Sie sind auf Seite 1von 16

OPTOMETRY

I INVITED REVIEW

Retinitis pigmentosa:
understanding the clinical presentation,
mechanisms and treatment options
Clin Ex$ Optom 2004; 87: 2: 65-80
Michael Kalloniatis' PhD
Erica L Fletcher' PhD
* Department of Optometry and Vision
Science, University of Auckland,
New Zealand
Department ofAnatomy and Cell Biology,
The University of Melbourne, Australia
Submitted: 19January 2004
Revised: 9 February 2004
Accepted for publication: 9 February 2004

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 mutations 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 outlined, 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 management 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 heterogenous group of inherited ocular diseases
representing the most recurrent retinal
dystrophies, with a worldwide prevalence of
1:3000 to 1:5000.' Inherited forms of retinal degeneration are largely focused on
gene mutations within photoreceptors or
W E cells leading to devastatingloss ofvisual
function, often progressing to functional
blindness. The chief gene mutations leading 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 manage patients with RP, it is important to review

the basic structure of the retina and the key


in the retina, with the major retinal glial
pathways involved in the visual response
cell being the Miiller cell" (Figure 1).
affected in the disease process.
Mtller cells are intimately involved in a
T h e visual process begins with light
range of activities essential for normal retiabsorption in the photoreceptor outer segnal functions that include neurotransmitment with subsequent encoding occurring
ter a n d metabolite metabolism, a n d
through interactions between the five
chromophore recycling. The retinal pigother major classes of retinal n e ~ l r o n s . ~ . ~ment epithelial (RPE) cells are considered
Photoreceptor cells (rods and cones), bipart of the retina and are involved in a
polar cells and ganglion cells form the
multitude of functions essential for neu'through' pathway, while horizontal cells,
ral retinal function. These include the
amacrine cells and interplexiform cells
transport and storage of vitamin A derivaform the 'lateral' or 'modulatory' pathways
tives (retinoids), recycling of the visual pig(Figure 1). The complex interplay bem e n t , o u t e r photoreceptor segment
tween the different retinal neurons leads
phagocytosis and anti-oxidant functions.'J3
to the encoding of spatial, temporal and
chromatic information that is subseActivation and deactivation of the
visual pigment
quently relayed by ganglion cells to the
central visual pathway~.~.'-'I
In addition to
The visual pigment within the photorecep
neural cells, several glial cells are localised
tor outer segment is composed of an inClinical 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 M U e r cells have long been known to
contain retinoid binding protein^.'^.'^ With
t h e 11-cis retinol available to c o n e
photoreceptors, pigment regeneration
can occur after cones convert 1 1 4 s retinol
to 1l-cis retinal: a reaction known to occur
in isolated cone photoreceptors but not
in rod photo receptor^.'^

Phototransduction cascade

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 (1l-cis to alltrans conformational change), that initiates 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 subsequent light can be detected. The rhodopsin 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 (alltrans retinal) detaches from the opsin and
begins its regeneration via the RPE or
Miiller cells'4 (Figures 3 and 4). The findClinical and Experimental Optometry 87.2 March 2004

66

Activation of the visual pigment (Rh* in


rod photoreceptors or the long- middleor short-wavelength photopigment for
cone photoreceptors), allows for the binding of a series of membrane-associated
proteins involved in the visual transduction cascade. The first protein to bind,
transducin, is in the Gfamily of proteins,
which are often associated with processes
linked with signal amplification. The binding and activation of transducin subsequently leads to activation of phosphodiesterase (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 hyperpolarisation to a light
The production 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 enzyme 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

Retinitis pigmentosa Kalloniatis and Fletcher

night blindness; g e n e mutations in


guanylate cyclase may cause cone dystrophy o r a form of Lebers congenital amaurosis; whereas a well-defined gene mutation 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 mutations may cause syndromes associated with
RP that include Laurence-Moon/BardetBiedl and Ushers syndrome as well as
other RP-like conditions involving metabolic defects such as Refsums disease.
Recent reviews on the molecular biology
of RP a n d other inherited retinal
dystrophies are provided by several articless.2328 and Daiger and co-worker? maintain an up-to-date website at the address

http://www.sph.uth.tmc.edu/Retnet/,
which outlines genes causing inherited retinal 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 electophysiological methods o r psychophysically) and progression of photoreceptor
dysfunction are classified as RP. Clinical presentation may he varied hut include
symptoms of night blindness (nyctalopia)
with elevated dark adaptation thresholds
(predominantly rod hut also cone thresholds), abnormal electroretinographic
a- and h-wave, difficulty with mobility secondary to visual field constriction, acquired blue-yellow (tritan) colour vision
defect, abnormal retinal pigmentation,

Figure 2. Simplifiedphotoreceptor outer segment under dark and light adaptation. The nonspecific 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, predisposition to myopia, posterior subcapsular cataract and vitreous changes.30-3z
Visual disturbances occur when visual acuity is relatively unaffected and careful
choice of colour vision tests may reveal the
acquired tritan colour vision defect early
M o st of the
in the disease pro~ess.~~~
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 d e p e n d e n t on t h e relative
involvement of rod a n d cone photoreceptors. 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 a n d multiple^.'^^^^^^
Heckenlively, Boughman and Friedmansfi
Clinical and Experimental Optometry 87.2 March 2004

67

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 approximately 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 published from around the world, with autosoma1 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 parentto-child transmission. Autosomal recessive
classification is based on established inheritance within different family groups
consistent with Mendelian characteristics.

