Download Complete Textbook of Vitreoretinal Diseases and Surgery 1st Edition Edition S Natarajan PDF for All Chapters
Download Complete Textbook of Vitreoretinal Diseases and Surgery 1st Edition Edition S Natarajan PDF for All Chapters
Download Complete Textbook of Vitreoretinal Diseases and Surgery 1st Edition Edition S Natarajan PDF for All Chapters
com
https://ebookultra.com/download/textbook-of-
vitreoretinal-diseases-and-surgery-1st-edition-
edition-s-natarajan/
https://ebookultra.com/download/comprehensive-textbook-of-surgery-1st-
edition-dinesh-vyas/
ebookultra.com
https://ebookultra.com/download/textbook-of-oral-and-maxillofacial-
surgery-1st-edition-rajiv-m-borle/
ebookultra.com
https://ebookultra.com/download/logan-turner-s-diseases-of-the-nose-
throat-and-ear-head-and-neck-surgery-11th-edition-hussain/
ebookultra.com
https://ebookultra.com/download/textbook-of-pleural-diseases-hodder-
arnold-publication-2nd-edition-richard-w-light/
ebookultra.com
Textbook of Small Animal Surgery 2 Volume Set 3rd Edition
Douglas Slatter
https://ebookultra.com/download/textbook-of-small-animal-
surgery-2-volume-set-3rd-edition-douglas-slatter/
ebookultra.com
https://ebookultra.com/download/acsi-textbook-on-cutaneus-and-
aesthetic-surgery-1st-edition-mysore-venkataram-et-al/
ebookultra.com
https://ebookultra.com/download/matrix-methods-of-structural-analysis-
theory-and-problems-1st-edition-c-natarajan/
ebookultra.com
https://ebookultra.com/download/vascular-and-endovascular-surgery-a-
comprehensive-review-textbook-with-cd-rom-7th-edition-wesley-moore/
ebookultra.com
https://ebookultra.com/download/textbook-of-neuro-oncology-1st-
edition-mitchel-s-berger/
ebookultra.com
Textbook of Vitreoretinal Diseases and Surgery 1st
Edition Edition S Natarajan Digital Instant Download
Author(s): S Natarajan , N Hussain
ISBN(s): 9788184486193, 8184486197
Edition: 1st Edition
File Details: PDF, 9.72 MB
Year: 2009
Language: english
Textbook of
Vitreoretinal Diseases
and Surgery
Textbook of
Vitreoretinal Diseases
and Surgery
Chief Editors
S Natarajan DO FMRF
Chief Managing Director
Aditya Jyot Eye Hospital Pvt Ltd
Mumbai, India
Co-Editor
Supriya Dabir MD DNB
Vitreoretinal Consultant
Aditya Jyot Eye Hospital Pvt Ltd
Mumbai, India
Foreword
SS Badrinath and Tarun Sharma
USA Office
1745, Pheasant Run Drive, Maryland Heights (Missouri), MO 63043, USA, Ph: 001-636-6279734
e-mail: jaypee@jaypeebrothers.com, anjulav@jaypeebrothers.com
All rights reserved. No part of this publication should be reproduced, stored in a retrieval system, or transmitted in any form
or by any means: electronic, mechanical, photocopying, recording, or otherwise, without the prior written permission of the
editors and the publisher.
This book has been published in good faith that the material provided by contributors is original. Every effort is made
to ensure accuracy of material, but the publisher, printer and editors will not be held responsible for any inadvertent
error(s). In case of any dispute, all legal matters to be settled under Delhi jurisdiction only.
vi
Foreword
After the great triumph of the 8th International Advanced Vitreoretinal Surgery Conference held in
picturesque Kuala Lumpur early in 2008 with world renowned faculty, it became imperative for me
to share the thoughts, speeches and ideas which made the conference a resounding success.
As the Chairman of the Scientific Committee for this conference, I feel extremely privileged to
bring you this book which marks the first such successful compilation of the collection of scientific
sessions with bridging plenary lectures focusing on the updates in vitreoretinal diseases.
This anthology addresses the latest treatment modalities in both medical as well as surgical
retina. It also highlights the painstaking research in genetics, stem cell therapy, lasers etc. Apart
from this, focus has been on the utility of various advanced diagnostic modalities in day-to-day
practice.
This book is meant to cater to not only to nascent retinal surgeons but also practicing surgeons as
an update to their knowledge. Since the conference limits the audience to a few, this book is meant
to rejuvenate the information on the updates in the field of vitreoretinal surgery to all those unable
to attend the meet.
I would like to thank all the guests and invited speakers from across the world from the bottom
of my heart. Their time and contributions are greatly cherished and undoubtedly provide the icing
on the academic cake. A book of this stature would be incomplete without its scientific programme
and I am duty bound to felicitate all those who have provided scientific contributions. I would also
like to thank and appreciate Dr Nazimul Hussain and Dr Supriya Dabir who have worked tirelessly
with me and contributed immensely in completing the book.
Ultimately, they have justified the efforts of the Organising and Scientific Committees.
Thank you for granting me the privilege of serving as the Chairman of the Committee.
S Natarajan
Contents
xii
Textbook of Vitreoretinal Diseases and Surgery
Abstract
Age-related macular degeneration (AMD) is a late-onset complex disorder with multifactorial etiologies. Both genetic
and environmental factors play a role in the disease pathogenesis. AMD is the third leading cause of blindness in the
elderly. Familial aggregation, segregation studies and linkage analysis have provided both qualitative and quantitative
evidence on the genetic basis in AMD. Several candidate loci have been earlier mapped in AMD but variants in genes viz.
APOE, ABCA4, FBLN6 and EFEMP1 harboring these loci have accounted for only a small proportion of cases. Recent
screening of two major loci has led to the identification of the Complement Factor H (CFH) on 1q32 and LOC387715 and
HTRA1 on the 10q26 gene cluster. Single nucleotide polymorphisms (SNPs) in CFH (Y402H), LOC387715 (A69S) and a
promoter variant in HTRA1 have been associated with AMD in large case-control cohorts. These SNPs exhibited large
effect sizes and high disease odds for the risk genotypes across different populations. Interestingly, these associations
have been widely replicated across multiple ethnic groups worldwide indicating their potential role in the disease
pathogenesis. In this article, we would outline the genetics of AMD with special emphasis on CFH followed by other
genetic variants based on studies done by our group and colleagues worldwide. We would also provide a brief overview
on the possible molecular mechanisms leading to AMD.
Introduction
Visual impairment leading to blindness is a major impediment towards growth and development in
populations worldwide. The burden of blindness is a global challenge that needs to be tackled on a
war footing. Recent estimates of the World Health Organization (WHO) indicate that around 45
million people are blind and 135 million are visually impaired worldwide and 90% of these people
live in the developing countries. Among the blinding conditions, cataract accounts for 60% of global
blindness, followed by glaucoma (12.3%). Age-related macular degeneration (AMD) is the third
leading cause of blindness and accounts for 8.7% of the world population (WHO Fact sheet no. 282).
Although AMD is more common among the elderly in developed countries, it is also becoming a
cause of concern in the developing countries, with fast demographic changes in lifestyle and
senescence.
