retinitis pigmentosa phenotypes in French-Canadian families. Am J Ophthalmol

    51. Tanino T, Ohba N, Mishima S. Studies on pigmentary retinal dystrophy. 111. A statisti- cal analysis of perimetric data. Klin Montsbl Augenheilkd 1977; 170: 808-813.

    52. Pearlman JK. Mathematical models of retinitis pigmentosa: study of the rate of progress in different genetic forms. Trans Amer Ophthalmol Soc 1979; 77: 643-656.

    53. Berson EL, Sandberg MA, Rosner B, Birch DG, Hanson AH. Natural course of retini- tis pigmentosa over a three-year interval. Am JOphthalmol1985; 99: 240-251.

    54. Berson EL, Rosner B, Sandberg MA, Weigel-DiFranco C, Dryja TP. Ocular find- ings in patients with autosomal dominant retinit is pigmentosa and rhodopsin, proline-347-leucine. Am J Ophthalmoll991; 111: 614623.

    55. Berson EL, Rosner B, Sandberg MA, Dryja TP. Ocular findings in patients with auto- soma1 dominant retinitis pigmentosa and a rhodopsin gene defect (Pro-23-His). Arch Ophthalmoll991; 109: 92-101.

    56. O h KT, Longmuir R, Oh DM, Stone EM, Kopp K, Brown J, Fishman GA, Sonkin P, Gehrs KM, Weleber RG. Comparison of the clinical expression of retinitis pigmentosa associated with rhodopsin mutations at codon 347 and codon 23. Am J Ophthalmol 2003; 136: 306313.

    57. Lewin AS, Drenser KA, Hauswirth WW, Nishikawa S, Yasumura D, Flannery JG, LaVail MM. Ribozyme rescue of photore-

    2003; 136: 678-687.

    Clinical and Experimental Optometry 87.2 March 2004

    77

  • Retinitis pigmentosa Kalloniatis and Fletcher

    ceptor cells in a transgenic rat model of autosomal dominant retinitis pigmentosa. Nut Med 1998; 4: 967-971.

    58. Olsson JE, GordonJW, Pawlyk BS, Roof D, Hayes A, Molday RS, Mukai S, Cowley GS, Berson EL, Dryja TP. Transgenic mice with a rhodopsin mutation (Pro23His): a mouse model of autosomal dominant retinitis pigmentosa. Neuron 1992; 9: 815-830.

    59. KijasJw, Cideciyan AV, Aleman TS, Pianta MJ, Pearce-Kelling SE, Miller BJ, Jacobson SG, Aguirre GD, Acland GM. Naturally oc- curring rhodopsin mutation in the dog causes retinal dysfunction and degenera- tion mimicking human dominant retinitis pigmentosa. Proc Natl Acad Sci USA 2002;

    60. Illing ME, Rajan RS, Bence NF, Kopito RR. A rhodopsin mutant linked to autosomal dominant retinitis pigmentosa is prone to aggregate and interacts with the ubiquitin proteasome system. J Biol Chem 2002; 277: 341 50-341 60.

    61. Sanyal S, Zeilmaker GH. Development and degeneration of retina in rds mutant mice: light and electron microscopic observa- tions in experimental chimaeras. Exp Eye

    62. Birch DG, Hood DC, Nusinowitz S, Pepperberg DR. Abnormal activation and inactivation mechanisms of rod transduc- tion in patients with autosomal dominant retinitis pigmentosa and the pro-23-his mutation. Invest Ophthalmol Vis Sci 1995; 36: 1603-1614.

    63. Kemp CM, Jacobson SG, Roman AJ, Sung CH, Nathans J. Abnormal rod dark adapta- tion in autosomal dominant retinitis pigmentosa with proline-23-histidine rho- dopsin mutation. Am J Ophthalmol 1992; 113: 165-174.

    64. Jin S, Cornwall MC, Oprian DD. Opsin ac- tivation as a cause of congenital night blind- ness. Nut Neurosci 2003; 6: 731-735.

    65. Lolley RN, Farber DB, Rayborn ME, Hollyfield JG. Cyclic GMP accumulation causes degeneration of photoreceptor cells: simulation of an inherited disease. Science 1977; 196: 664-666.