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 (Mataand 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 1l-cis-retinol and 114s-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 family inheritance based on Mendelian characteristics. 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 simplex cases compared with predictions by
Mendelian genetic^.^^^^^^'^^* FishmanSg

reported 60 per cent with a known Mendelian 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,40a key consideration in this classification is the exclusion of consanguinity. Consanguinity
would significantly increase the odds of
carrying the same mutated gene.'
A new evolving concept in the inheritClinical and Experimental Optometry 87.2 March 2004

68

ance of many conditions, including some


forms of RP or syndromes associated with
RP (Usher type 1; RDS/ROMl; BardetBiedl), is referred to as ~ l i g o g e n i cPoly.~~
genic traits are disorders that occur
through poorly understood interactions
between many genes and the environment. Such an environmental interaction
could explain the higher correlation between perinatal stress and some simplex
forms of RF' reported by Stone and coll e a g u e ~ . ~T*h, e~ ~authors suggest the
existence of a non-hereditary explanation
for photoreceptor death (such as perinatal stress), for some simplex RP. This
suggestion awaits further experimental evidence 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 phen ~ 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 phenotype despite the fact that the mutations
were in two distinct genes.49In addition,
it may be possible to modify the expression 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,',35or other non-genetic mechanisms are in p l a ~ e . ~For
~ *example,
~'
we
now know that human subjects with specific gene mutations such as that affecting
a photoreceptor structural protein
peripherin (peripherin/RDS gene mutation), 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 associates45outlined
the following clinical implications of gene
mutations. Different gene mutations of the
same gene result in different clinical pres-

Retinitis pigmentosa Kalloniatis and Fletcher

entations, (for example, different mutations in the rhodopsin gene give rise to
different clinical presentations: allelic heterogeneity); 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 Usher's syndrome: locus heterogeneity); the
expression is different for the same genetic defect (expressivity) ; the defective
gene is not expressing leading to variations 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 Xlinked RP, female carriers should display
signs and symptoms of RP; a common finding, although there is considerable varia b i l i t ~ . ~Female
" . ~ ~ carriers may display a
tapetal reflex, irregular pigmentation in
the posterior pole, WE atrophy and pigment stippling and may express a phenotype typical of more advanced RP.4R-50

Visual function in Rp
Traditionally it has been thought that the
progression of visual dysfunction is slowest in those with autosomal dominant RP,
followed by recessive (including simplex/
multiplex) RP and the fastest are those
that are X-linked.35The advent of molecular biological techniques to classify RP patients has provided useful insights to complement 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 different genetic subgroups.52In a major
study of 172 typical RP patients followed
from 2.5 to 10 years, Massof and coll 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 onset). These parameters reflect photopic
visual fields that represent cone loss secondary to rod loss or concurrent rod and
cone loss. It follows that such measurements overestimate the age of onset of scotopic visual field loss by an unknown
amount because we d o 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
Clinical and Experimental Optometry 87.2 March 2004

69

in a time constant = 8.4 f 4.9 years ( E D


range of 3.5 to 13.3years) ..y5 There was no
significant difference in the time constant
for visual field progression across the different 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 progression of patients with RP.The second
parameter investigated by Massof and colleaguess5 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 differences in type 1 versus type 2 RF', implying
that the age of onset demonstrates some
variability. T h e 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

Retinitis pigmentosa Kalloniatis and Fletcher

patients likely to be encountered in optometric practice.


Berson and associates53followed 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 finding was that a minimum of two years was
required to follow visual fields. Visual acuity and dark adaptation ( 1 1 degrees central) remained unchanged over three years
and 77 per cent showed significant reduction in the electroretinogram (16 per cent
amplitude loss per year). There was no significant difference in loss of electroretinogram 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 biological genotyping, several studies have
focused on visual function measurements
in patients with specific gene mutations
s~~
causing RP. Berson and c o - ~ o r k e rstudied 17 cases with an autosomal dominant
form of RP (P23H:proline to histidine substitution 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 mutations. 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 patients 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 cow o r k e r ~reported
~~
considerable he terogeneity 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 compared with 140 unrelated subjects with
autosomal dominant rhodopsin gene mutations. Mobility was impaired between 12
and 27 years of age.55Although 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 subsequent study encompassing a spectrum
of rhodopsin gene mutations at these two
terminals.56The G and N-terminus are illustrated in Figure 3. The same amino acid
switch (proline to alanine) at two different locations (P23A versus P347A), resulted in a greater loss in all visual functions measured for the Gterminus located
gene mutation (P347A).56Gene mutations
at loci encoding the C-terminal may impact 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 below for further comments). The use of
genetic phenotyping invariably will lead to
a better understanding of the RP phenotype including visual function progression
characteristics.
The following two case studies illustrate
the usefulness of appreciating the inheritance 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 presentation secondary to functional mobility difficulties was within the critical age prediction from the results of Massof and
colleague^.^^ The fact that a male cousin
was affected with a similar condition indicates that autosomal recessive may be a
mode of inheritance, although the possibility of oligogenic inheritance should also
be con~idered.~'
O n further questioning,
it was ascertained that the parents of both
affected members were related and therefore 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 'tripping over her children's toys'. Nyctalopia
was not a problem at the time of diagnosis. A first cousin (male) was also affected
but there was no other family history reported (her parents and the parents of her
affected cousin had normal vision). Her
siblings were examined and were unaffected (Figure 5a). Visual acuity with spectacle correction was RE 6/18, LE 6/21 but
corneal topography indicated a keratoconus-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
Clinical and Experimental Optometry 87.2 March 2004

