Sie sind auf Seite 1von 9

ARTICLE IN PRESS

Progress in Retinal and Eye Research 23 (2004) 523–531

Do blue light filters confer protection against age-related


macular degeneration?
T.H. Margraina,*, M. Boultona, J. Marshallb, D.H. Slineyc
a
School of Optometry and Vision Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, UK
b
Department of Ophthalmology, The Rayne Institute, St. Thomas’ Hospital, London, UK
c
US Army Center for Health Promotion and Preventive Medicine, Aberdeen Proving Ground, Maryland, MD 21010-5403, USA

Abstract

Age-related macular degeneration (AMD) is a major health problem in the developed world accounting for approximately half of
all blind registrations. Current treatment options are unsuitable for the majority of patients and therefore the identification of
modifiable risk factors that may inform disease prevention programmes is a priority. This review evaluates the long-held belief that
blue light exposure has a role in the pathogenesis of AMD. Laboratory evidence has demonstrated that photochemical reactions in
the oxygen-rich environment of the outer retina lead to the liberation of cytotoxic reactive oxygen species (ROS). These ROS cause
oxidative stress which is known to contribute to the development of AMD. The precise chromopore that may be involved in the
pathogenesis of AMD is unclear but the age pigment lipofuscin is a likely candidate. Its aerobic photoreactivity and adverse effects
on antioxidant activity combined with its gradual accumulation over time suggests that its in vivo phototoxicity increases with age
despite changes in the absorption characteristics of the crystalline lens. Evidence from animal studies confirms blue light’s damaging
potential but the results are not directly applicable to macular degeneration in humans. Studies of human macular pigment density
and the risk of AMD progression following cataract surgery lend further weight to the hypothesis that blue light exposure has a role
in the pathogenesis of AMD but the epidemiological evidence is equivocal. On balance the evidences suggests but does not yet
confirm that blue light is a risk factor for AMD. Given the socio-economic impact of this disease and urgent need to identify
modifiable risk factors, future work should include a large-scale clinical trial to evaluate the effect of blue blocking filters on AMD
progression rates.
r 2004 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 524
2. Light and the pathogenesis of AMD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 524
2.1. Acute light exposure studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 525
2.2. Chronic light exposure studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 526
3. Ageing, lipofuscin and the potential for chronic light damage in humans . . . . . . . . . . . . . . . . . 526
4. Clinical evidence for the role of light in the pathogenesis of AMD . . . . . . . . . . . . . . . . . . . . 527
5. Epidemiological evidence for the role of light in the pathogenesis of AMD . . . . . . . . . . . . . . . . 528
6. Blue light filters . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 528
7. Conclusion and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 529
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 529

*Corresponding author.
E-mail address: margrainth@cardiff.ac.uk (T.H. Margrain).

1350-9462/$ - see front matter r 2004 Elsevier Ltd. All rights reserved.
doi:10.1016/j.preteyeres.2004.05.001
ARTICLE IN PRESS
524 T.H. Margrain et al. / Progress in Retinal and Eye Research 23 (2004) 523–531

1. Introduction electromagnetic spectrum may result in chronic or acute


tissue damage when it is absorbed by any one of a
Age-related macular degeneration (AMD) is the number of photosensitisers or chromophores e.g. the
leading cause of blindness in the developed world and visual pigments, melanin, melanopsin, lipofuscin, flavins
currently affects 12.7 million people in Europe and and flavoproteins. Evidence collected since the 1970s
North America (Klein et al., 1995; 1999). At present, the suggests that light may damage the retina in a number of
term AMD is used generically to describe two distinct ways involving different chromopores and cellular
pathologies: geographic atrophy (GA) and exudative events (Kremers and Van Noren, 1988). For example,
AMD. In the early stages, both conditions are asso- short-intense exposures (100 ms–10 s) may result in
ciated with pigmentary disturbances and the develop- thermal damage when incident energy is absorbed by a
ment of drusen at the posterior pole. In GA, the early tissue and there is a significant rise in temperature. The
changes lead to degeneration of the retinal pigment action spectra for this type of damage is dependent on
epithelium and receptors and in exudative AMD they the absorption characteristics of the chromopore but
lead to the growth of new vessels under the retina. GA typically peaks in the blue/green, green and red regions
accounts for the majority of AMD cases and for these of the visible spectrum (Boulton et al., 2001). Although
people there is a gradual loss of central vision eventually such exposures are used therapeutically by surgeons, to
resulting in an absolute central scotoma. For those with produce retinal photocoagulation, they are not encoun-
the exudative form of the disease the loss of central tered in the natural world.
vision is often acute. Longer exposures (typically quoted as being 10 s and
Laser photocoagulation and photodynamic therapy longer) to much less-intense light sources may cause
have been shown to reduce the risk of severe visual loss retinal damage by a photochemical mechanism and it is
for some people with the exudative form of the disease this mechanism that is most likely to be of relevance to
but for the vast majority of people with AMD there is the development of AMD.
no effective treatment. Low vision aids can ameliorate Photochemical reactions take place in normal ambi-
some of the problems associated with visual loss but ent conditions and involve a reaction between energetic
they can do little to alleviate the substantial individual photons and an absorbing molecule. In the presence of
and socio-economic costs of this disease (Margrain, oxygen this reaction leads, via a number of intermediary
1999). steps, to the production of reactive oxygen species
Ultimately, the key to reducing the number of people (ROS) including singlet oxygen, superoxide, hydrogen
with AMD is likely to be found in the axiom that peroxide and hydroxyl radicals. These ROS are highly
‘‘prevention is better than cure’’. In this paper, we toxic and can cause lipid peroxidation, protein oxidation
evaluate the evidence collected over the last three and mutagenesis (Boulton et al., 2001). ROS production
decades implicating blue light damage in the pathogen- is also a natural by-product of respiration and is
esis of AMD. We show that blue light phototoxicity believed to have a major role in ageing (Barja, 2002).
increases substantially with age and suggest that there is To minimise the destructive potential of ROS, cells
now a powerful case for the establishment of a large- have developed a number of defence mechanisms, for
scale clinical trial to evaluate the ability of blue light example enzymic (e.g. superoxide dismutase, catalase,
filters to confer protection against AMD. haeme oxygenase and phospholipases) and non-enzymic
(e.g. vitamin E, A and C) antioxidants. The RPE is
further protected from the ravages of ROS damage by
melanin which is an ROS scavenger. In particular, it has
2. Light and the pathogenesis of AMD
the capacity to minimise the production of hydroxyl,
one of the most potent oxidative moieties (Boulton et al.,
It is well established that ultraviolet (UV) and visible
2001). However, it is the photoreceptors which have
radiation has the potential to damage the retina and
perhaps developed the ultimate solution to oxidative
pigment epithelium (Noell et al., 1966). Fortunately, the
damage i.e. the continuous replacement of their cellular
human retina is protected from short-wavelength radia-
constituents (Marshall, 1985).
tion, which is particularly damaging, by the cornea
Ideally the cellular defence mechanisms, such as those
which absorbs below 295 nm and the lens which absorbs
described above, should be sufficient to combat the toxic
strongly below 400 nm.1 The human retina is therefore
effects of ROS damage. In reality, however, the defence
only exposed to the ‘‘visible component’’ of the
mechanisms are imperfect systems and the resulting
electromagnetic spectrum from 400–760 nm and some
lifetime accumulation of ROS damage may contribute
short-wavelength infra red (IR). This part of the
not just to age-related eye disease but to ageing itself
1
In childhood the retina is exposed to a small amount of UV (Barja, 2002). Indeed it has been suggested that ROS
(320 nm) because the lens transmits about 4% of incident radiation at damage to mitochondrial DNA in the RPE is a primary
this wavelength. mechanism in the pathogensis of AMD (Liang and
ARTICLE IN PRESS
T.H. Margrain et al. / Progress in Retinal and Eye Research 23 (2004) 523–531 525