The Problem
AMD causes progressive impairment of central vision and is a leading cause of irreversible vision
loss worldwide. The overall prevalence of late stage AMD varies from 1.4 to 1.7% across different
epidemiological cohorts (Klein et al, 1992; Mitchell et al, 1995) and increases significantly with age.
The prevalence of AMD in India ranges from 1.84-2.7% (Nirmalan et al, 2004), similar to the global
prevalence. It is estimated that by the year 2020, around 8 million people will have vision loss due to
retinal complications including AMD (Bressler, 2002). As it leads to irreversible blindness, managing
AMD is a global public health challenge.
GENDER
The incidences of early and late AMD are 2 and 7 times more in females above > 75 years of age,
compared to males below 75 years of age (Klaver et al, 1998). The high risk in females may be due to
the loss of a protective effect of estrogens in postmenopausal women.
RACE
The prevalence of AMD was shown to vary greatly among different races. All forms of AMD are
more prevalent in the white population than darkly pigmented races (Bressler et al, 2008; Friedman
et al, 1999). Although the exact mechanism is not known, it was suggested that may melanin protect
against the formation of lipofuscin, which is a marker of cellular senescence and promoter of oxidative
damage (Kawasaki et al, 2008). A recent study found no differences in macular or melanin pigment
densities between eyes with and without early AMD (Kayatz et al, 2001).
CIGARETTE SMOKING
Smoking has been postulated to cause AMD by depression of serum antioxidant levels and alteration
of choroidal blood flow and detoxification of the retinal pigment epithelium (RPE). It has been
hypothesized that decrease in luteal pigments in human retina due to cigarette smoking may cause
oxidative damage to the macula, thereby leading to an increased risk of developing AMD. Many
population-based studies have shown the association of smoking with increased risk of AMD
(Berendschot et al, 2002; AREDS 2000; Delcourt et al, 1998). It was also shown that prior and current
smokers are prone to develop AMD atleast 5 to 10 years before non-smokers. A risk ratio of 2 or
higher was observed for neovascular AMD (Klein et al). Stryker et al, reported that men who
smoked one pack of cigarettes per day had only 72% of the plasma α-carotene levels of nonsmokers,
even after adjusting for dietary differences between smokers and nonsmokers. The APEDS study
reported an odds ratio of 3.29 [95%CI, 1.42–7.57] for AMD in individuals with the history of cigar
smoking (Nirmalan et al, 2004).
ALCOHOL CONSUMPTION
Alcohol intake causes tissue damage by increasing the oxidative stress or affecting mechanisms that
protect against oxidative damage to retina. The inconsistent findings among various studies, however,
suggest that consumption of alcoholic beverages is not likely to be an important risk factor for the
incidence of AMD at this point of time (Eye Disease Case–Control Study Group; 1992).
DIET
Diet has been related to several chronic conditions including cancer, coronary heart disease, and
cataract. It may also have an important role in preventing and slowing the development of AMD
(Seddon et al, 1994).
LIGHT EXPOSURE
There have been conflicting reports on association of ultraviolet or visible light with AMD.
Blumenkranz and associates found a statistically significant difference in the incidence of AMD
between sun-exposed and sun-protected skin, whereas the Eye Disease Case-Control Study Group
did not find sunlight exposure a risk factor in AMD. Majority of studies however, found no
associations between estimated ambient UV-B exposure/sunlight exposure and age-related
maculopathy (Stryker et al, 1988). 3
Textbook of Vitreoretinal Diseases and Surgery
HYPERTENSION
Systemic hypertension was found to be associated with neovascular AMD. Hyman et al, conducted
a case-control study of risk factors for neovascular and non-neovascular age-related macular
degeneration (AMD). It was observed that neovascular AMD was positively associated with diastolic
blood pressure greater than 95 mm Hg (odds ratio [OR] = 4.4), as opposed to non-neovascular AMD
(Khan et al, 2006). The association of hypertension with advanced AMD was also reported by AREDS
study [OR = 1.45] (AREDS, 2005).
CATARACT SURGERY
There are few studies, which reported cataract surgery as a risk factor for AMD. Wang et al, studied
the association of cataract surgery and 5-year incidence of late-stage age-related maculopathy in the
patients from the Beaver Dam and Blue Mountains eye studies. It was found that over a period of
five years either neovascular AMD or geographic atrophy developed in 6.0 to 7.5% of aphakic eyes
(10 of 168 right and 11 of 147 left eyes), compared with 0.7% of phakic eyes (40 of 5504 right and
37 of 5572 left eyes) (Abecasis et al, 2004). According to APEDS, prior cataract surgery was significantly
associated with increased incidence of AMD (adjusted OR 3.79; 95% CI, 2.1– 6.78) (Nirmalan et al,
2004).
Genetics of AMD
The genetic basis of AMD was relatively ignored for many years as other causes for the disease
were explored. Genetic epidemiological studies have revealed that genetic differences between
populations might play an important role in explaining the prevalence among diverse ethnic groups.
Higher concordance among the monozygotic twins, familial aggregation and segregation analyses
have suggested a strong genetic basis for the disease (Seddon et al, 1997, 2005; Klaver et al, 1998).
TWIN ANALYSIS
Twin studies allows the exact comparison of the relative contribution by genetic as well as non-
genetic factors on a trait. Such studies provide an exact quantification of the contribution of genetic
factors or heritability which is the proportion of variance of a trait that is attributable to genetic
factors. Such studies have provided more direct evidence of AMD heritability by comparing disease
concordance rates in monozygotic (MZ) vs. dizygotic (DZ) twins. In a large USA population based
survey of 840 elderly male twins, including 210 MZ twin pairs, 181 DZ twin pairs, and 58 singletons
268 twins were having signs of maculopathy, of which 106 had advanced disease. Analyses of this
4 twin data yielded heritability estimates of 0.67 for intermediate and advanced disease (grades 3, 4,
Genetics of Age-related Macular Degeneration
and 5), and 0.71 for advanced disease only, including geographic atrophy and exudative AMD
(grades 4 and 5). Hammond and colleagues evaluated concordance rates for early ARM in a
cohort of 506 female volunteer twin pairs (226 MZ twin pairs and 280 DZ twin pairs). ARM was
defined as the presence of soft drusen 0.63 mm in diameter in the macular area or the presence of
pigmentary changes along with any type of drusen. The concordance for ARM in MZ twins was 0.37
compared with 0.19 in DZ twins, and the heritability of ARM was estimated to be 45% (95% CI 0.35
to 0.53).
1991). BF is an important activator of the alternative complement pathway and thus may result in
AMD by abnormal BF activity.