70

CASE 2
A 54year-old male with typical RP had
been diagnosed at age 44 years. Nyctalopia was first noticed about 10 years before
diagnosis. There was no history of consanguinity 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 going 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 reassessment of visual fields using Goldmann
perimetry. Visual acuities were 6/6 R and L
a n d dark adaptation thresholds were

Retinitis pigmentosa Kalloniatis and Fletcher

Patient#l

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 adaptation 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 inheritance include X-linked or autosomal dominant. 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 quantitative 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 colleagueP 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 progression 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 obtain 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 predictor 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
example,
p i g r n e n t o ~ a ~(for
~.~
~ - ~ ~ P23H rat).
Why a rhodopsin gene mutation causes
retinal degeneration is unclear. Rhodopsin
is an integral membrane protein that comprises 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 associatesMshowed 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 susceptible to degradation by the ubiquitindependent 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 encode 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 autosoma1 dominant RP in human^.^^^^^^ On the
other hand, the mutant rhodopsin may
have an alteration to protein structure that
subsequently modifies the phototransduction 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 reflect a mechanism similar to light-induced
retinal damage. Other findings suggesting
abnormal photopigment kinetics include
reduced rod bwave amplitude and delayed
implicit time54and delayed time course of
dark adaptation after large bleach.63In s u p
port of this concept of abnormal activation, in a model of congenital night blindness due to rhodopsin gene mutations, the
mutant rhodopsin remained constitutively
activated and thus would cause desensitisation of photoreceptors.M
Photoreceptor degenerations such as a
human form of autosomal recessive retinitis 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
Clinical and Experimental Optometry 87.2 March 2004

72

cGMP appears to precede both photoreceptor and inner retinal


In vitro experiments have demonstrated a
causal link between increases in cCMP
levels and retinal d e g e n e r a t i ~ 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 exposure. 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 combination of PDE inhibitor and cGMP analogue
in the incubation medium virtually destroyed every rod in the specimen over the
same incubation p e r i ~ d . ~T'h e use of
iodoacetate, a known inhibitor of
glycolysis, which also inhibits sulfhydrylcontaining enzymes such as PDE, results
in elevated cGMP levels (within 35 minutes) and visible photoreceptor degeneration within a day.72
Despite such studies and the established
elevation of cGMP in animal models of
RP,67*68the mechanism by which cCMP
leads to photoreceptor degeneration remains unresolved. Elevated cyclic nucleotide (cGMP) levels in the outer segment
of photoreceptors would place photoreceptors effectively in a state of high
activity or 'constant darkness'. The functional significance of 'constant darkness' is
that photoreceptors must maintain their
dark current at a considerable energy cost.79
It has been established by S t e i r ~ b e r gthat
~~
photoreceptors are in a state of physiological hypoxia in the dark: a hypoxia that does
not lead to functional deficits, presumably
due to the lowered energy demands during the light cycle.
Anomalies in the delivery or recycling
of the chromophore leads to a spectrum
of RP conditions including a well characterised form of Leber's congenital amaurosis involving RPE65, an RPE protein
associated with c h r o m o p h o r e recyling.^.^^ 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

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 normal rhodopsin and mutated rhodopsin
were present in the same retina, uniform
retinal degeneration was evident, although
the progression was slower than in the corresponding transgenic mice.RgThe finding
of Huang and associatesHShas important
implications for the autosomal dominant
form of RP. I t implies that the gene mutation per se does not lead to photoreceptor
degeneration but that the mutant product triggers other not yet fully understood
events (see discussion earlier), leading to
degeneration of photoreceptors that do
not have defective rhodopsin. These findings for the rhodopsin gene mutation differ from previous work using chimeras, for
rds mouse6' and the
the RCS rat
rd mouse.a In these three chimeric models (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 assoit was not clear whether
c i a t e ~However,
.~~
the chimeric models (RCS, rd, rds) were
followed to completion considering the
different time courses in the diverse animal models of RE4
From a clinical viewpoint, variations in
rod and cone sensitivity profiles suggest that
the diversity in photoreceptor degeneration is evident in human forms of the condition. Massof and Finkelsteins4 assessed
rod and cone sensitivity and reported the
existence of three groups of patients:
1. subjects with only cone mediated responses throughout their visual field
2. subjects with both rod and cone threshold elevation with the rod/cone threshold differences being similar to those
expected in normal subjects
3. subjects with cone mediated central
vision, rod and cone mediated midperipheral 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

+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 electroretinogram 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 effectivelyno 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.44Although
there was variability in the degree of visual
dysfunction within each g r o u p , t h e
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 mechan 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
Clinical and Experimental Optometry 87.2 March 2004

73

retinal function a n d 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 eGlucose
.~~~~
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
~
mitochondria] 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 hydrolysis of guanyl tri-phosphate to form cCMP
3. multiple phosphorylations of photo-

Retinitis pigmentosa Kulloniutis and Fletcher

pigment (an essential step in deactivation of opsins) .