Godley, 2003). The rate at which ROS damage 1


accumulates is likely to accelerate over time because
cellular defence mechanisms are known to decline with
age (Boulton et al., 2001). For reasons to be outlined

Normalised reactivity
later, we suggest that the RPE is particularly vulnerable
0.1
to age-related ROS injury because the decline in its
melanin content, associated with advancing years, is
accompanied by an increase in its lipofuscin content.
Lipofuscin is not just a potent generator of ROS but
also has an inhibitory effect on antioxidant activity 0.01
(Shamsi and Boulton, 2001).

2.1. Acute light exposure studies

Animal studies suggest that there are two distinct 0.001


types of photochemical damage, one associated with 300 350 400 450 500 550 600
short exposures operating at the level of the RPE and Wavelength (nm)
the other associated with longer, relatively less-intense
Fig. 1. Action spectra for light damage to the primate retina (symbols)
exposures, at the level of the photoreceptor outerseg- and for the aerobic photoreactivity of ocular lipofuscin (solid line).
ments. The solid line is taken directly from Fig. 1 in Rozanowska et al. (1995)
Short exposures, up to about 12 h, to relatively intense and describes the photoreactivity of lipofuscin in isolated RPE cells
short-wavelength light, sometimes referred to as the from human donors. The symbols describe the action spectra for light
‘‘blue light hazard’’, can produce damage at the level of damage to the primate retina normalised to the reactivity of lipofuscin
at 460 nm (Ham et al., 1982). The similarity of the data is remarkable.
the RPE in primates (Ham et al., 1978). The dependence
of this type of light damage on oxygen concentration
(Ham et al., 1984; Jaffe et al., 1988; Ruffolo et al., 1984)
and the ability of various antioxidants to reduce light type of ‘‘blue light damage’’. A number of components
damage (Dillon, 1991; Organisciak and Winkler, 1994) are likely to contribute to lipofuscin’s photoreactivity.
confirm its oxidative origins. There are a number of One of these components, known as A2E, has been
candidate chromopores for this type of damage most shown to produce ROS, trigger RPE cell apoptosis,
notably, melanin and lipofuscin. damage DNA and lead to RPE cell death (Sparrow and
Although capable of producing reactive oxygen Cai, 2001; Sparrow et al., 2000, 2002, 2003). However,
species at high irradiance levels, melanin seems an A2E is only weakly photoreactive and is unlikely to
unlikely candidate for the mediation of light-induced explain the photoreactivity of lipofuscin (Boulton et al.,
damage in AMD. According to Mellerio (1994) the 2001).
action spectra for ‘blue-light-damage’ does not coincide Longer exposures (typically, 12–48 h) to less-intense
with the absorption spectrum of melanin nor its action exposures produce damage at the level of the photo-
spectrum for the uptake of oxygen (Mellerio, 1994). receptors. This type of damage was initially demon-
Total melanin content of the retina reduces with age strated in the rat where rod photoreceptor degeneration
(Schmidt and Peisch, 1986) and its spatial distribution was noted after a period of constant illumination from
does not coincide with AMD. Indeed, melanin is fluorescent lamps (Noell et al., 1966). Similar findings
generally thought to be photoprotective and increased were obtained in young primates with the notable
pigmentation is generally associated with a reduced risk exception that cones were more vulnerable than rods
of AMD (Klein et al., 1999). showing degeneration after a 12 h exposure when the
Laboratory studies have shown lipofuscin to be a retinal irradiance was between 195 and 361 mW/cm2
potent generator of ROS including singlet oxygen, (Sykes et al., 1981). However, the apparent susceptibility
superoxide anion and hydrogen peroxide (Rozanowska of cones is unlikely to reflect a difference in damage
et al., 1995). These products are cytotoxic and give rise, threshold but rather a difference in repair capacity i.e.
directly or indirectly, to lipid peroxidation, protein rod outersegments are replaced more rapidly than cone
oxidation, loss of lysosomal integrity, cytoplasmic outer segments (Marshall, 1985). A number of mechan-
vacuolation and cell death (Davies et al., 2001). Most isms may contribute to light-induced photoreceptor
importantly the action spectra for photochemical damage. The photopigments themselves have long been
damage to the RPE in primate retina (Ham et al., implicated in receptoral damage. Early studies have
1976, 1982) and the aerobic photoreactivity of lipofuscin shown that the degree of light-induced damage posi-
(Rozanowska et al., 1995) are coincident (see Fig. 1) tively correlated with pre-exposure rhodopsin content
suggesting that lipofuscin is largely responsible for this and that the action spectrum for photoreceptor damage
ARTICLE IN PRESS
526 T.H. Margrain et al. / Progress in Retinal and Eye Research 23 (2004) 523–531