Recently two studies reported significant associations between AMD and variants in the
complement component 3 (C3) gene on chromosome 19p13. C3 plays an important role in activation
of both the classical and the alternative complement pathways and the plasma complement C3a
levels are significantly elevated in AMD cases compared to controls. A fourth recent study also
found ARM associated variants in the C7 and MBL2 (Gene ID 4153) complement pathway genes by
complement pathway focused analysis of an earlier genome-wide association scan.
dry AMD (Cameron et al, 2007) eyes and in promoter-based assays, this finding was not replicated
in another study (Kanda et al, 2007). Thus, the precise role of the LOC387715 and HTRA1 SNPs in
AMD is as yet unknown, and their interactions with different factors in the complement pathway
remain speculative. However, such speculations cannot be addressed unless extensive data on gene–
gene interactions in the background of other nongenetic factors leading to the pathophysiology of
AMD are elucidated (Swaroop et al, 2007)
APOE
In the past few years, association of variants in genes involved in lipid metabolism such as
Apolipoprotein E (APOE) have increased our understanding of the underlying mechanism of AMD
development. Based on common pathogenic features including lipid deposition (drusen and plaque
formation), thickening of connective tissue (Bruch’s membrane and arterial inner lining) and elevated
levels of CRP in serum, a common mechanism for the development of AMD and atherosclerotic
cardiovascular diseases was proposed (Freidman 2000). However, it was noted that the effect of
APOE alleles on AMD risk was contrastingly different than that of atherosclerosis and cardiovascular
diseases (Baird et al, 2004; Zareparsi et al, 2004). Association of APOE in AMD has been reported by
several groups (Klaver et al, 1998; Schmidt et al, 2002; Baird et al, 2004; Zareparsi et al, 2004) showing
a reduced risk of AMD with APOE-e2 allele and higher risk with allele APOE-e4. However, APOE
polymorphisms have exhibited varied geographical and ethnic variations across AMD patients
worldwide (Kaur et al, 2006).
shown to induce choroidal neovascualrization (CNV) in retinal tissues. In-vitro treatment of human
RPE cells with CEP dipeptide or CEP-HSA did not induce increased VEGF secretion and suggesting
that anti-CEP therapeutic modalities might be of value in limiting CNV in AMD (Ebrahem et al,
2006). CNV in retinal tissues can also be stimulated by the complement components (C3a and C5a)
present in the drusen. It was shown that genetic ablation of receptors for C3a or C5a reduces VEGF
expression, leukocyte recruitment and CNV formation after laser injury. These experiments suggested
that antibody-mediated neutralization of C3a or C5a or pharmacological blockade of their receptor
could be therapeutic modalities for AMD (Nozaki et al, 2005).
Conclusions
Genetic association studies have led to the implication of several candidate genes in AMD in the last
few years. These association studies have been meaningful as they have been widely replicated
across multiple populations with varied ethnic backgrounds in clinically well characterized cohorts
(Todd 2006). Moreover, the effect sizes of these variants, particularly the CFH (Y402H), LOC387715
(A69S) and HTRA1 have been substantially large that permitted a proper association amidst varied
sample sizes in different studies. In future, more such studies are required across wider geographical
regions to identify candidates that contribute to the AMD pathogenesis. Genetic typing of AMD
patients would also permit clinicians to develop correlations with genotypes for estimating disease
risk and progression. Identification of susceptible gene variant(s) would allow early intervention in
subjects ‘at-risk’ of developing AMD for a better prognosis. While the underlying biological functions
of these candidate genes and their interactions are yet to be characterized, large multicentre studies
with these variants should be undertaken with respect to the treatment modalities in order to
understand the therapeutic mechanisms in subjects carrying the risk genotype(s).
Bibliography
1. Age-related Eye Disease Study research Group. Risk factors associated with age-related macular degeneration. A
case-control study in the Age-related Eye Disease Study; Age-related Eye Disease Study Report Number 3.
Ophthalmology 2000;107: 2224-32.
2. Age-related Eye Disease Study research Group. Risk factors for the incidence of advanced Age-Related
macular degeneration in the Age-Related Eye Disease Study (AREDS) AREDS Report No. 19. Ophthalmology
2005;11: 532-9.
3. Akimoto A, Akimoto M, Kuroiwa S, Kikuchi T, Yoshimura N. Lack of association of mutations of the bestrophin
gene with age-related macular degeneration in non-familial Japanese patients. Graefes Arch Clin Exp Ophthalmol
2001;239: 66–8.
4. Allikmets R, Seddon JM, Bernstein PS, Hutchinson A, Atkinson A, et al. Evaluation of the Best disease gene in
patients with age-related macular degeneration and other maculopathies. Hum Genet 1999;104:449–53.
5. Allikmets R, Shroyer NF, Singh N, Seddon JM, Lewis RA, et al. Mutation of the Stargardt disease gene (ABCR) in
age-related macular degeneration. Science 1997;277:1805–7.
6. Ayyagari R, Zhang K, Hutchinson A, Yu Z, Swaroop A, et al. Evaluation of the ELOVL4 gene in patients with age-
related macular degeneration. Ophthalmic Genet 2001;22: 233–9.
7. Baird PN, Guida E, Chu DT, Vu HT, Guymer RH. The epsilon 2 and epsilon 4 alleles of the apolipoprotein gene are
associated with age-related macular degeneration. Invest Ophthalmol Vis Sci 2004;45:1311-5.
8. Baird PN, Islam FM, Richardson AJ, Cain M, Hunt N, et al. Analysis of the Y402H variant of the complement factor-
H gene in age-related macular degeneration. Invest Ophthalmol Vis Sci 2006;47: 4194-8.
9. Balasubramanian D. Molecular and cellular approaches to understand and treat some diseases of the eye. Curr Sci
10 2002;82: 948-57.
Genetics of Age-related Macular Degeneration
10. Blander JM and Medzhitov R. Regulation of phagosome maturation by signals from toll-like receptors. Science
2004;304, 1014-18.
11. Bressler NM. Early detection and treatment of neovascular age-related macular degeneration. J Am Board Fam
Pract 2002;15:142-52.
12. Burchard EG, Ziv E, Coyle N, Gomez SL, Tang H, et al. The importance of race and ethnic background in biomedical
research and clinical practice. N Engl J Med 2003;348: 1170-5.
13. Cameron DJ, Yang Z, Gibbs D, Chen H, Kaminoh Y, et al. HTRA1 variant confers similar risks to geographic
atrophy and neovascular age-related macular degeneration. Cell Cycle 2007;6:1122-5.
14. Castrillo A, Joseph SB, Vaidya SA, Haberland M, Fogelman AM, et al. Crosstalk between LXR and toll-like receptor
signaling mediates bacterial and viral antagonism of cholesterol metabolism. Mol Cell 2003;12: 805-16.
15. Clark SJ, Higman VA, Mulloy B, Perkins SJ, Lea SM, et al. His-384 allotypic variant of factor H associated with age-
related macular degeneration has different heparin binding properties from the non-disease-associated form. J
Biol Chem 2006;281:24713-20.
16. Conley YP, Jakobsdottir J, Mah T, Weeks DE, Klein R, et al. CFH, ELOVL4, PLEKHA1 and LOC387715 genes and
susceptibility to age-related maculopathy: AREDS and CHS cohorts and meta-analyses. Hum Mol Genet 2006;15:
3206-18.
17. Cook DN, Pisetsky DS, Schwartz DA. Toll-like receptors in the pathogenesis of human disease. Nat Immunol
2004;5: 975-9.
18. Dandona L, Dandona R, Srinivas M, Giridhar P, Vilas K, et al. Blindness in the Indian State of Andhra Pradesh.
Invest Ophthalmol Vis Sci 2001;42: 908-16.
19. De La Paz MA, Pericak-Vance MA, Lennon F, Haines JL, Seddon JM. Exclusion of TIMP3 as a candidate locus in age-
related macular degeneration. Invest Ophthalmol Vis Sci 1997;38: 1060–5.
20. DeWan A, Liu M, Hartman S, Zhang S, Liu DTL, et al. HTRA1 promoter polymorphism in wet age-related macular
degeneration. Science 2006;314: 989-92.