One role of glial cells appears to involve
initial glucose catabolism with the carbon
skeleton provided to neurons predominantly 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 retinal metabolism involving the trafficking of
the carbon skeleton of glucose between
glial cells and neurons, the 'lactate trafficking 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 aerobic metabolism. In the retina, PoitryYamate, Poitry and Tsacopoulossg have
demonstrated tracking of lactate from
neurons to glia. Disruption of this shuttle
using a range of lactate transport inhibitors severely impairs retinal functiongs
however, the fact that photoreceptors can
use both glycolytic and aerobic metabolic
pathwdysy5suggests that lactate trafficking
reflects only part of the metabolic pathways available to retinal neurons.
A stable source of metabolic substrate is
required to maintain a high metabolic activity 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.73In theory, the continuously high metabolic demand in cases
of elevated cGMP provides a parsimonious explanation for a mechanism inducing photoreceptor degeneration. In contrast, retinal degeneration caused by light
damage, or that due to continuously activated rhodopsin, would have a different
underlying cause because the cyclic nucleotide channel would not be makinga high
contribution to energy demand.
The interconversion of pyruvate/lactate

is catalysed by LDH, which comes in varishow major differences between normal


ous isoenzymes. Graymoreg9first reported
especially at P15 to P20.
and rd mice,108,Lo9
t h e metabolic alteration in LDH
T h e changes in oxygen consumption,
glucose utilisation and lactic acid producisoenzyme distribution in the RCS rat compared to control rat retina. He showed a
tionlo7identify an early metabolic dysfuncdramatic alteration in the distribution of
tion in the rd mouse, well before photoreLDH isoenzymes with a marked reduction
ceptor cells die through apoptosis at
of one of the LDH isoenzymes (LDH5) at
around P10 in the rd mouse."n The chief
P7, an age when photoreceptors have not
conclusion that can be drawn from such
differentiated in the rat retina and well
work is that in two disparate models of
before anatomical alterations are evident
retinal degeneration, the RCS rat (caused
in the RCS rat.7g*'w~Lo'
The RCS rat also disby a mutation of the tyrosine kinase gene
played little change in total LDH activity
(MERTK8i*82)
and the rd mouse (caused
before degenerationIo2 b u t t h e LDH
by a mutation of the PDE genem), both
display metabolic anomalies before phoisoenzyme distribution was altered early
(at P7), before photoreceptor degeneratoreceptor degeneration.
t i ~ n However,
. ~ ~
Bonavita, Ponte a n d
AmoreIo2demonstrated a major loss of
Adult onset retinal degeneration
LDH activity after degeneration in the RCS
mimicking RP
There are forms of adult onset photorerat retina indicating that LDH activity is a
ceptor degeneration mimicking RP assosensitive indicator of photoreceptor retinal function. In addition, the RCS rat
ciated with malignancies, including
shows altered glucose utilisation and transmelanoma associated retinopathy (MAR)
a n d cancer associated retinopathy
port 9s*105-105 and neurochemical studies
(CAR).1"-114
In these conditions, individuhave demonstrated alterations before,
during and after the d e g e n e r a t i ~ n . ' ~ . ' ~ ' als suffering from small-cell carcinoma of
the lung/breast or melanomas produce
Miiller cells display abnormal metabolism
of glutamate and altered neurochemistry
antibodies as part of the body's response
to cancer. These antibodies are produced
before degeneration that continue during
against epitopes on the cancer cells that
the degenerative phase involving the
also recognise a range of proteins in the
amino acids glutamate, GABA, aspartate,
retina.'"."2."5 Circulating serum antibodglutamine and arginine. The latter change
involves neurochemical changes during/
ies must enter the retina, presumably
after degeneration, involving the amino
through breaks in the blood retinal baracids glycine and taurine, as well as u p
rier and consequently destroy retinal neutake of glutamate, GABA and g l y ~ i n e . ~ ~ , " rons
' ~ leading to RP-like signs and symptoms
in these individuals. '"J"J~~
Although proMore recently, Kalloniatis and associates Io6
showed that cation channels are constituposed, a n immunological aetiology in
typical RP has not been supported by
tively open in many photoreceptors desexperimental data.llfiHeckenlively and cotined to degenerate, before apoptotic
markers are evident. Open cation chanworkerstJ7found eight instances of serum
anti-recoverin immunoglobulins in a samnels would place photoreceptors in a state
ple of 521 typical RP cases, implying that
of constant depolarisation increasing the
an immunological aetiology is not a likely
energy demand'* or may initiate apoptotic
mechanisms.
mechanism in most cases of RP.
I n the rd mouse model, metabolic
anomalies have also been demonstrated
TREATMENT OPTIONS: WHAT DO
before degeneration. NoellIo7showed an
YOU T E U YOUR PATIENT WITH
increase in oxygen uptake, glucose utiliINHERITED RETINAL DYSTROPHY?
sation and lactic acid production (aerobic)
In certain inherited forms of retinal dysdetectable at P8 in the rd mouse retina,
trophy, where biochemical pathway
followed by a rapid decrease from P12.
anomalies lead to the degeneration, diet
Metabolic substrate concentrations and
control of foods containing ornithine or
high energy phosphate compounds do not
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 medical intervention, for example, vitamin A,
cardizem (diltiazem); activation of inhere n t protection systems, for example,
growth factors may reduce apoptosis; replacement of mutated gene; replacement
of affected cells, for example, RPE/neural
retinal transplants; and retinal prosthesis
(Sharma and Ehinger116and Lund and coworkers'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 transplant,IZ2creating chimera^,'^ growth facor differtors or apoptosis inhibitors123,124
ent light levels.'25Frasson 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 reported that an L-type calcium channel
blocker slowed the progression of retinal
degeneration in the rd mouse. L-type calcium channels are present on photor 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.IZ8The fact that D-cis-diltiazem
modulated cone calcium channels is important because it means that pharmacological manipulation may be a viable treatment 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 proposed that elevated cGMP levels lead to
photoreceptor depolarisation, with calcium 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 differences 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 targeting 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 cosuccessfully applied gene
therapy in congenital amaurosis. They reported ERG and other parameters consistent with improved visual function over 100
days post-treatment, providing the first
strong evidence of gene therapy for a condition causing a devastating early loss of
vision.'% This model also requires perinatal 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 factors may work by reducing apoptosis but
other apoptosis retarding methods are in
experimental phases. They require repeated intra-vitreal injections and growth
factors may have other undesirable actions."' Even when rod death is delayed
by survival factors, photoreceptors still
experience deconstruction of their phenotypes 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 meaningful contacts for the transplanted neural retina in doubt. More importantly, for
both retinal transplants, the degenerating
Clinical and Experimental Optometry 87.2 March 2004