was similar to that of rhodopsin (Noell and Albrecht, and repair mechanisms are compromised is likely to be
1971; Organisciak and Noell, 1977). However, more quite different. On the basis that ROS damage has a
recent evidence has shown that deep blue light is 50–80 major role in ageing, it seems likely that the damage
times more efficient at causing receptoral damage than caused by chronic light exposure will manifest as
green light despite the fact that it is less efficiently accelerated ageing (Barja, 2002; Liang and Godley,
absorbed (Rapp and Smith, 1992). This observation has 2003).
been explained on the basis of rhodopsin photoreversal
(Grimm et al., 2000). That is, blue light promotes the
photoisomerisation of all-trans-retinal which leads to 3. Ageing, lipofuscin and the potential for chronic light
the regeneration of rhodopsin and an increase in damage in humans
phototransduction signalling this in turn leads to
photoreceptor apoptosis. The exact sequence of events The studies described above have shown that two
remains unclear but in relatively dim light photoreceptor distinct retinal layers are susceptible to photochemical
apoptosis is mainly dependent on a transducin-depen- injury i.e. the photoreceptor outer segments and the
dent pathway (Hao et al., 2002). Receptoral damage RPE, and that this damage is likely to be mediated by
may also result from the liberation of ROS by all-trans- the visual pigments and lipofuscin, respectively.
retinal which is a well-known photosensitiser and Although both may contribute to the development of
capable of producing singlet oxygen and superoxide AMD, and in particular geographic atrophy, there are
anions after photoexcitation with blue light (Boulton several reasons for believing that lipofuscin is of
et al., 2001). particular importance. Firstly, RPE cells are retained
On a cautionary note it should be remembered that throughout life, their repair systems operate at a
the majority of the animal studies mentioned above have molecular level and this type of ‘‘closed system’’ is more
used nocturnal rodents and it has not been established likely to accumulate ROS damage than cells of the
that the same damage pathways have a significant role in ‘‘open system’’ type in which there is abundant
diurnal species. component renewal e.g. photoreceptors (Marshall,
Furthermore, all of these investigations described 1985). Secondly, the chronology of lipofuscin accumula-
above have used exposures that are shorter and more tion in the RPE is coincident with the development of
intense than might normally be encountered in the AMD (Feeneyburns et al., 1984). Thirdly, longitudinal
natural environment i.e. they are all ‘acute’ light studies of in vivo autofluorescence (which has been
exposure studies where the damage inflicted by photo- ascribed to the presence of lipofuscin) have shown that it
chemical reactions has outstripped, at least in the short is areas of retina with the highest autofluorescence that
term, the capacity of the various defence and repair are most susceptible to degeneration (Holz et al., 2001).
mechanisms. Consequently, they are only indirectly Fortunately, the crystalline lens attenuates the shorter
related to the effects of ‘chronic’ light exposure in wavelengths reducing the potential phototoxicity of
elderly humans. What these studies do show, however, is lipofuscin. This is exemplified in Fig. 2 which shows
that light and in particular blue light has the potential to
cause retinal damage via a photochemical mechanism
and that the nature of the damage is dependent not just 30.00
on the photoreactivity of a variety of chromopores but
on the capacity of the defence and repair systems. 25.00
Relative Phototoxicity

2.2. Chronic light exposure studies 20.00

Chronic exposure to ambient lighting produces ROS 15.00


in the same way that acute exposures do because all
photochemical reactions demonstrate reciprocity be- 10.00
tween irradiance and exposure duration (Sliney, 1991).
That is, both are capable of producing damage. The 5.00
nature of that damage will reflect the capabilities of the
various defence and repair mechanisms, and the time at 0.00
which the damage is observed. For example, the acute 300 400 500 600 700
light exposure studies described above typically obtain Wavelength (nm)
measures of threshold damage in young animals after
Fig. 2. Action spectra for lipofuscin-mediated ‘blue light damage’ in
defence systems have been overwhelmed and repair the intact human eye. The curve is based on the action spectra of
systems have had insufficient time to act. Damage from lipofuscin adjusted for the transmission characteristics of the ocular
chronic light exposure in elderly humans whose defence media for a person in their 80s.
ARTICLE IN PRESS
T.H. Margrain et al. / Progress in Retinal and Eye Research 23 (2004) 523–531 527