    66. Farber DB, Shuster TA. Cyclic nucleotides in retinal function and degeneration. In: Adler R, Farber D, eds. The Retina: a Model for Cell Biology Studes. Part I. Florida: Academic Press; 1986. p239-296.

    67. Chader GJ. Studies of animal models or retinal degeneration. In: Tso MDM. Reti- nal Diseases. Philadelphia: JB Lippincott; 1988. p80-89.

    68. Farber DB, Flannery JG, Bowes-Rickman C. The rd mouse story: seventy years of re- search on an animal model of inherited retinal degeneration. P..g Retinal Eye Res

    69. McLaughlin ME, Ehrhart TL, Berson EL,

    99: 6328-6333.

    R ~ s 1984; 39: 231-246.

    1994; 13: 31-63.

    Dryja TP. Mutation spectrum of the gene encoding the beta subunit of rod phos- phodiesterase among patients with auto- soma1 recessive retinitis pigmentosa. Proc Nut1 Acad Sci USA 1995; 92: 3249-3253.

    70. Ulshafer RJ, Fliesler SJ, Hollyfield JG. Dif- ferential sensitivity to protein synthesis in human retina to a phosphodiesterase in- hibitor and cyclic nucleotides. Cum Eye Res 1984; 3: 383-392.

    71. Ulshafer RJ, Garcia CA, Hollyfield JG. Sen- sitivity of photoreceptors to elevated levels of cGMP in the human retina. Invest Ophthalmol Vis Sci 1980; 19:1236-1241.

    72. Farber DB Souza DW, Chase DG. Cone visual cell degeneration in ground squir- rel retina: disruption of morphology and cyclic nucleotide metabolism by iodoace- tic acid. Invest Ophthalmol Vis Sci 1983; 24: 1236-1249.

    73. Ames A, Li Y-Y, Heher EC, Kimble R. En- ergy metabolism of rabbit retina as related to function: the high cost of Na transport. JNeurosci 1992; 12: 840-853.

    74. Steinberg RH. Monitoring communica- tions between photoreceptors and pigment epithelial cells: effects of mild systemic hypoxia. Invest Ophthalmol Vis Sci 1987; 28:

    75. Seeliger MW, Biesalski HK, Wissinger B, Gollnick H, Gielen S, Frank J, Beck S, Zrenner E. Phenotype in retinol deficiency due to a hereditary defect in retinol bind- ing protein synthesis. Invest Ophthalmol Vis Sci 1999; 40: 3-1 1.

    76. Bok D, Hall MO. The role of the pigment epithelium in the etiology of inherited reti- nal dystrophy in the rat.JCel1 B i d 1971; 49: 664-682.

    77. Mullen RJ, LaVail MM. lnherited retinal dystrophy: primary defect in pigment epi- thelium determined with experimental rat chimeras. Science 1976; 192: 799-801.

    78. Tso MO, Zhang C, Abler AS, Chang CJ, Wong F, Chang GQ, Lam TT. Apoptosis leads to photoreceptor degeneration in in- herited retinal dystrophy of RCS rats. In- vest Ophthalmol Vis Sci 1994; 35: 2693-2699.

    79. Fletcher EL, Kalloniatis M. Neurochemical architecture of the normal and degenerating rat retina. JComp Neumll996; 376 343-360.

    80. Strauss 0, Stumpff F, Mergler S, Weinrich M, Wiederholt M. The Royal College of Sur- geons rat: An animal model for inherited retinal degeneration with a still unknown genetic defect. Acta Anatomica (Basel) 1999;

    81. DCruz PM, Yasumura D, Weir J, Matthes MT, Abderrahim H, LaVail MM, Vollrath D. Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Hum Mol Gen 2000; 9: 645-651.

    82. Gal A, LiY, Thompson DA, Weir J, Orth U, Jacobson SG, Apfelstedt-Sylla E, Vollrath D.

    1888-1 904.

    162: 101-111.