75

retina shows modified neuronal and glial


s t r u c t ~ r e , ' ~implying
~ J ~ ~ that new, rogue
circuits are being formed. In retinal remodelling secondary to rod-cone retinal
degeneration, there is neurite remodelling, followed by global remodelling (see
animation at the following address: http:/
/prometheus.med.utah.edu/-marclab/
PER-full-remodeling.htm1). One exciting outcome of this research139is that the
neural retina is 'plastic' secondary to degeneration. 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 intramuscular and periorbital injection of
extracted RNA from y e a ~ t . ~ ~ . ' ~ ~
Treatment options with untested promise 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 levels, although modifying the ERG during
the treatment period, may not alter the
natural course of the disease.I4' In addition, 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 implants in RP patients'43resulted in patients
seeing spots of light that were usually
coloured (yellow/blue/yellow-green) .
Humayun and de Juan143proposed that
the resolution was up to about 1.8 degrees.
This group'44also reported that a patient
could detect spots of light and possibly the

Retinitis pigmentosa Kalloniatis and Fktcher

direction of movement using a 16-electrode array. In a study that included both


RP patients and a normal volunteer, a resolution limit of 2.25 degrees to 4.50 degrees
was achieved with an electrode array in
contact with the retina.'45z'46Often the
patients did not report percepts that
matched the stimulation pattern and frequently described multiple percepts when
one electrode was driven. The differing
results obtained from the normal-sighted
volunteer indicate that retinal degeneration alone does not explain the limited
results in blind p a t i e n t ~ , I ~providing
~.I~~
functional evidence supporting the retinal remodeling concerns of Marc and cow 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 stimulation. The long-term effect of such implants 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 encoding of visual information before it is transmitted via ganglion cells and such sensory
encoding would be required by any prosthetic device. Technological development
is still required.
A large double-masked treatment trial
of vitamin A and/or vitamin E by Berson
and associates14g 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 useful 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 advising patients on vitamin A considering
the known toxicity of this retinoid.l16This
mode of treatment has been shown to be
beneficial in other forms of retinal dystrophy and is currently recommended by the
National Institute of Health (USA)'I6

(http://www.nei.nih.gov/news/

subjects with RP for a further trial on the


benefits of vitamin A ( h t t p : / /
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 patients 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 diagnosis (PGD), where the zygome that does
not contain the mutated gene is chosen
for implantation, ensures that the identified mutation is not transferred. This technique has been applied extensively to inherited conditions such as blood disorders
and neurodegenerative diseases, providing an additional 'treatment' option in
cases where the mutation is k n ~ w n . ' ~ " J ~ '
Vitamin A at 15,000 IU daily is the recommended 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-workers152found 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 become available but other potential treatment options are at the laboratory experimental phase.

clinicalalerts/alert-rp.htm: http://

www.nei.nih.gov/news/pressreleases/
rppressrelease.htm). The National Institutes of Health is also currently recruiting

ACKNOWLEDGEMENTS

We thank Riki Gibson for compiling Figure 7 and contribution to the schematics
Clinical and Experimental Optometry 87.2 March 2004

76

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 . Haim M. Epidemiology o f retinitis
pigmentosa in Denmark. Acta Ophthalmol
Scand Suppl2002; 233: 1-34.
2. Cibis GW, Abdel-Latif AA, Bron AJ, Chalam
KV, Tripathi BJ, Tripathi RC, Wiggs J, Aaby
AA. Basic and Clinical Science CourseSection 2: Fundamental and Principles of
Ophthalmology. The foundation of the
American Academy of Ophthalmology.
2001. ~355-386.
3. Bessant DAR, Kaushal S, Bhattacharya SS.
Genetics and biology of the inherited retinal dystrophies. In: Kaufman PL, Alm A,
eds. Adler's Physiology of the Eye, 10th ed.
St Louis: Mosby; 2003. ~358-381.
4. Pacione LR, Szego MJ, Ikeda S, Nishina PM,
McInnes RR. Progress toward understanding the genetic and biochemical mechanisms of inherited photoreceptor degenerations. Annu Rev Neumsci 2003; 26: 657-700.
5. Dowling JE. The Retina: An Approachable
Part of the Brain. Cambridge MA The
Belknap Press; 1987.
6. Wu SM, Maple BR. Amino acid neurotransmitters in the retina: a functional overview.
Vision Res 1998; 38: 1371-1384.
7. Werblin FS, Dowling JE. Organization of
the retina of the mudpuppy, Necturus
maculosus. 11. Intracellular recording. J
N ~ ~ h 1969;
y s 32: 339-355.
8. Kaneko A. Receptive field organization of
bipolar and amacrine cells in the goldfish
retina. JPhysiol (Lond) 1973; 235, 133-153.
9. Stell WK, Lightfoot DO, Wheeler TG,
Leeper HF. Goldfish retina: functional polarization of cone horizontal cell dendrites
and synapses. Science 1975; 190: 989-990.
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
1985; 4: 181-219.
13. La Cour M. The retinal pigment epithelium. In: Kaufman PL, Alm A, eds. Adler's