21. Ebrahem Q, Renganathan K, Sears J, Vasanji A, Gu X, et al. Carboxyethylpyrrole oxidative protein modifications
stimulate neovascularization: Implications for age-related macular degeneration. Proc Natl Acad Sci USA
2006;103:13480-4.
22. Edwards AO, Ritter R III, Abel KJ, Manning A, Panhuysen C, et al. Complement factor H polymorphism and age-
related macular degeneration. Science 2005;308: 421-4.
23. Fisher SA, Rivera A, Fritsche LG, Babadjanova G, Petrov S, et al. Assessment of the contribution of CFH and
chromosome 10q26 AMD susceptibility loci in a Russian population isolate. Br J Ophthalmol 2006;91: 576-8.
24. Friedman E. The role of the atherosclerotic process in the pathogenesis of age-related macular degeneration. Am
J Ophthalmol 2000;130: 658-63.
25. Gold B, Merriam JE, Zernanat J, Hancox LS, Taiber AJ, et al. Variation in factor B (BF) and complement component
2 (C2) genes is associated with age-related macular degeneration. Nat Genet 2006;38: 458-62.
26. Gotoh N, Yamada R, Hiratani H, Renault V, Kuroiwa S, et al. No association between complement factor H gene
polymorphism and exudative age-related macular degeneration in Japanese. Hum Genet 2006;120:139-43.
27. Gu X, Meer SG, Miyagi M, Rayborn ME, Hollyfield JG, et al. Carboxyethylpyrrole protein adducts and autoantibodies,
biomarkers for age-related macular degeneration. J Biol Chem 2003;278: 42027-35.
28. Haddad S, Chen AC, Santangelo SL, Seddon JM. The Genetics of Age-Related Macular Degeneration: A Review of
Progress to Date. Surv Ophthalmol 2006;51:316-3.
29. Hageman GS, Anderson DH, Johnson LV, Hancox LS, Taiber AJ, et al. A common haplotype in the complement
regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proc Natl
Acad Sci, USA 2005;102:7227-32.
30. Haines JL, Hauser MA, Schmidt S, Scott WK, Olson LM, et al. Complement factor H variant increases the risk of
age-related macular degeneration. Science 2005;308: 419-21.
31. Haines JL, Schnetz-Boutaud N, Schmidt S, Scott WK, Agarwal A, et al. Functional candidate genes in age-related
macular degeneration: Significant association with VEGF, VLDLR and LRP6. Invest Ophthalmol Vis Sci 2006;47:
329-35.
32. Jakobsdottir J, Conley YP, Weeks DE, Mah TS, Ferrell RE, et al. Susceptibility genes for age-related maculopathy on
chromosome 10q26. Am J Hum Genet 2005;77: 389-407.
33. Johnson PT, Betts KE, Radeke MJ, Hageman GS, Anderson GH, et al. Individuals homozygous for the age-related
macular degeneration risk-conferring variant of complement factor H have elevated levels of CRP in the choroids.
Proc Natl Acad Sci USA 2006;103: 17456-61.
34. Kaur K, Hussain A, Hussain N, Das T, Pathangay A, et al. Analysis of CFH, TLR4 and APOE polymorphism in India
suggests the Tyr402His variant of CFH to be a global marker for age-related macular degeneration. Invest
Ophthalmol Vis Sci 2006;47: 3729-35.
11
Textbook of Vitreoretinal Diseases and Surgery
35. Kiechl S, Lorenz E, Reindl M, Wiedermann CJ, Oberhollenzer F, et al. Toll-like receptor 4 polymorphisms and
atherogenesis. N Engl J Med 2002;347:185-92.
36. Kindzelskii AL, Elner VM, Elner SG, Yang D, Hughes BA, et al. Toll-Like Receptor 4 (TLR4) of Retinal Pigment
Epithelial Cells Participates in Transmembrane Signaling in Response to Photoreceptor Outer Segments. J Gen
Physiol 2004;124: 139-49.
37. Klaver CC, Kliffen M, van Duijn CM, Hofman A, Cruts M, et al. Genetic association of apolipoprotein E with age-
related macular degeneration. Am J Hum Genet 1998;63: 200-206. Erratum in Am J Hum Genet 1998; 63:1252.
38. Klaver CCW, Wolf RCW, Assink JJM, van DuJin CM, Hofman A, et al. Genetic risk of age-related maculopathy.
Population-Based Familial Aggregation Study. Arch Ophthalmol 1998;116: 646-51.
39. Klein ML, Schultz DW, Edwards A, Matise TC, Rust K, et al. Age-related macular degeneration: clinical features in
a large family and linkage to chromosome 1q. Arch Ophthalmol 1998;116: 1082-88.
40. Klein R, Klein BE, Linton KL. Prevalence of age-related maculopathy. The Beaver Dam Eye Study.
Ophthalmology1992;99: 933-43.
41. Klein R, Klein BE, Franke T. The relationship of cardiovascular disease and its risk factors to age-related maculopathy:
The Beaver Dam Eye study. Ophthalmology 1993;100: 406-14.
42. Klein R, Peto T, Bird A, Vannewkirk MR. The epidemiology of age related macular degeneration. Am J Ophthalmol
2004;1376: 486-95.
43. Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, et al. Complement factor H polymorphism in age-related macular
degeneration. Science 2005;308: 385-9.
44. Krishnaiah S, Das TP, Nirmalan PK, Nutheti R, Shamanna BR, et al. Risk factors for age-related macular degeneration:
Findings from Andhra Pradesh Eye Disease Study in south India. Invest Ophthalmol Vis Sci 2005;6: 4442-9.
45. Lokki ML, Koskimies SA. Allelic differences in hemolytic activity and protein concentration of BF molecules are
found in association with particular HLA haplotypes. Immunogenetics 1991;34: 242-6.
46. Magnusson KP, Duan S, Sigurdsson H, Petursson H, Yang Z, et al. CFH Y402H confers similar risk of soft drusen
and both forms of advanced AMD. PLoS Medicine 2006;3:1-5.
47. Majewski J, Schultz DW, Weleber RG, Schain MB, Edwards AO, et al. Age-related macular degeneration-a genome
scan in extended families. Am J Hum Genet 2003;73:540-50.
48. Mitchell P, Smith W, Attebo K, Wang JJ. Prevalence of age-related maculopathy in Australia. The Blue Mountains
Eye Study. Ophthalmology 1995;102:1450-60.
49. Nirmalam PK, Katz J, Robin AL, Tielsch JM, Namperumalsamy P, et al. Prevalence of vitreoretinal disorders in a
rural population of Southern India: The Aravind Comprehensive Eye Survey. Arch Ophthalmol 2004;122: 581-6.
50. Nozaki M, Raisler BJ, Sakurai E, Sarma JV, Barnum SR, et al. Drusen complement components C3a and C5a
promote choroidal neovascularization. Proc Natl Acad Sci, USA 2006;103: 2328-33.
51. Okamoto H, Umeda S, Obazawa M, Minami M, Noda T, et al. Complement factor H polymorphisms in Japanese
population with age-related macular degeneration. Mol Vis 2006;12:156-8.