30.00 10
9

Relative phototoxicity
25.00

Relative Phototoxicity
8
7
20.00
6
15.00 5
4
10.00 3
2
5.00
1
0.00 0
300 400 500 600 700 1st 4th 9th

(a) Wavelength (nm) (b) Decades of life

Fig. 3. Phototoxicity action spectra for three different age groups (a) and their relative phototoxicty (b). The curves are based on the aerobic
photoreactivity of lipofuscin, the age-related reduction in the transmission characteristics of the human crystalline lens and the increase in lipofuscin
content with age (Barker and Brainard, 1991; Delori et al., 2001; Rozanowska et al., 1995).

the predicted action spectra for lipofuscin-mediated 4. Clinical evidence for the role of light in the
oxidative damage in vivo. pathogenesis of AMD
Phototoxicity contributed by lipofuscin increases
substantially with age because the protective effects of The phototoxic effects of light from ophthalmic
lens senescence are offset by a substantial increase in the instruments are well documented (Davidson and Stern-
concentration of photoreactive elements in the retina berg, 1993; Jaffe et al., 1988; Michels et al., 1992;
(Delori et al., 2001). Fig. 3 describes the relative Michels and Sternberg, 1990). Although these exposures
phototoxicity of light for three age groups. Analysis of are acute, the oxygen-dependent nature of the resulting
the area under the curves shows that the potential for damage suggests an oxidative mechanism.
blue light damage increases nine fold over a life-span. Of more relevance to the pathogenesis of AMD,
This figure, which contradicts earlier suggestions that it several studies have reported the effects of an increase in
may be light exposure in childhood that is important in chronic short-wavelength light exposure. The use of
AMD, (Simons, 1993) is likely to underestimate the intraocular lenses without UV filters was found to
effect of age on lipofuscin mediated photodamage increase the incidence of cystic macular oedema follow-
because of lipofuscin’s inhibitory effect on antioxidant ing cataract surgery (Kraff et al., 1985) and epidemio-
activity (Shamsi and Boulton, 2001). logical evidence has shown that progression of AMD is
Although this phototoxicity increases steadily with approximately 2.7 times more likely following cataract
age the potential for blue light damage is likely to be extraction and intraocular lens implantation (Klein
many times greater for those developing AMD where et al., 1998; Pollack et al., 1996). The increased risk of
lipofuscin ‘hot spots’ may act as a focus for local AMD progression has been directly attributed to an
oxidative damage (Holz et al., 2001). Indeed, massive increase in blue light exposure (Cruickshanks et al.,
quantities of lipofuscin have been demonstrated in 1993; Klein et al., 1998). (Werner et al., 1989) showed a
the RPE of eyes with atrophic AMD (Sarks et al., loss of blue colour vision in pseudoaphakic eyes without
1988). The hypothesis, that lipofuscin-mediated photo- UV filtration.
chemical damage increases with age, and particularly so Additional evidence suggesting that blue light has a
for those developing AMD, may explain why antiox- role in the development of AMD comes from studies
idant therapy is beneficial in these groups (Kassoff et al., which indicate that macular pigment is protective
2001). (Beatty et al., 1999; Weiter et al., 1988). Macular
If light were to have a role in the pathogenesis of pigment has a broad band absorbance spectra peaking
AMD the evidence outlined above enables us to make at 460 nm. It is therefore, particularly effective at
several predictions. Firstly, increased exposure to short- reducing the potentially damaging effects of lipofuscin
wavelength radiation, both in terms of intensity and whose photoreactivity peaks at 450 nm in elderly adults
duration, should increase the risk of AMD. Secondly, (see Fig. 2). Estimates of macular pigment density in the
exposure to short-wavelength radiation in old age will human eye suggest that it typically absorbs about 70%
be a greater risk than exposure at other times of life. of incident light at this wavelength. Although, macular
Thirdly, antioxidants should reduce the risk of AMD pigment is found in all retinal layers it reaches its highest
and more specifically their protective effects should be concentration at the receptor axon and the inner
greatest for those with the greatest concentration of plexiform layers and it is therefore well placed to act
lipofuscin. The following paragraphs examine the as a light filter. Unfortunately, despite its name macular
evidence for these predictions. pigment only confers protection to the fovea because its
ARTICLE IN PRESS
528 T.H. Margrain et al. / Progress in Retinal and Eye Research 23 (2004) 523–531