    Mutations in Mertk, the human orthologue of the RCS rat retinal dystrophy gene, cause retinitis pigmentosa. Nature Gen 2000; 26:

    83. Huang PC, Gaitan AE, Hao Y, Petters RM, Wong F. Cellular interactions implicated in the mechanism of photoreceptor degenera- tion in transgenic mice expressing a mu- tant rhodopsin gene. Proc Natl Acad Sci USA 1993; 90: 8484-8488.

    84. Massof RW, Finkelstein D. Rod sensitivity relative to cone sensitivity in retinitis pigmentosa. Invest Ophthalmol Vis Sci 1979; 18: 263-272.

    85. Lolley RN. The rd gene defect triggers pro- grammed rod cell death. The Proctor Lec- ture. Invest Ophthalmol Vis Sci 1994; 35:

    86. Rich KA, Figueroa SL, Zhan Y, Blanks JC. Effects of Muller cell disruption on mouse photoreceptor cell development. Exp Eye

    87. Pow DV, Crook DK. Direct immunocyto- chemical evidence for the transfer of glutamine from glial cells to neurons: use of specific antibodies directed against the d-stereoisomers of glutamate and glutamine. Neumsci 1995; 70: 295-302.

    88. Kalloniatis M, Tomisich G. Amino acid neu- rochemistry of the vertebrate retina. h o g Retinal Eye Res 1999; 18: 81 1-866.

    89. Poitry-Yamate CL, Poitry S, Tsacopoulos M. Lactate released by Miller glial cells is me- tabolized by photoreceptors from mamrna- lian retina.JNeurosci 1995; 15: 5179-5191.

    90. Tsacopoulos M, Poitry-Yamate CL, MacLeish PR, Poitry S. Trafficking of mol- ecules and metabolic signals in the retina. Prog Retin Eye Res 1998; 17: 429-442

    91. Winkler BS. Glycolytic and oxidative me- tabolism in relation to retinal function. J Gen Physioll981; 77: 667-692.

    92. Winkler BS, Arnold MJ, Brassell MA, Puro DG. Energy metabolism in human retinal Miiller cells. Invest Ophthalmol Vis Sci 2000;

    93. Brotherton J. Studies on the metabolism of the rat retina with special reference to retinitis pigmentosa. I. Anaerobic glycoly- sis. Exp Eye Res 1962; 1: 234-245.

    94. AlderVA, Cringle SJ, Constable IJ. The reti- nal oxygen profile of cats. Invest Ophthalmol Vis Sci 1983; 24: 30-36.

    95. Bui BV, Kalloniatis M, Vingrys AJ. The con- tribution of glycolytic and oxidative path- ways to retinal photoreceptor function. Invest Ophthalmol Vis Sci 2003; 44: 2708-2715.

    96. Kuwabara T, Cogan DG. Retinal glycogen. Arch Ophthalmoll961; 6 6 96-104.

    97. Tsacopoulos M, Magistretti PJ. Metabolic coupling between glia and neurons. J Neurosci 1996; 16: 877-885.

    98. Bui BV, Kalloniatis M, Vingrys AJ. Retinal function loss after monocarboxylate trans-

    270-271.

    41 82-4 19 1.

    R ~ s 1995; 61: 235-248.

    41: 3183-3190.

    Clinical and Experimental Optometry 87.2 March 2004

    78

  • Retinitis pigmentosa Kulloniutis and Fletcher

    port inhibition. Invest Ophthalmol Vis Sci 2004: 45: 584593.

    99. Graymore C. Possible significance of the isoenzymes of lactic dehydrogenase in the retina of the rat. Nature 1964; 201: 615-616.

    100. Fletcher EL, Kalloniatis M. Localization of amino acid neurotransmitters during postnatal development of the rat retina. J Comp Neurol1997; 380: 449-471.

    101. Fletcher EL, Kalloniatis M . Neurochemi- cal development of the degenerating rat retina.J Comp Neuroll997; 388: 1-22.

    102. Bonavita V, Ponte F, Amore G. Neuro- chemical studies on the inherited retinal degeneration of the rat. Vision Res 1963; 3:

    103. Reading HW, Sorsby A. The metabolism of the dystrophic retina. I: Comparative studies on the glucose metabolism of the developing rat retina, normal and dys- trophic. Vision Hes 1962; 2: 315-325.