52. Rivera A, Fisher SA, Fritsche LG, Keilhauer CN, Lichtner P, et al. Hypothetical LOC387715 is a second major
susceptibility gene for age related macular degeneration contributing independently from complement factor H to
disease risk. Hum Mol Genet 2005;14: 3227-36.
53. Ross RJ, Bojanowski CM, Wang JJ, Chew EW, Rochtchina E, et al. The LOC387715 and age related macular
degeneration: Replication in three case-control samples. Invest Ophthalmol Vis Sci 2007;48:1128-32.
54. Schmidt S, Klaver C, Saunders A, Postel E, De La Paz M, et al. A pooled case-control study of the apolipoprotein E
(APOE) gene in age-related maculopathy. Ophthalmic Genet 2002;23:209-23.
55. Schmidt S, Saunders AM, De La Paz MA, Postel EA, Heinis RM, et al. Association of the Apolipoprotein E gene with
the age-related macular degeneration: Possible effect modification by family history, age, and gender. Mol Vis
2000;6: 287-93.
56. Schmidt S, Scott WK, Postel EA, Agarwal A, Hauser ER, et al. Ordered subset linkage analysis supports a susceptibility
locus for age-related macular degeneration on chromosome 16p12. BMC Genet 2004;5:18.
57. Schmidt S, Hauser MA, Scott WK, Postel EA, Aggarwal A, et al. Cigarette smoking strongly modifies the association
of LOC387715 and age-related macular degeneration. Am J Hum Genet 2006;78: 852-64.
58. Seddon JM, Ajani UA, Mitchell BD. Familial aggregation of age-related maculopathy. Am J Ophthalmol 1997;123:
199-206.
59. Seddon JM, Ajani UA, Sperduto RD, Hiller R, Blair N, et al. Dietary carotenoids, vitamins A, C, and E, and advanced
age-related macular degeneration. Eye Disease Case-Control Study Group. JAMA 1994;272: 1413-20. Erratum in:
JAMA 1995; 273: 622.
60. Seddon JM, Cote J, Page WF, Aggen SH, Neale MC. The US twin study of age related macular degeneration:
Relative roles of genetic and environmental influences. Arch Ophthalmol 2005;123: 321-7.
61. Seddon JM, Santangelo SL, Book K, Chong S, Cote J. A genome-wide scan for age-related macular degeneration
12 provides evidence for linkage to several chromosomal regions. Am J Hum Genet 2003;73: 780-90.
Genetics of Age-related Macular Degeneration
62. Seddon JA, Francis PJ, George S, Schultz DW, Rosner B, et al. Association of CFH Y402H and LOC387715 A69S with
progression of age-related macular degeneration. JAMA 2007; 297:1793-1800.
63. Seitsonen S, Lemmela S, Holopainen J, Tommila P, Ranta P, et al. Analysis of variants in the complement factor H,
the elongation of very long chain fatty acids-like 4 and the hemicentin 1 genes of age-related macular degeneration
in the Finnish population. Mol Vis 2006;12: 796-801.
64. Sepp T, Khan JC, Thurlby DA, Shahid H, Clayton DG, et al. Complement Factor H variant Y402H is a major risk
determinant for geographic atrophy and choroidal neovascularization in smokers and nonsmokers. Invest
Ophthalmol Vis Sci 2006;47: 536-40.
65. Shastry BS, Trese MT. Evaluation of the peripherin/RDS gene as a candidate gene in families with age-related
macular degeneration. Ophthalmologica 1999;213:165–70.
66. Simonelli F, Frisso G, Testa F, di Fiore R, Vitale DF, et al. Polymorphsim p.402Y>H in the complement factor
H protein is a risk factor for age-related macular degeneration in an Italian population. Br J Ophthalmol 2006;90:1142-
5.
67. Souied EH, Leveziel N, Richard F, Dragon-Durey MA, Coscas G, et al. Y402H Complement factor H polymorphism
associated with exudative age-related macular degeneration in the French population. Mol Vis 2005;11:1135-40.
68. Stone EM, Lotery AJ, Munier FL, Heon E, Piguet B, et al. A single EFEMP1 mutation associated with both Malattia
Leventinese and Doyne honeycomb retinal dystrophy. Nat Genet 1999;22:199-202.
69. Tanimoto S, Tamura H, Ue T, Yamane K, Maruyama H, et al. A polymorphism of LOC387715 gene is associated
with age-related macular degeneration in the Japanese population. Neurosci Lett 2006;414: 71-4.
70. The Eye Disease Case – Control Study Group. Risk factors for neovascular age-related macular degeneration. Arch
Ophthalmol 1992;110:1701-8.
71. Todd JA. Statistical false positive or true disease pathway? Nat Genet 2006;38: 731-3.
72. Tomany SC, Cruickshanks KJ, Klein R, Klein BE, Knudtson MD. Sunlight and the 10-year incidence of age-related
maculopathy: the Beaver Dam Eye Study. Arch Ophthalmol 2004;122: 750-7. Erratum in: Arch Ophthalmol. 2005;
123: 362.
73. Tomany SC, Wang JJ, Van Leeuwen R, Klein R, Mitchell P, et al. Risk factors for incident age-related macular
degeneration: Pooled findings from three continents. Ophthalmology 2004;111:1280-7.
74. Weeks DE, Conley YP, Mah TS, Paul TO, Morse L, et al. A full genome scan for age-related maculopathy. Hum Mol
Genet 2000;9:1329-49.
75. Weeks DE, Conley YP, Tsai HJ, Mah TS, Schmidt S, et al. Age-related maculopathy: a genome-wide scan with
continued evidence of susceptibility loci within the 1q31, 10q26, and 17q25 regions. Am J Hum Genet 2004;75:
174-89.
76. World Health Organization. Fact sheet no. 282. Magnitude and causes of visual impairment. http://www.who.int/
mediacentre/factsheets/fs282/, 2004.
77. Yang Z, Camp NJ, Sun H, Tong Z, Gibbs D, et al. A variant of the HTRA1 gene increases susceptibility to age-
related macular degeneration. Science 2006;314: 992-3.
78. Yoshida T, DeWan A, Zhang H, Sakamoto R, Okamoto H, et al. HTRA1 promoter polymorphism predisposes
Japanese to age-related macular degeneration. Mol Vis 2007;13: 545-8.
79. Zareparsi S, Branham KE, Li M, Shah S, Klein RJ, et al. Strong association of the Y402H variant in complement factor
H at 1q32 with susceptibility to age-related macular degeneration. Am J Hum Genet 2005;77:149-53.
80. Zareparsi S, Buraczynska M, Branham KE, Shah S, Eng D, et al. Toll-like receptor 4 variant D299G is associated with
susceptibility to age-related macular degeneration. Hum Mol Genet 2005;14:1449-55.
81. Zareparsi S, Reddick AC, Branham KE, Moore KB, Jessup L, et al. Association of apolipoprotein E alleles with
susceptibility to age-related macular degeneration in a large cohort from a single center. Invest Ophthalmol Vis Sci
2004;45: 1306-10.
13
Textbook of Vitreoretinal Diseases and Surgery
Introduction
Cell therapy is an exciting area of research that is aimed at regeneration of tissues or organs in
patients with irreparable damage to these tissues or organs. Cell therapy is no longer restricted to
transfer of cells from a healthy subject to a patient, as in bone marrow transplant or corneal transplant.