Retinitis pigmentosa Kalloniatis and Fletcher

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


Mosby; 2003. p 348-357.
14. Mata NL, Radu RA, Clemmons RC, Travis
GH. Isomerization and oxidation of vitamin
A in conedominant retinas: a novel pathway for visual-pigment regeneration in daylight. Neuron 2002; 36: 69-80.
15. Bunt-Milam AH, Saari JC. Immunocytochemical localization of two retinoidbinding proteins in vertebrate retina. J Cell
Bioll983; 97: 703-712.
16. Bok D, Ong DE, Chytil F. Immunocytochemical 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 conductance 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 Physiology of the Eye, 10th ed. St Louis: Mosby;
2003. ~382-408.
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: 469529.
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 t p : / /
www.els.net.
23. Van Soest S, Westerveld A, de Jong PTVM,
Bleeker-Wagemakers E, Bergen AAB.
Retinitis pigmentosa: defined from a molecular point of view. Surv Ophthalmoll999;
43: 321-334.
24. Phelan JK, Bok D. A brief review of retinitis 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: http://
www.els.net.
26. Koenig R. Bardet-Biedl syndrome and
Usher syndrome. D m Ophthalmol2003; 37:
126140.
27. Daiger SP, Rossiter BJF, Greenberg J ,
Christoffels A, Hide W. Data services and
software for identifying genes and mutations causing retinal degeneration. Invest

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;
18: 689-735.
29. Marmor MF, Aguirre G, Arden G, Berson
E, Birch DG, Boughman JA, C a r 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. Edinburgh: Butterworth Heinemann: 2003.
~491-496.
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. Bilateral symmetry of vision disorders in typical retinitis pigmentosa. Br J Ophthalmol
1979; 63: 90-96.
35. Massof RW, Dagnelie G, Benzschawel T,
Palmer RW, Finkelstein D. First order dynamics 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: 405416.
38. Boughman JA, Fishman GA. A genetic
analysis of retinitis 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 surveys in Australia a n d the USA. Can J
Ophthalmol2001; 36: 315-322.
43. Kajiwara K, Berson EL, Dryja TP. Digenic
retinitis pigmentosa due to mutations at the

Clinical and Experimental Optometry 87.2 March 2004

77

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 molecular 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. MethodsMol
Biol2000; 135: 289-295.
48. Bird AC. Management of retinitis pigmentosa-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
2003; 136: 678-687.
51. Tanino T, Ohba N, Mishima S. Studies on
pigmentary retinal dystrophy. 111. A statistical 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 retinitis pigmentosa over a three-year interval.
Am JOphthalmol1985; 99: 240-251.
54. Berson EL, Rosner B, Sandberg MA,
Weigel-DiFranco C, Dryja TP. Ocular findings in patients with autosomal dominant
retinitis pigmentosa a n d rhodopsin,
proline-347-leucine. Am J Ophthalmoll991;
111: 614623.
55. Berson EL, Rosner B, Sandberg MA, Dryja
TP. Ocular findings in patients with autosoma1 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-

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 occurring rhodopsin mutation in the dog
causes retinal dysfunction and degeneration mimicking human dominant retinitis
pigmentosa. Proc Natl Acad Sci USA 2002;
99: 6328-6333.
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:
34150-34160.
61. Sanyal S, Zeilmaker GH. Development and
degeneration of retina in rds mutant mice:
light and electron microscopic observations in experimental chimaeras. Exp Eye
R ~ 1984;
s
39: 231-246.
62. Birch DG, Hood DC, Nusinowitz S ,
Pepperberg DR. Abnormal activation and
inactivation mechanisms of rod transduction 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 adaptation in autosomal dominant retinitis
pigmentosa with proline-23-histidine rhodopsin mutation. Am J Ophthalmol 1992;
113: 165-174.
64. Jin S, Cornwall MC, Oprian DD. Opsin activation as a cause of congenital night blindness. 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 o r
retinal degeneration. In: Tso MDM. Retinal Diseases. Philadelphia: JB Lippincott;
1988. p80-89.
68. Farber DB, Flannery JG, Bowes-RickmanC.
The rd mouse story: seventy years of research on an animal model of inherited
retinal degeneration. P..g Retinal Eye Res
1994; 13: 31-63.
69. McLaughlin ME, Ehrhart TL, Berson EL,