    104. Stramm LE, Pautler EL. Glucose uptake by normal and dystrophic rat retinas and cili- ary bodies. Exp Eyp Res 1980; 30: 709-718.

    105. Di Mattio J, Zadunaisky JA. Reduced ocu- lar glucose transport and increased non- electrolyte permeability in rats with retinal degeneration (RCS). Exp Eye Hes 1983; 37:

    106. Kalloniatis M, Tomisich G, Wellard JW, Foster LE Mapping photoreceptor and post-receptoral labelling patterns using a channel permeable probe (agmatine) dur- ing development in the normal and RCS rat retina. Visual Neurosci 2002; 19: 61-70.

    107. Noell WK. Aspects of experimental and hereditary retinal degnerat ion. In: Graymore C. Biochemistry of the Retina. London: Academic Press; 1965. p51-72.

    108. Lolley RN. Changes in glucose and energy metabolism in vivo in developing retinae from visually-competent (DBA/lJ) and mutant (C3H/HeJ) mice.JNmrorhem 1972;

    109. Lolley RN, Racz E. Changes in levels of ATPase activity in developing retinae of normal (DBA) and mutant (C3H) mice. Vision Res 1972; 12: 567-571.

    110. Portera-Cailliau C , Sung CH, Nathans J, Adler R. Apoptotic photoreceptor cell death in mouse models of retinit is pigmentosa. h c Natl Acad Sci USA 1994; 91: 974978.

    111. Goldstein SM, Syed NA, Milam AH, Maguire AM, Lawton TJ, Nichols CW. Cance r-assoc ia ted re tin o pa t h y. Arrh Ophthalmoll999; 117: 1641-1645.

    112. Ohguro H, Ogawa K, Maeda T, Maruyama I, Maeda A, TakanoY, Nakazawa M. Retinal dysfunction in cancer-associated retinopa- thy is improved by Ca(2+) antagonist ad- ministration and dark adaptation. Invest Ophthalmol Vis Sci 2001; 42: 2589-2595.

    113. Maeda A, Maeda T, Ohguro H, Palczewski

    2 7 1-280.

    217-223.

    19: 175-185.

    K, Sato N. Vaccination with recoverin, a cancer-associated retinopathy antigen, in- duces autoimmune retinal dysfunction and tumor cell regression in mice. EurJImmunol

    114. KeltnerJL, Thirkill CE,Kp PT. Clinical and immunologic characteristics of melanoma- associated retinopathy syndrome: eleven new cases and a review of 51 previously published cases. JNeuroophthalmol2001; 21: 173-187.

    15. Lei B. Bush RA, Milam AH, Sieving PA. Human melanoma-associated retinopathy (MAR) antibodies alter the retinal ON- response of the monkey ERG in vivo. Invest Ophthalmol Vis Sci 2000; 41: 262-266.

    16. Sharma RK, Ehinger B. Management of hereditary retinal degeneration: present sta- tus and future directions. Sun, Ophthalml 1999; 43: 427-444.

    117. Heckenlively JR, Fawzi AA, Oversier J, Jordan, BL, Aptsiauri N. Autoimmune retin- opathy: patients with antirecoverin immu- noreactivity and panretinal degeneration. ATch Ophthalmol2000; 1 18: 1525-1 533.

    118. Wang T, Steel G, Milam AH, Valle D. Correction of ornithine accumulation pre- vents retinal degeneration in a mouse model of gyrate atrophy of the choroid and retina. R o c NatlAcad Sci USA 2000; 97: 1224 1229

    119. Lund RD, Kwan AS, Keegan DJ, Sauve Y, Coffey PJ, Lawrence JM. Cell transplanta- tion as a treatment for retinal disease. Prog Retin Eye Res 2001; 20: 415449.

    120. Maslim J, Valter K, Egensperger R, Hol- lander H, Stone J. Tissue oxygen during a critical developmental period controls the death and survival of photoreceptors. In- vest Ophthalmol Vis Sci 1997; 38: 1667-1677.