concentration drops precipitously from the foveal pit i.e. and particularly for those with the early signs of AMD
the optical density of macular pigment drops to half its that is the quantity of interest. In support of the notion
value 1.5 from fixation (Kilbride et al., 1989). The that oxidative stress is likely to be most damaging for
topographical distribution of macular pigment may those at high risk of developing AMD, a recent large-
explain the pattern of cell loss observed in histological scale clinical trial found that the therapeutic effects of
studies of early AMD (Curcio, 2001; Curcio et al., 2000; antioxidant therapy were greatest in those with estab-
Kilbride et al., 1989). Of course, macular pigment also lished disease (Kassoff et al., 2001).
has the potential to protect the retina by neutralising
reactive oxygen species (Schalch, 1992) but its location
in the receptor axon layer and the inner plexiform layer, 6. Blue light filters
some distance from the site where reactive oxygen
species are produced does not facilitate this potential. A number of investigators have proposed the use of
The potential for light to cause retinal damage may be sunglasses which attenuate short-wavelength light to
increased in people with a history of above-average light reduce the phototoxic potential of light (Fishman, 1986;
exposure because macular pigment density is inversely Ham et al., 1976; Sliney, 2001; Werner and Schmidt,
related to light exposure (Mellerio et al., 2002). That is, 1975). However, apart from two small scale trials on
the potentially damaging effects of light exposure are three people with retinitis pigmentosa the possible
compounded by a reduction in the optical density of MP therapeutic effect of short-wavelength light deprivation
associated with chronic light exposure. has not been evaluated (Berson, 1980; Werner and
It is also of interest to note that the optical density of Schmidt, 1975).
macular pigment is significantly reduced in smokers The ideal absorption characteristics of sunglasses or
(Hammond et al., 1996). The reduction in optical intraocular lenses (IOL) have not yet been established
density would increase blue light exposure and this (Mainster and Sparrow, 2003). However, sunglasses or
would be consistent with a blue-light contribution to the intraocular lenses with the transmission characteristics
heightened risk of AMD in smokers (Smith et al., 1996). shown in Fig. 4 reduce the amount of short-wavelength
light reaching the retina and have a yellow/orange and
bronze appearance. Calculations indicate that these
5. Epidemiological evidence for the role of light in the filters would reduce the in vivo aerobic photoreactivity
pathogenesis of AMD of lipofuscin by approximately 90%. The notion that a
yellow filter can provide this level of protection is
There have been several population-based studies that supported by a recent laboratory study which has shown
have evaluated the role of ultraviolet and visible light on that a ‘yellow’ IOL can produce an 80% reduction in
the development of AMD (Cruickshanks et al., 1993; death of A2E laden RPE cells exposed to light (Sparrow
Darzins et al., 1997; Hyman et al., 1983; Taylor et al., et al., 2004). For reasons given earlier the prophylactic
1990, 1992; West et al., 1989). effect of such filters is likely to be greatest in the elderly
An extensive survey of the watermen in the Chesa- and those at high risk AMD. There may be an added
peake Bay area concluded that chronic exposure to blue
or visible light may be related to the development of 100
AMD (Taylor et al., 1992). Similarly, the authors of the yellow /orange
Beaver Dam Eye Study also suggest that their measures
indicate that visible light rather than UV might be 80

associated with AMD (Cruickshanks et al., 1993).


Transmission %

Conversely, Darzins et al. (1997) and the Eye Disease 60


Case Control Study Group (1992) found no such bronze
relationship (Darzins et al., 1997; Eye Disease Case
40
Control Study Group, 1992).
The equivocal findings reported in the epidemiologi-
cal literature are quite unremarkable. Firstly, the 20

absence of a relationship with UV simply confirms that


the adult lens absorbs almost all radiation below 0
400 nm. Secondly, the assumption has been that it is 350 400 450 500 550 600 650 700
lifetime exposure to sunlight that is the relevant variable Wavelength (nm)
but this is unlikely to be the case. As already stated the
Fig. 4. Transmission characteristics of two filters that have a yellow/
phototoxicity of blue light increases with age and is orange and bronze appearance. Both filters reduce in vivo lipofuscin-
likely to be particularly great for those with lipofuscin mediated photoreactivity (see Fig. 3a) by approximately 90% in people
‘‘hot spots’’. Therefore, it is light exposure in old age in their 80s.
ARTICLE IN PRESS
T.H. Margrain et al. / Progress in Retinal and Eye Research 23 (2004) 523–531 529