We can now envisage cell therapy that involves generation of patient-specific cells for transplantation,
manipulate them in vitro into the desired cell type and transplant these cells into a patient for functional
restoration of the damaged tissue. Although many cell therapies are currently being tested in animal
models and their clinical application might take much longer, results from these studies are encouraging
and suggest that regeneration of custom-made organs might indeed be possible in real life.
Cell therapy in the form of simple blood transfusion and bone marrow transplant has been in use
for a long time. This was limited to collecting healthy cells from matched donors and introducing
these cells into the patient. The cells were not propagated or manipulated in any way in vitro prior to
transplantation. The first culture and transplantation of dermal epithelial cells was performed to
replace damaged skin in burns patients1, 2 and taking cues from these experiments, similar approach
was used to regenerate corneal epithelium in patients with limbal stem cell deficiency.3 Simultaneously
embryonic stem cells were first established in 19814,5 and further understanding of mechanisms
underlying self-renewal and differentiation of stem cells into various types of cells led to designing
of new therapeutic strategies.
Degenerative diseases of retina like retinitis pigmentosa (RP), age-related macular degeneration
(AMD) and diabetic retinopathy lead to irreparable damage to the retina and are a major cause of
blindness in developed countries (Figure 2-1). Several genetic disorders such as retinitis pigmentosa
or Leber congenital amaurosis also lead to retinal degeneration and blindness. A therapeutic approach
that can regenerate retinal cells or the entire retina would help improve visual acuity and quality of
life of these patients. Use of cell therapy for retina however is not easy as the retina consists of
several cell types and retinal disorders can occur due to damage to any of these cell types. The
neural retina consists of rod and cone photoreceptors, bipolar cells, ganglion cells, horizontal cells,
Müller cells and amacrine cells that are arranged in a definite three-dimensional structure. The
neural retina rests on the retinal pigment epithelium (RPE), which is in close contact with
photoreceptors and is vital for maintenance of photoreceptor homeostasis. Degeneration of any
type of cells in the neural retina or RPE can lead to retinal degeneration and loss of vision. Retinal
disorders could also occur due to uncontrolled growth of endothelial cells leading to
neovascularization in the retina that can eventually lead to retinal detachment in diseases like diabetic
retinopathy and AMD. The current attempts at retinal cell therapy are therefore aimed at regeneration
of cells in neural retina, regeneration of RPE and re-establishment of correct retinal vasculature. For
treatment of inherited retinal disorders, the cell therapy also might involve manipulations to correct
the genetic defect by gene therapy.
Cell therapy is an interdisciplinary field that requires inputs from the field of stem cell biology,
regenerative biology, material science, developmental biology, genetic engineering and clinicians.
The success and feasibility of any cell therapy depends on a number of factors, such as availability of
suitable source of stem cells, expansion and manipulation of cells to generate the desired type of cell,
introduction of cells in vivo, survival and integration of cells to reconstruct degenerated tissue and
restoration of its physiological function (Figure 2-2).
An appropriate source of stem cells that can generate and sustain cells/tissue of interest is an
16 important starting point for cell therapy. Stem cells are the cells that have the ability to renew
Cell Therapy in Retinal Diseases
FIGURE 2-1: Fundus photograph showing wet (Top left) and geographic atrophy (Top right) age-related macular
degeneration, and proliferative diabetic retinopathy (bottom)
themselves and also differentiate into any other cell of the body. They could be embryonic stem cells,
which are derived from the inner cell mass of blastula or adult stem cells, which are present in the adult
tissues such as bone marrow, liver, muscles, skin and cornea. Embryonic stem cells are totipotent, i.e.
they have the ability to differentiate into any other cell type. Adult stem cells on the other hand have
limited capacity to divide and can differentiate only into specific types of cells. They can be either
multipotent stem cells that can differentiate into many types of cells, such as the bone marrow stem
cells, or unipotent stem cells that can differentiate into only one type of cell, e.g. limbal stem cells. For
retina, several sources of stem cells such as embryonic, fetal and adult stem cells from eye and bone
marrow have been investigated as discussed below. An autologous source of cells would be preferred
in order to avoid complications of rejection associated with graft rejection, but it may not be possible
to use an autologous source if retinal degeneration is due to an inherited disorder.
The next important issue is the ability to culture these stem cells in vitro to expand them. The
number of cells available for transplantation, especially if they are from adult tissues, is often limited
and the cells need to be cultured in vitro to obtain sufficient number of cells. Expansion of cells while
maintaining their characteristics such as their stemness or their differentiation status is a challenge
and requires standardization of culture conditions. This involves identification of correct culture 17
Textbook of Vitreoretinal Diseases and Surgery
medium, growth factors, feeder cells and it may also involve standardization of correct matrix that
will help cellular growth and differentiation or harvesting of cell at the time of transplantation.
After expansion, stem cells might be directly introduced in the animal to allow it to differentiate
in the niche provided by the recipient or the cells might be coaxed to undergo differentiation in vitro
to generate the cell or cells of interest. Directed differentiation of a stem cell into a cell of interest is
extremely challenging and requires understanding of mechanisms underlying the development of
that particular tissue during embryogenesis. The differentiation of a stem cell into the desired cell
type is regulated by both intrinsic information present within the cell and the cues provided by the
niche it is residing in. Therefore for directed differentiation, stem cells need to be cultured under
specific conditions that would allow their differentiation into cells of interest. Extensive
experimentation to understand the developmental biology and cell differentiation of the tissue of
interest needs to be carried out using various animal models to understand the conditions required
for such in vitro manipulations. This is particularly complicated for tissues like retina, which is made
up of several cell types. For treatment of genetic disorders, the defective copy of the gene needs to
be replaced with the wild type or the correct copy of the gene at this stage.
The correct route of introduction of these differentiated cells in vivo also needs to be investigated.
In retinal cell therapy, the mode of introduction, whether subretinal or intravitreal, is important.
Once introduced in the body, the transplanted cells are monitored for their survival. The cells are
also monitored to check if they maintain their state of differentiation. If undifferentiated cells have
been introduced, whether they can differentiate in vivo into the desired cell type needs to be
monitored. The cells should also integrate well in the tissue. In the retina, it is important that
18 transfected cells establish synaptic connections for restoration of retinal function. Similarly in the
Cell Therapy in Retinal Diseases
case of RPE cells, the cells should integrate and establish contact with photoreceptors in order to
maintain their homeostasis. All these steps are important in ensuring that the transplanted cells
function well in vivo and are a true substitute for the degenerated or damaged tissue.
Last but not the least transplanted cells should not cause any undesirable changes in the recipient.
Cell therapy involves removal of stem cells from their normal niche and introduction into a new
environment. Changes in the stem cell niche could lead to unsafe situations like uncontrolled cell
division leading to tumor formation. The application of cell therapy to humans should be performed
only after ensuring that the cell therapy is free of such safety issues and it restores normal
functioning.
the superior colliculus region. A more systematic study in mouse to check cells at what stage are able
to integrate in adult retina showed that postnatal P1-P7 retinal cells which express retina-specific
transcription factor Nrl, but not embryonic cells, could integrate well in the retina.9 The integrated
cells formed functional synaptic junctions and also improved visual function as seen by pupillometry
and extracellular field potential recordings from ganglion cell layer. This study identified the specific
ontogenic stage of transplanted cells that is important for integration of retinal cells and can restore
retinal function.