    121. Valter K, Maslim J, Bowers F, Stone J. Pho- toreceptor dystrophy in the RCS rat: roles of oxygen, debris and bFGF. Invest Ophthalmol Vis Sci 1998; 39: 2427-2442.

    122. Li LX, Turner JE. Inherited retinal dys- trophy in the RCS rat: prevention of pho- toreceptor degenerat ion by pigment epithelial cell transplantation. Exp Eye Res

    123. Faktorovich EG, Steinberg RH,Yasumura D, Matthes MT, LaVail MM. Photoreceptor degeneration in inherited retinal dystrophy delayed by basic fibroblast growth factor. Nature 1990; 347: 83-86.

    124. Katai N, Kikuchi T, Shibuki H, Kuroiwa S, Arai J , Kurokawa T, Yoshimura N. Caspaselike proteases activated in apoptotic photoreceptors of Royal College of Sur- geons rats. Invest Ophthalmol Vis Sci 1999;

    125. Organisciak DT, Darrow RM, Barsalou L, Kutty RK, Wiggert B. Susceptibility to reti- nal light damage in transgenic rats with rhodopsin mutations. Invest Ophthalmol Vis Sci 2003; 44: 486-492.

    2002; 32: 2300-2307.

    1988; 47: 911-917.

    40: 1802-1807.

    126. Frasson M, Sahel JA, Fabre M, Simonutti M, Dreyfus H , Picaud S. Retinitis pigmentosa: rod photoreceptor rescue by a calcium-channel blocker in the rd mouse. Nat Med 1999; 5:1183-1187.

    127. Taylor WR, Morgans C. Localization and properties of voltage-gated calcium chan- nels in cone photoreceptors of Tupaia belangeri. Visunl Neurosci 1998; 15: 541-552.

    128. HartJ, Wilkinson MF, Kelly MEM, Barnes S. Inhibitory action of diltiazem on voltage- gated calcium channels in cone photo- receptors. Exp Eye Res 2003; 7 6 597-604.

    129. Edward DP, Edward DP, Lam TT, Shahinfar S, Li J, Tso MO. Ameliaration of light-induced retinal degeneration by a cal- cium overload blocker. Arch Ophthalmol 1991; 109: 554562.

    130. Pawlyk BS, Li T, Scimeca MS, Sandberg MA, Berson EL. Absence of photoreceptor rescue with Dcisdiltiazem in the rd mouse. Invest Ophthalml VisSci2002; 43: 1912-1915.

    131. Bush RA, Kononen L, Machida S, Sieving PA. The effect of calcium channel blocker diltiazem on photoreceptor degeneration in the rhodopsin Pro23His rat. Invest Ophthalmol Vis Sci 2000; 41: 2697-2701.

    132. Pearce-Kelling SE, Aleman TS, Nickle A, Laties AM, Aguirre GD, Jacobson SG, Acland GM. Calcium channel blocker D-cisdiltiazem does not slow retinal degen- eration in the PDEGB mutant rcdl canine model of retinitis pigmentosa Mol Vision 2001; 7: 4247.

    33. Reichel MB, Ali RR, Hunt DM, Bhattacharya SS. Gene therapy for retinal degeneration. Ophthalmic Res 1997; 29: 261- 268.

    34. Lem J, Flannery JG, Li T, Applebury ML, Farber DB, Simon MI. Retinal degenera- tion is rescued in transgenic rd mice by ex- pression of the cGMP phosphodiesterase beta subunit. Roc Natl Arad Sri USA 1992; 89: 4422-4426.

    135. Travis GH, Groshan KR, Lloyd M, Bok D. Complete rescue of photoreceptor dyspla- sia and degeneration in transgenic retinal degeneration slow (rds) mice. Neuron 1992; 9: 113-119.

    136. Acland GM, Aguirre GD, Ray J, Zhang Q Aleman TS, Cideciyan AV, Pearce-Kelling SE, Ananad V, Zeng Y, Maguire AM, Jacob- son SG, Hauswirth WW, Bennett J. Gene therapy restores vision in a canine model of childhood blindness. Nat Gaet2001; 28:

    137. LaVail MM, Yasumura D, Matthes MT, Lau-Villacorta C, Unoki K, Sung CH, Steinberg RH. Protection of mouse photoreceptors by survival factors in reti- nal degenerations. Invest Ophthalmol Vis Sci 1998; 39: 592-602.