Dryja TP. Mutation spectrum of the gene


encoding the beta subunit of rod phosphodiesterase among patients with autosoma1 recessive retinitis pigmentosa. Proc
Nut1 Acad Sci USA 1995; 92: 3249-3253.
70. Ulshafer RJ, Fliesler SJ, Hollyfield JG. Differential sensitivity to protein synthesis in
human retina to a phosphodiesterase inhibitor and cyclic nucleotides. Cum Eye Res
1984; 3: 383-392.
71. Ulshafer RJ, Garcia CA,Hollyfield JG. Sensitivity of photoreceptors to elevated levels
of cGMP in the h u m a n retina. Invest
Ophthalmol Vis Sci 1980; 19:1236-1241.
72. Farber DB Souza DW, Chase DG. Cone
visual cell degeneration in ground squirrel retina: disruption of morphology and
cyclic nucleotide metabolism by iodoacetic acid. Invest Ophthalmol Vis Sci 1983; 24:
1236-1249.
73. Ames A, Li Y-Y, Heher EC, Kimble R. Energy metabolism of rabbit retina as related
to function: the high cost of Na transport.
JNeurosci 1992; 12: 840-853.
74. Steinberg RH. Monitoring communications between photoreceptors and pigment
epithelial cells: effects of mild systemic
hypoxia. Invest Ophthalmol Vis Sci 1987; 28:
1888-1904.
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 binding protein synthesis. Invest Ophthalmol Vis
Sci 1999; 40: 3-11.
76. Bok D, Hall MO. The role of the pigment
epithelium in the etiology of inherited retinal dystrophy in the rat.JCel1 B i d 1971; 49:
664-682.
77. Mullen RJ, LaVail MM. lnherited retinal
dystrophy: primary defect in pigment epithelium 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 inherited retinal dystrophy of RCS rats. Invest 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 Surgeons rat: An animal model for inherited
retinal degeneration with a still unknown
genetic defect. Acta Anatomica (Basel) 1999;
162: 101-111.
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.

Clinical and Experimental Optometry 87.2 March 2004

78

Mutations in Mertk, the human orthologue


of the RCS rat retinal dystrophy gene, cause
retinitis pigmentosa. Nature Gen 2000; 26:
270-271.
83. Huang PC, Gaitan AE, Hao Y, Petters RM,
Wong F. Cellular interactions implicated in
the mechanism of photoreceptor degeneration in transgenic mice expressing a mutant 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 programmed rod cell death. The Proctor Lecture. Invest Ophthalmol Vis Sci 1994; 35:
4182-4191.
86. Rich KA, Figueroa SL, Zhan Y,Blanks JC.
Effects of Muller cell disruption on mouse
photoreceptor cell development. Exp Eye
R ~ 1995;
s
61: 235-248.
87. Pow DV, Crook DK. Direct immunocytochemical 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 neurochemistry 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 metabolized by photoreceptors from mamrnalian retina.JNeurosci 1995; 15: 5179-5191.
90. Tsacopoulos M, Poitry-Yamate CL,
MacLeish PR, Poitry S. Trafficking of molecules and metabolic signals in the retina.
Prog Retin Eye Res 1998; 17: 429-442
91. Winkler BS. Glycolytic and oxidative metabolism 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;
41: 3183-3190.
93. Brotherton J. Studies on the metabolism of
the rat retina with special reference to
retinitis pigmentosa. I. Anaerobic glycolysis. Exp Eye Res 1962; 1: 234-245.
94. AlderVA, Cringle SJ, Constable IJ. The retinal oxygen profile of cats. Invest Ophthalmol
Vis Sci 1983; 24: 30-36.
95. Bui BV, Kalloniatis M, Vingrys AJ. The contribution of glycolytic and oxidative pathways 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 a n d neurons. J
Neurosci 1996; 16: 877-885.
98. Bui BV, Kalloniatis M, Vingrys AJ. Retinal
function loss after monocarboxylate trans-