Human studies have also been carried out which show quite encouraging results. Transplantation
of fetal retina in patients with retinitis pigmentosa showed that fetal retina integrates well into the
degenerating retina and can improve visual acuity.10, 11 A recent report of clinical trial from the same
group shows that transplantation of fetal retinal sheets led to improvement in visual acuity of 7 out
of 10 patients suffering from RP and AMD.12 No adverse events were reported in these studies and
the grafts survived well. This is encouraging although its application to large population could have
ethical and practical problems. However with the information available now it might be possible to
differentiate stem cells in vitro to a similar ontogenic stage before transplanting these cells into the
retina.
Studies show that retinal stem cells (RSCs) present in neonatal retinas can certainly be isolated,
propagated and differentiated in vitro to generate retinal cells. This is important, as it would be
helpful if cells from one fetal retina could be used for several transplantations. Neonatal/fetal tissues
would likely have less number of stem cells/progenitor cells and ability to increase their number
before transplantation would increase success rate of this treatment. Using epidermal growth factor
(EGF), retinal progenitors from neonatal rats could be differentiated into both early and late neuronal
cells.13 Similar studies were also performed in pigs, where progenitor cells from neural retina of pig
could be cultured in vitro and when injected subretinally in laser-injured retina, could integrate and
express photoreceptor cells markers.14 Mouse retinal progenitors from postnatal day 1, when
transplanted into hosts lacking rhodopsin, were able to develop into mature neurons, express
recoverin, rhodopsin or cone opsin and integrate into the retina.15 Interestingly these cells could also
rescue cells in the outer nuclear layer, with improvement in light-mediated behavior as compared to
uninjected animals. On the other hand, subretinal injection of radial glial cells from retina of new-
born mice into rd1 or VPP mice led differentiation of injected cells into either ganglion cells or glial
cells.16 Another study has shown that incubation of mouse radial glia cells with FGF-2 and B27 also
can lead to differentiation of these cells into photoreceptors with high yield.17 More extensive studies
need to be carried out to investigate whether these cells are functionally as competent as normal
retinal cells.
RPE Cells
RPE cells also have been investigated for their potential to generate retinal cells, as both neural
retina and RPE develop from neuroepithelium and follow similar developmental signal during initial
development. Indeed rat RPE cells from early embryonic stages have the potential to develop into
retinal neurons.18 In the presence of bFGF, RPE cells from stages E12-E14 could differentiate into
cells expressing markers of rod cells, amacrine cells and RGC cells. Although RPE cells appear to
have the potential for differentiation into retinal cells, they are difficult to harvest, this might hinder
their use for cell therapy.
On the other hand, homologous transplantation of RPE cells has shown promising results. Replacing
20 degenerating RPE cells by transplanting healthy, autologous RPE cells from peripheral retina has
Cell Therapy in Retinal Diseases
been used a therapeutic approach for treating AMD. Dissociated RPE cells19 as well as RPE explants
from the mid-periphery have been transplanted in patients with neovascular AMD20, 21 and this led
to some improvement in visual acuity. The procedure however can lead to complications such as
retinal detachment and improvement in visual acuity can be transient.22
noggin, followed by incubation with fibroblast growth factor (FGF) and epidermal growth factor
(EGF) in serum-free medium. This treatment promoted differentiation of ES cells into neural
progenitors and these neural progenitors also expressed several markers of retinal cells. In another
study human ES cells were treated with noggin, Dkk1 and insulin-like growth factor (IGF) to generate
embryoid bodies, followed by incubation with bFGF for three weeks to generate retinal progenitors.47
This treatment was more efficient and generated retinal progenitors with high efficiency, up to 80%.
The cells were also able to integrate into degenerating mouse retina. In addition both human and
mouse ES cells have been shown to differentiate into RPE and other types of neurons when cultured
on denuded, gelatin-coated human amniotic membrane.48 This method suggests an alternative to
use of xenogenic feeder cells as has been reported previously.42, 44
The studies mentioned above use fetal calf serum or fetal tissues as feeder layers to direct
differentiation of ES cells into photoreceptors. The differentiation medium used here is not defined
and would involve use of xenogenic agents and hence may not be suitable for transplantation in
human patients. Recent study by Osakada and coworkers reported the use of a completely defined
medium to generate photoreceptors and RPE cells from mouse, monkey and human ES cells.49 The
authors used a combination of purified signaling molecules in a step-wise manner and combined this
with selection of cells based on progenitor-specific markers at each step to finally generate
photoreceptors from ES cells. They first generated neural retinal precursors from mouse ES cells
using serum-free suspension culture in the presence of Wnt and nodal antagonists as described
earlier.50 From these cells, retinal progenitors were selected based on expression of retinal progenitor
marker Rx. These cells were further cultured on laminin-fibronectin coated dishes along with DAPT,
an inhibitor of notch signaling. Around 22% of the treated cells expressed Crx, photoreceptor-
specific marker and were post-mitotic. 10% of these cellls expressed cone-specific markers. A further
incubation of these cells with combination of aFGF, bFGF, taurine, sonic hedgehog and retinoic acid,
led to generation of 17% cells that expressed rod-specific markers. A similar approach was also used
for generation of human and monkey photoreceptors. RPE cells were also generated from monkey
ES cells using defined medium, only this time selection was done for expression of Mitf, marker for
RPE progenitor, instead of Rx.49 It remains to be seen whether these cells can establish connections
with neurons and whether they are functionally active.
ES cells can also provide a niche for degenerating retinal cells that can prevent further damage
under certain conditions. Mouse ES cells differentiated to neural lineages have been shown to delay
retinal degeneration in mouse models. The ES cells differentiated into neuronal cells after removal
LIF and addition of retinoic acid, these neuralised cells were introduced in mnd mice, which suffer
from lysosomal storage disorder that causes retinal degeneration.51 These cells could integrate into
the retina, reduce lysosomal storage bodies and delay photoreceptor degeneration. This suggests
that neuralised ES cells could prevent or delay disease progression if not completely rescue the
disease.
While use of ES cells for therapeutic purpose in humans is questionable at this stage, development
of two important techniques that can reprogram differentiated adult cells into stem cells have caused
excitement among scientists and such cells could be further differentiated into retinal progenitors
using the information that is already available from research on embryonic stem cells. The first
technique is that of somatic nuclear transfer, which involves transfer of nucleus from an adult cell
into an enucleated oocyte (Figure 2-3A). The combination of factors present in the oocyte can
reprogram the adult nucleus and the resulting cell acquires characteristics of a totipotent stem cell
which, when transferred into a surrogate mother, can develop into a complete animal. Dolly the
23
Textbook of Vitreoretinal Diseases and Surgery
A B
FIGURES 2-3A and B: Reprogramming of adult nucleus. A: Reprogramming by somatic cell nuclear transfer.
B: Reprogramming by introduction of four transcription factors to generate induced pluripotent cells
sheep was the first animal developed by this technique.52 The technique although promising, again
requires use of oocytes, which may not be ethically acceptable and the success rate of this technique
is low.