    138. Marc RE, Jones BW, Watt CB, Strettoi E. Neural remodeling in retinal degeneration.

    92-95.

    Clinical and Experimental Optometry H7.2 March 2004

    79

  • Retinitis pigmentosa Kalloniatis and Fletcher

    Prog Retin Eye Res 2003; 22: 607-655. 139. Jones BW, Watt CB, Frederick JM, Baehr

    W, Chen CK, Levine EM, Milam AH, Lavail MM, Marc RE. Retinal remodeling trig- gered by photoreceptor degenerations. J Comp Neurol2003; 464 1-16.

    140. Weleber RG. The Cuban experience: false hope for a cure for retinitis pigmentosa. Arch Ophthalmoll996; 114 606607.

    141. Berson EL, Remulla JF, Rosner B, Sandberg MA, Weigel-DiFranco C. Evalua- tion of patients with retinitis pigmentosa receiving electric stimulation, ozonated blood and ocular surgery in Cuba. Arch Ophthalmoll996; 114: 560-563.

    142. Vingolo EM, Pelaia P, Forte R, Rocco M, Giusti C, Rispoli E. Does hyperbaric oxy- gen (HBO) delivery rescue retinal photo- receptors i n retinitis pigmentosa? Doc Ophthalmoll998; 97: 33-39.

    143. Humayun MS, de Juan E Jr. Artificial vision. Eye 1998; 12: 605607.

    144. Humayun MS, Weiland JD, Fujii GY, Greenberg R, Williamson R, Little J, Mech B, Cimmarusti V, Van Boemel G, Dagnelie G, de Juan E. Visual perception in a blind subject with a chronic microelectronic reti- nal prosthesis. Vision Res2003; 43: 2573-2581.

    145. Rizzo JF 3rd, WyattJ, Loewenstein J, Kelly S, Shire D. Methods and perceptual thresh- olds for short-term electrical stimulation of human retina with microelectrode arrays. Invest Ophthalmol Vis Sci 2003; 44: 5355-5361.

    146. Rizzo JF 3rd, WyattJ, Loewenstein J, Kelly S, Shire D. Perceptual efficacy of electrical st imulation of human ret ina with a microelectrode array during short-term sur- gical trials. Invest Ophthalmol Vzs Sci 2003; 44: 5362-5369.

    147. Chow AY, Chow W. Subretinal electrical stimulation of the rabbit retina. Neurosn'Lett 1997; 225: 13-16.

    148. Zrenner E, Miliczek KD, Gabel VP, Graf HG, Guenther E, Haemmerle H , Hoeminger B, Kohler K, Nisch W, Schubert M, Stett A, Weiss S. The development of subretinal microphotodiodes for replace- ment of degenerated photoreceptors. Oph- thalmic Res 1997; 29: 269-280.

    149. Berson EL, Rosner B, Sandberg MA, Hayes KC, Nicholson BW, Weigel-DiFranco C, Willett W. A randomized trial of vitamin A and vitamin E supplementation for retini- tis pigmentosa. Arch Ophthalmoll993; 111:

    150. Oyesiku JO, Turner CF. Reproductive choices for couples with haemophilia. H a - mophilia 2002; 8: 348-352.

    151. V Chan J, TKChan J. Prenatal diagnosis of common single gene disorders by DNA tech- nology. HongKongMedJ1997; 3 173-178

    152. Sibulesky L, Hayes KC, Pronczuk A, Weigel-DiFranco C, Rosner B, Berson EL. Safety of < 7500 RE (< 25000 IU) vitamin A

    761-772.

    daily in adultswith retinitis pigmentosa. Am J Clin N U ~ T 1999; 69: 656-663.

    Author's address: Professor Michael Kalloniatis Robert G Leitl Professor of Optometry Department of Optometry and Vision Science The University of Auckland Private Bag 92019 Auckland NEW ZEALAND

    Clinical and Experimental Optometry 87.2 March 2004

    80