Retinitis pigmentosa Kulloniutis and Fletcher

K, Sato N. Vaccination with recoverin, a


port inhibition. Invest Ophthalmol Vis Sci
cancer-associated retinopathy antigen, in2004: 45: 584593.
duces autoimmune retinal dysfunction and
99. Graymore C. Possible significance of the
tumor cell regression in mice. EurJImmunol
isoenzymes of lactic dehydrogenase in the
2002; 32: 2300-2307.
retina of the rat. Nature 1964; 201: 615-616.
114. KeltnerJL, Thirkill CE,Kp PT. Clinical and
100. Fletcher EL, Kalloniatis M. Localization
immunologic characteristics of melanomaof amino acid neurotransmitters during
associated retinopathy syndrome: eleven new
postnatal development of the rat retina. J
cases and a review of 51 previously published
Comp Neurol1997; 380: 449-471.
cases. JNeuroophthalmol2001; 21: 173-187.
101. Fletcher EL, Kalloniatis M . Neurochemi15. Lei B. Bush RA, Milam AH, Sieving PA.
cal development of the degenerating rat
Human melanoma-associated retinopathy
retina.J Comp Neuroll997; 388: 1-22.
(MAR) antibodies alter the retinal ON102. Bonavita V, Ponte F, Amore G. Neuroresponse of the monkey ERG in vivo. Invest
chemical studies on the inherited retinal
Ophthalmol Vis Sci 2000; 41: 262-266.
degeneration of the rat. Vision Res 1963; 3:
16. Sharma RK, Ehinger B. Management of
2 7 1-280.
hereditary retinal degeneration: present sta103. Reading HW, Sorsby A. The metabolism
tus and future directions. Sun, Ophthalml
of the dystrophic retina. I: Comparative
1999; 43: 427-444.
studies on the glucose metabolism of the
117. Heckenlively JR, Fawzi AA, Oversier J,
developing rat retina, normal and dysJordan, BL, Aptsiauri N. Autoimmune retintrophic. Vision Hes 1962; 2: 315-325.
opathy: patients with antirecoverin immu104. Stramm LE, Pautler EL. Glucose uptake
by normal and dystrophic rat retinas and cilinoreactivity and panretinal degeneration.
ary bodies. Exp Eyp Res 1980; 30: 709-718.
ATch Ophthalmol2000; 1 18: 1525-1533.
118. Wang T, Steel G, Milam AH, Valle D.
105. Di Mattio J, Zadunaisky JA. Reduced ocular glucose transport and increased nonCorrection of ornithine accumulation preelectrolyte permeability in rats with retinal
vents retinal degeneration in a mouse
degeneration (RCS). Exp Eye Hes 1983; 37:
model of gyrate atrophy of the choroid and
217-223.
retina. R o c NatlAcad Sci USA 2000; 97: 1224
106. Kalloniatis M, Tomisich G, Wellard JW,
1229
Foster LE Mapping photoreceptor and
119. Lund RD, Kwan AS, Keegan DJ, Sauve Y,
Coffey PJ, Lawrence JM. Cell transplantapost-receptoral labelling patterns using a
tion as a treatment for retinal disease. Prog
channel permeable probe (agmatine) during development in the normal and RCS
Retin Eye Res 2001; 20: 415449.
rat retina. Visual Neurosci 2002; 19: 61-70.
120. Maslim J, Valter K, Egensperger R, Hol107. Noell WK. Aspects of experimental and
lander H, Stone J. Tissue oxygen during a
hereditary retinal degneration. In:
critical developmental period controls the
Graymore C. Biochemistry of the Retina.
death and survival of photoreceptors. InLondon: Academic Press; 1965. p51-72.
vest Ophthalmol Vis Sci 1997; 38: 1667-1677.
108. Lolley RN. Changes in glucose and energy
121. Valter K, MaslimJ, Bowers F, StoneJ. Phometabolism in vivo in developing retinae
toreceptor dystrophy in the RCS rat: roles
from visually-competent (DBA/lJ) and
of oxygen, debris a n d bFGF. Invest
mutant (C3H/HeJ) mice.JNmrorhem 1972;
Ophthalmol Vis Sci 1998; 39: 2427-2442.
19: 175-185.
122. Li LX, Turner JE. Inherited retinal dys109. Lolley RN, Racz E. Changes in levels of
trophy in the RCS rat: prevention of phoATPase activity in developing retinae of
toreceptor degeneration by pigment
normal (DBA) and mutant (C3H) mice.
epithelial cell transplantation. Exp Eye Res
Vision Res 1972; 12: 567-571.
1988; 47: 911-917.
110. Portera-Cailliau C , Sung CH, Nathans J,
123. Faktorovich EG, Steinberg RH,Yasumura
Adler R. Apoptotic photoreceptor cell
D, Matthes MT, LaVail MM. Photoreceptor
death in mouse models of retinitis
degeneration in inherited retinal dystrophy
pigmentosa. h c Natl Acad Sci USA 1994;
delayed by basic fibroblast growth factor.
91: 974978.
Nature 1990; 347: 83-86.
111. Goldstein SM, Syed NA, Milam AH,
124. Katai N, Kikuchi T, Shibuki H, Kuroiwa
Maguire AM, Lawton TJ, Nichols CW.
S, Arai J , Kurokawa T, Yoshimura N.
Cance r-assoc ia ted re tin o pa t h y. Arrh
Caspaselike proteases activated in apoptotic
Ophthalmoll999; 117: 1641-1645.
photoreceptors of Royal College of Sur112. Ohguro H, Ogawa K, Maeda T, Maruyama
geons rats. Invest Ophthalmol Vis Sci 1999;
I, Maeda A, TakanoY, Nakazawa M. Retinal
40: 1802-1807.
dysfunction in cancer-associated retinopa125. Organisciak DT, Darrow RM, Barsalou L,
thy is improved by Ca(2+) antagonist adKutty RK, Wiggert B. Susceptibility to retiministration and dark adaptation. Invest
nal light damage in transgenic rats with
Ophthalmol Vis Sci 2001; 42: 2589-2595.
rhodopsin mutations. Invest Ophthalmol Vis
113. Maeda A, Maeda T, Ohguro H, Palczewski
Sci 2003; 44: 486-492.

Clinical and Experimental Optometry H7.2 March 2004

79

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 channels 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 voltagegated calcium channels in cone photoreceptors. 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 calcium 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 degeneration 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. OphthalmicRes 1997; 29: 261268.
34. Lem J, Flannery JG, Li T, Applebury ML,
Farber DB, Simon MI. Retinal degeneration is rescued in transgenic rd mice by expression 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 dysplasia 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, Jacobson SG, Hauswirth WW, Bennett J. Gene
therapy restores vision in a canine model
of childhood blindness. Nat Gaet2001; 28:
92-95.
137. LaVail MM, Yasumura D, Matthes MT,
Lau-Villacorta C, Unoki K, Sung CH,
Steinberg RH. Protection of mouse
photoreceptors by survival factors in retinal degenerations. Invest Ophthalmol Vis Sci
1998; 39: 592-602.
138. Marc RE, Jones BW, Watt CB, Strettoi E.
Neural remodeling in retinal degeneration.

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 triggered by photoreceptor degenerations. J
Comp Neurol2003;4 6 4 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. Evaluation 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 oxygen (HBO) delivery rescue retinal photoreceptors i n retinitis pigmentosa? Doc
Ophthalmoll998;97: 33-39.
143. Humayun MS, d e 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 retinal prosthesis. Vision Res2003; 43: 2573-2581.
145. Rizzo JF 3rd, WyattJ, Loewenstein J, Kelly
S, Shire D. Methods and perceptual thresholds 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
stimulation of human retina with a
microelectrode array during short-term surgical 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 replacement of degenerated photoreceptors. Ophthalmic 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 retinitis pigmentosa. Arch Ophthalmoll993; 111:
761-772.
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 technology. 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

daily in adultswith retinitis pigmentosa. Am


J Clin N U ~1999;
T
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

Das könnte Ihnen auch gefallen