visual benefit for this group of people when using the However, it would be premature to recommend the
yellow/orange filter because blue light is selectively wide spread use of blue blocking lenses in the elderly
scattered by the ocular media and its attenuation has because although there is considerable circumstantial
been associated with improvements in contrast sensitiv- evidence for such a measure there is no direct evidence
ity and a reduction in glare sensitivity (Frennesson and that environmental light causes retinal damage. How-
Nilsson, 1993; Leat et al., 1990; Wolffsohn et al., 2000). ever, we conclude that there are now three compelling
Indeed many people with low vision show a preference reasons for undertaking a large-scale clinical trial to
for yellow spectacle lenses despite their adverse impact evaluate the prophylactic effects of blue light filtration
on colour vision (Rosenblum et al., 2000). However, for in AMD. Firstly, there are now sound reasons for
those with normal vision, and in particular for those suspecting that blue light exposure in old age may
who drive, the bronze filter may be preferable because contribute to the development of AMD. Secondly, the
although such a filter attenuates blue light is does not debate on the role of blue light exposure and AMD has
block blue light altogether and therefore it would have a continued for almost three decades and only a large-
less detrimental effect on colour vision. scale randomised clinical trial will have sufficient power
Although, it seems likely that filters that attenuate to provide conclusive evidence. Thirdly, even a modest
blue light will have a beneficial effect in terms of retinal beneficial effect is likely to be associated with substantial
photoprotection there may be unwanted side effects. For individual and socio-economic benefits because AMD is
example, the action spectra for photosensitive ganglion reaching epidemic proportions (Evans and Wormald,
cells, that are thought to have a role in setting our 1996).
circadian clock, peaks at 484 nm (Berson et al., 2002).
Consequently, it is possible that blue light filters may
disrupt sleep cycles. However, although sleep disruption
References
has been reported in people who are ‘blind’ (Tabandeh
et al., 1998), the effect of the blue light filters proposed is Barja, G., 2002. Endogenous oxidative stress: relationship to aging,
unlikely to be dramatic. This is because the action longevity and caloric restriction. Ageing Res. Rev. 1, 397–411.
spectra is only weakly tuned to short-wavelength light Barker, F.M., Brainard, G.C., 1991. The direct spectral transmission
and multiple types of receptors are thought to con- of the excised human lens as a function of age. FDA Research
tribute to photic entrainment (Berson et al., 2002; Ruby Report No. 785346, PA, USA.
Beatty, S., Boulton, M., Henson, D., Koh, H.H., Murray, I.J., 1999.
et al., 2002). Macular pigment and age related macular degeneration. Br. J.
Ophthalmol. 83, 867–877.
Berson, D.M., Dunn, F.A., Takao, M., 2002. Phototransduction by
7. Conclusion and future directions retinal ganglion cells that set the circadian clock. Science 295,
1070–1073.
Berson, E.L., 1980. Light deprivation and retinitis pigmentosa. Vision
On the basis of the evidence presented here we believe Res. 20, 1179.
that there is support for the long-held belief that light Boulton, M., Rozanowska, M., Rozanowski, B., 2001. Retinal
has a role in the pathogenesis of AMD. That is, the photodamage. J. Photochem. Photobiol. B-Biol. 64, 144–161.
recent finding that antioxidant therapy has a protective Cruickshanks, K.J., Klein, R., Klein, B.E.K., 1993. Sunlight and age-
related macular degeneration—the Beaver Dam Eye Study. Arch.
effect confirms that oxidative stress has a role in the
Ophthalmol. 111, 514–518.
pathogenesis of AMD and laboratory studies have Curcio, C.A., 2001. Photoreceptor topography in ageing and age-
demonstrated that light, and in particular blue light, is a related maculopathy. Eye 15, 376–383.
source of oxidative stress via its interaction with retinal Curcio, C.A., Owsley, C., Jackson, G.R., 2000. Spare the rods, save the
chromophores. Therefore, a reduction in blue light cones in aging and age-related maculopathy. Invest. Ophthalmol.
exposure might reasonably be expected to reduce Vis. Sci. 41, 2015–2018.
Darzins, P., Mitchell, P., Heller, R.F., 1997. Sun exposure and age-
progression in AMD. related macular degeneration—an Australian case-control study.
The precise chromopore(s) that may be involved in Ophthalmology 104, 770–776.
the pathogenesis of AMD remains unclear, but the age Davidson, P.C., Sternberg, P., 1993. Potential retinal phototoxicity.
pigment lipofuscin which has long been associated with Am. J. Ophthalmol. 116, 497–501.
AMD is a likely candidate. Its aerobic photoreactivity Davies, S., Elliott, M.H., Floor, E., Truscot, T.G., Zareba, M., et al.,
2001. Photocytotoxicity of lipofuscin in human retinal pigment
and adverse effects on antioxidant activity combined epithelial cells. Free Radical Biol. Med. 31, 256–265.
with its gradual accumulation over time suggests that Delori, F.C., Goger, D.G., Dorey, C.K., 2001. Age-related accumula-
retinal phototoxicity increases substantially with age tion and spatial distribution of lipofuscin in RPE of normal
despite changes in the absorption characteristics of the subjects. Invest. Ophthalmol. Vis. Sci. 42, 1855–1866.
Dillon, J., 1991. The photophysics and photobiology of the eye.
crystalline lens. This hypothesis suggests that the
J. Photochem. Photobiol. B-Biol. 10, 23–40.
potential for blue light to cause retinal damage is Evans, J., Wormald, R., 1996. Is the incidence of registrable age-
greatest in the elderly and those at risk of AMD related macular degeneration increasing? Br. J. Ophthalmol. 80,
progression. 9–14.
ARTICLE IN PRESS
530 T.H. Margrain et al. / Progress in Retinal and Eye Research 23 (2004) 523–531

Eye Disease Case Control Study Group, 1992. Risk factors for Leat, S.J., North, R.V., Bryson, H., 1990. Do long wavelength pass
neovascular age-related macular degeneration. Arch. Ophthalmol. filters improve low vision performance. Ophthalmic Physiol. Opt.
110, 1701–1708. 10, 219–224.
Feeneyburns, L., Hilderbrand, E.S., Eldridge, S., 1984. Aging human Liang, F.Q., Godley, B.F., 2003. Oxidative stress-induced mitochon-
RPE—morphometric analysis of macular, equatorial, and periph- drial DNA damage in human retinal pigment epithelial cells: a
eral cells. Invest. Ophthalmol. Vis. Sci. 25, 195–200. possible mechanism for RPE aging and age-related macular
Fishman, G.A., 1986. Ocular phototoxicity—guidelines for selecting degeneration. Exp. Eye Res. 76, 397–403.
sunglasses. Surv. Ophthalmol. 31, 119–124. Mainster, M.A., Sparrow, J.R., 2003. How much blue light should an
Frennesson, I.C., Nilsson, U.L., 1993. Contrast sensitivity peripheral IOL transmit? Br J. Ophthalmol. 87, 1523–1529.
to an absolute central scotoma in age-related macular degeneration Margrain, T.H., 1999. Minimising the impact of visual impairment:
and the influence of a yellow or an orange filter. Doc. Ophthalmol. low vision aids are a simple way of alleviating impairment. Br.
84, 135–144. Med. J. 318, 1504–1504.
Grimm, C., Reme, C.E., Rol, P.O., Williams, T.P., 2000. Blue light’s Marshall, J., 1985. Radiation and the ageing eye. Ophthalmic Physiol.
effects on rhodopsin: photoreversal of bleaching in living rat eyes. Opt. 5, 241–263.
Invest. Ophthalmol. Vis. Sci. 41, 3984–3990. Mellerio, J., 1994. Light effects on the retina. In: Albert, D.M.,
Ham, W.T., Mueller, H.A., Ruffolo, J.J., Guerry, D., Guerry, R.K., Jakobiec, F.A. (Eds.), Principles and Practice of Ophthalmology:
1982. Action spectrum for retinal injury from near-ultraviolet Basic Sciences. Saunders, Philadelphia, pp. 1326–1345.
radiation in the Aphakic monkey. Am. J. Ophthalmol. 93, 299–306. Mellerio, J., Ahmadi-Lari, S., van Kuijk, F.J.G.M., Pauleikhoff, D.,
Ham, W.T., Mueller, H.A., Ruffolo, J.J., Millen, J.E., Cleary, S.F., Bird, A.C., Marshall, J., 2002. A portable instrument for
et al., 1984. Basic mechanisms underlying the production of measuring macular pigment with central fixation. Curr. Eye Res.
photochemical lesions in the mammalian retina. Curr. Eye Res. 3, 25, 37–47.
165–174. Michels, M., Lewis, H., Abrams, G.W., Han, D.P., Mieler, W.F., et al.,
Ham, W.T., Mueller, H.A., Sliney, D.H., 1976. Retinal sensitivity to 1992. Macular phototoxicity caused by fiberoptic endoillumination
damage from short-wavelength light. Nature 260, 153–155. during pars-plana vitrectomy. Am. J. Ophthalmol. 114, 287–296.
Ham, W.T., Ruffolo, J.J., Mueller, H.A., Clarke, A.M., Moon, M.E., Michels, M., Sternberg, P., 1990. Operating microscope-induced
1978. Histologic analysis of photochemical lesions produced in retinal phototoxicity–pathophysiology, clinical manifestations and
rhesus retina by short-wavelength light. Invest. Ophthalmol. Vis. prevention. Surv. Ophthalmol. 34, 237–252.
Sci. 17, 1029–1035. Noell, W.K., Albrecht, R., 1971. Irreversible effects of visible light on
Hammond, B.R., Wooten, B.R., Snodderly, D.M., 1996. Cigarette the retina: role of vitamin A. Science 172, 76–80.
smoking and retinal carotenoids: implications for age-related Noell, W.K., Walker, V., Kang, B., Berman, S., 1966. Retinal damage
macular degeneration. Vision Res. 36, 3003–3009. by visible light. Invest. Ophthalmol. Vis. Sci. 5, 450–473.
Hao, W.S., Wenzel, A., Obin, M.S., Chen, C.K., Brill, E., et al., 2002. Organisciak, D.T., Noell, W.K., 1977. The rod outer segment
Evidence for two apoptotic pathways in light-induced retinal phospholipid/opsin ratio of rats maintained in darkness or cyclic
degeneration. Nature Genet. 32, 254–260. light. Invest. Ophthalmol. Vis. Sci. 16, 188–190.
Holz, F.G., Bellman, C., Staudt, S., Schutt, F., Volcker, H.E., 2001. Organisciak, D.T., Winkler, B.S., 1994. Retinal light damage: practical
Fundus autofluorescence and development of geographic atrophy and theoritical considerations. Prog. Ret. Eye Res. 13, 1–29.
in age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. Pollack, A., Marcovich, A., Bukelman, A., Oliver, M., 1996. Age
42, 1051–1056. related macular degeneration after extracapsular cataract extrac-
Hyman, L.G., Lilienfeld, A.M., Ferris, F.L., Fine, S.L., 1983. Senile tion with intraocular lens implantation. Ophthalmology 103,
macular degeneration: a case-control study. Am. J. Epidemiol. 118, 1546–1554.
213–227. Rapp, L.M., Smith, S.C., 1992. Morphologic comparisons between
Jaffe, G.J., Irvine, A.R., Wood, I.S., Severinghaus, J.W., Pino, G.R., rhodopsin-mediated and short-wavelength classes of retinal light
et al., 1988. Retinal phototoxicity from the operating microscope: damage. Invest. Ophthalmol. Vis. Sci. 33, 3367–3377.
the role of inspired oxygen. Ophthalmology 95, 1130–1141. Rosenblum, Y.Z., Zak, P.P., Ostrovsky, M.A., Smolyaninova, I.L.,
Kassoff, A., Kassoff, J., Buehler, J., Eglow, M., Kaufman, F., et al., Bora, E.V., et al., 2000. Spectral filters in low-vision correction.
2001. A randomized, placebo-controlled, clinical trial of high-dose Ophthalmic Physiol. Opt. 20, 335–341.
supplementation with vitamins C and E, beta carotene, and zinc for Rozanowska, M., Jarvisevans, J., Korytowski, W., Boulton, M.E.,
age-related macular degeneration and vision loss—AREDS Report Burke, J.M., et al., 1995. Blue light-induced reactivity of retinal age
No. 8. Arch. Ophthalmol. 119, 1417–1436. pigment—in-vitro generation of oxygen-reactive species. J. Biol.
Kilbride, P.E., Alexander, K.R., Fishman, M., Fishman, G.A., 1989. Chem. 270, 18825–18830.
Human macular pigment assessed by imaging fundus reflectometry. Ruby, N.F., Brennan, T.J., Xie, X.M., Cao, V., Franken, P., et al.,
Vision Res. 29, 663. 2002. Role of melanopsin in circadian responses to light. Science
Klein, R., Klein, B.E.K., Cruickshanks, K.J., 1999. The prevalence of 298, 2211–2213.
age-related maculopathy by geographic region and ethnicity. Prog. Ruffolo, J.J., Ham, W.T., Mueller, H.A., Millen, J.E., 1984.
Retin. Eye Res. 18, 371. Photochemical lesions in the primate retina under conditions of
Klein, R., Klein, B.E.K., Jensen, S.C., Cruickshanks, K.J., 1998. The elevated blood-oxygen. Invest. Ophthalmol. Vis. Sci. 25, 893–898.
relationship of ocular factors to the incidence and progression of Sarks, J.P., Sarks, S.H., Killingsworth, M.C., 1988. Evolution of
age-related maculopathy. Arch. Ophthalmol. 116, 506–513. geographic atrophy of the retinal-pigment epithelium. Eye 2,
Klein, R., Wang, Q., Klein, B.E.K., Moss, S.E., Meuer, S.M., 1995. 552–577.
The relationship of age-related maculopathy, cataract, and glauco- Schalch, W., 1992. Caretenoids in the retina—a review of their possible
ma to visual-acuity. Invest. Ophthalmol. Vis. Sci. 36, 182–191. role in preventing or limiting damage caused by light and oxygen.
Kraff, M.C., Sanders, D.R., Jampol, L.M., Lieberman, H.L., 1985. In: Emerit, E., Chance, B. (Eds.), Free Radicles and Aging.
Effect of an ultraviolet-filtering intraocular-lens on cystoid macular Birkhauser, Basel, pp. 280–298.
edema. Ophthalmology 92, 366–369. Schmidt, S.Y., Peisch, R.D., 1986. Melanin concentration in normal
Kremers, J.J.M., Van Noren, D., 1988. Two classes of light damage of human retinal pigment epithelium. Invest. Ophthalmol. Vis. Sci.
the retina. Lasers Light Ophthalmol 2, 41–53. 27, 1063–1067.
ARTICLE IN PRESS
T.H. Margrain et al. / Progress in Retinal and Eye Research 23 (2004) 523–531 531

Shamsi, F.A., Boulton, M., 2001. Inhibition of RPE lysosomal and Sparrow, J.R., Miller, A.S., Zhou, J., 2004. Blue light-absorbing
antioxidant activity by the age pigment lipofuscin. Invest. intraocular lens and retinal pigment epithelium protection in vitro.
Ophthalmol. Vis. Sci. 42, 3041–3046. J. Cataract Refract. Surg. 30, 873–878.
Simons, K., 1993. Artificial-light and early life exposure in age-related Sykes, S.M., Robison, W.G., Waxler, M., Kuwabara, T., 1981.
macular degeneration and in cataractogenic phototoxicity. Arch. Damage to the monkey retina by broad-spectrum fluorescent light.
Ophthalmol. 111, 297–298. Invest. Ophthalmol. Vis. Sci. 20, 425–434.
Sliney, D.H., 1991. Measurement of light and the geometry Tabandeh, H., Lockley, S.W., Buttery, R., Skene, D.J., Defrance, R.,
of exposure of the human eye. In: Marshall, J. (Ed.), The Arendt, J., Bird, A.C., 1998. Disturbance of sleep in blindness. Am.
Susceptible Visual Apparatus., Vol. 16. Macmillan Press, London, J. Ophthalmol. 126, 707–712.
pp. 20–23. Taylor, H.R., Munoz, B., West, S., Bressler, N.M., Bressler, S.B., et al.,
Sliney, D.H., 2001. Photoprotection of the eye—UV radiation and 1990. Visible light and risk of age-related macular degeneration.
sunglasses. J. Photochem. Photobiol. B-Biol. 64, 166–175. Trans. Am. Ophthalmol. Soc. 88, 163–173.
Smith, W., Mitchell, P., Leeder, S.R., 1996. Smoking and age-related Taylor, H.R., West, S., Munoz, B., Rosenthal, F.S., Bressler, S.B.,
maculopathy—the Blue Mountains Eye Study. Arch. Ophthalmol. et al., 1992. The long-term effects of visible-light on the eye. Arch.
114, 1518–1523. Ophthalmol. 110, 99–104.
Sparrow, J.R., Cai, B.L., 2001. Blue light-induced apoptosis of A2E- Weiter, J.J., Delori, F., Dorey, C.K., 1988. Central sparing in annular
containing RPE: involvement of caspase-3 and protection by bcl-2. macular degeneration. Am. J. Ophthalmol. 106, 286–292.
Invest. Ophthalmol. Vis. Sci. 42, 1356–1362. Werner, A., Schmidt, I., 1975. Photic damage in retinitis pigmentosa
Sparrow, J.R., Nakanishi, K., Parish, C.A., 2000. The lipofuscin and a suggestion for a protective device. J. Ame. Optom. Assoc. 46,
fluorophore A2E mediates blue light-induced damage to retinal 380–386.
pigmented epithelial cells. Invest. Ophthalmol. Vis. Sci. 41, Werner, J.S., Steele, V.G., Pfoff, D.S., 1989. Loss of human
1981–1989. photoreceptor sensitivity associated with chronic exposure to
Sparrow, J.R., Zhou, J., Ben-Shabat, S., Vollmer, H., Itagaki, Y., et al., ultraviolet-radiation. Ophthalmology 96, 1552–1558.
2002. Involvement of oxidative mechanisms in blue-light-induced West, S.K., Rosenthal, F.S., Bressler, N.M., Bressler, S.B., Munoz, B.,
damage to A2E-laden RPE. Invest. Ophthalmol. Vis. Sci. 43, et al., 1989. Exposure to sunlight and other risk-factors for age-
1222–1227. related macular degeneration. Arch. Ophthalmol. 107, 875–879.
Sparrow, J.R., Zhou, J.L., Cai, B.L., 2003. DNA is a target of the Wolffsohn, J.S., Cochrane, A.L., Khoo, H., Yoshimitsu, Y., Wu, S.,
photodynamic effects elicited in A2E-laden RPE by blue-light 2000. Contrast is enhanced by yellow lenses because of selective
illumination. Invest. Ophthalmol. Vis. Sci. 44, 2245–2251. reduction of short-wavelength light. Optom. Vis. Sci. 77, 73–81.

Das könnte Ihnen auch gefallen