The second and more recent breakthrough in stem cell biology is the generation of induced
pluripotent cells (Figure 2-3B). For the first time scientists were able to completely dedifferentiate a
differentiated dermal fibroblast to generate a pluripotent stem cell by transfection of four transcription
factors; Oct3/4, Klf4, c-myc and Sox2.53 When introduced into a blastula, these cells could give rise to
all tissues of the fetus, including germ cells, indicating that reprogrammed adult cells behaved like
embryonic stem cells.54 This technique will allow generation of stem cells from patient’s own cells,
which can be further manipulated to generate cells of our interest. It also does not require use of
oocytes or embryonic tissue and the use of induced pluripotent cells should therefore be free of ethical
issues. Use of these cells will also avoid graft rejection. Such cells can be induced into photoreceptors
as described49 and then introduced into patients with retinal degeneration to prevent further damage.
At the same time such therapeutic approaches should be used with caution as ES cells have
unlimited potential to self-renewal and introducing these cells in a different environment could
trigger their division through some unidentified factor. Indeed animals generated using induced
pluripotent stem cells had a higher rate of tumor formation.54 Also neural precursors generated
from mouse ES cells led to teratoma formation in 50% mice when injected in mouse retina.55 These
health and safety concerns need to be addressed before application of stem cell therapy.
In vivo bone marrow hematopoietic stem cells can differentiate into cells of lymphoid and myeloid
origin while bone marrow mesenchymal cells form endothelial cells, chondrocytes, skeletal muscles
and osteocytes. In addition in vitro they can also be differentiated into cardiocytes, renal cells,
pancreatic cells and hepatocytes. Bone marrow cells can also differentiate into neurons in vitro56 and
as retina is also of neural lineage, the potential of bone marrow cells has been explored for retinal
cell therapy. In addition potential of bone marrow cells for revascularization of retina and for trophic
support for survival retinal cells has also been explored.
Bone marrow cells can differentiate into retinal cells both in vitro and in vivo. When chimeric mice
with hematopoietic cells expressing GFP, were subjected to laser-induced Bruch’s membrane rupture,
GFP-positive astrocytes, vascular endothelial cells and RPE cells were found in the retina.57 Systemically
injected bone marrow stromal cells also migrated to the subretinal space in rats with NaIO¯induced
3
RPE degeneration and expressed RPE65.58 These studies indicate a role for bone marrow cells in
retinal wound repair in vivo and their ability to home the site of retinal injury. CD90+ marrow
stromal cells have also been used to generate retinal progenitor cells using a combination of activin
A, taurine and EGF in vitro.59 After incubation with this cocktail, these cells expressed markers of
photoreceptors such as rhodopsin, opsin and recoverin. When injected into the subretinal space of
RCS rats, these cells integrated into the retina and formed structures similar to photoreceptor layer.
However restoration of cone and rod function by these cells need to be further examined. Bone
marrow stromal cells also acquired RPE-like characteristics when transduced with adenovirus and
co-cultured with RPE cells.60 When injected subretinally into RCS rats, these cells integrated into
host RPE and prevented further photoreceptor degeneration of RCS rats for up to two months after
injection. This effect was enhanced when the stromal cells were transduced with PEDF prior to
injection, suggesting a role for PEDF in this trophic support by stromal cells.60
Interestingly bone marrow cells can also rescue retinal degeneration by contributing to angiogenesis
in the retina. Lineage negative (Lin¯) hematopoietic cells contain a population of endothelial cells.61
Intravitreally injected Lin¯ cells incorporated into the retinal vasculature of neonatal mice and also
in the vasculature of injury-induced angiogenesis in adults.62 Importantly these cells could also rescue
retinal vascular degeneration in rd1 and rd10 mice, which are models of retinitis pigmentosa, and
which in turn could rescue photoreceptors.63 The vasculature preferentially rescued cones from
degeneration and this was accompanied by increased expression of anti-apoptotic genes. A detectable,
although abnormal, electroretinogram recording was also observed in injected mice. Retinal
vasculature thus appears to be an important regulator of retinal integrity and restoration of retinal
vasculature could be a feasible approach for treating retinal degeneration.
that the retina itself can provide the correct cues for differentiation of neuronal cells into retinal cells
till a certain stage of development. The studies carried out using NSCs so far do not provide any
evidence for functional rescue of retinal cells during retinal degeneration.
Future Perspectives
Cell therapy holds great promise as potential therapeutic approach for treatment of retinal disorders.
Studies carried out so far provide evidence that stem cells from a number of sources have the
potential to differentiate into cells with retinal phenotype. These cells when transplanted in animal
models can also survive and integrate into the retina in some cases but the evidence for restoration
of retinal function is available only for a few of these studies. This suggests that more stringent tests
investigating function of retinal cells should be performed in addition to investigating the expression
of various retinal markers. Exactly which characteristics of transplanted cells and recipient’s retina
are important for functional integration is not clear yet. But fetal and neonatal retinas probably hold
the answers to these questions. Fetal/neonatal retinas provide both progenitor cells that can restore
some visual function when transplanted into a degenerating retina and a niche that can differentiate
stem cells into retinal cells. The challenge ahead of us is to generate retinal cells, which are at the
appropriate stage of differentiation so that they are capable of restoring retinal function. At the
same time caution should be exercised to ensure the safety of these cell therapies and to avoid
undesirable outcomes of cell therapy. With the advances in stem cell biology, it is also now possible
to generate patient-specific pluripotent stem cells that are capable of generating all types of cells, if
the correct milieu is provided. It remains to be seen whether these cells can be coaxed in vitro to
differentiate into retinal cells that can integrate and restore retinal function.
References
1. Green H, Kehinde O, Thomas J. Growth of cultured human epidermal cells into multiple epithelia suitable for
grafting. Proc Natl Acad Sci USA 1979;76 (11): 5665-8.
2. Gallico GG, 3rd, O’Connor NE, Compton CC, Kehinde O, Green H. Permanent coverage of large burn wounds
with autologous cultured human epithelium. N Engl J Med 1984;311 (7): 448-51.
3. Pellegrini G, Traverso CE, Franzi AT, Zingirian M, Cancedda R, De Luca M. Long-term restoration of damaged
corneal surfaces with autologous cultivated corneal epithelium. Lancet 1997; 349 (9057): 990-3.
4. Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature 1981; 292
(5819):154-6.
5. Martin GR. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by
teratocarcinoma stem cells. Proc Natl Acad Sci USA 1981; 78 (12): 7634-8.
6. Aramant RB, Seiler MJ, Ball SL. Successful cotransplantation of intact sheets of fetal retina with retinal pigment
26 epithelium. Invest Ophthalmol Vis Sci 1999; 40 (7):1557-64.
Cell Therapy in Retinal Diseases
7. Van Hoffelen SJ, Young MJ, Shatos MA, Sakaguchi DS. Incorporation of murine brain progenitor cells into the
developing mammalian retina. Invest Ophthalmol Vis Sci 2003; 44 (1): 426-34.
8. Sagdullaev BT, Aramant RB, Seiler MJ, Woch G, McCall MA. Retinal transplantation-induced recovery of retinotectal
visual function in a rodent model of retinitis pigmentosa. Invest Ophthalmol Vis Sci 2003; 44 (4):1686-95.
9. MacLaren RE, Pearson RA, MacNeil A, et al. Retinal repair by transplantation of photoreceptor precursors. Nature
2006; 444 (7116): 203-7.
10. Radtke ND, Seiler MJ, Aramant RB, Petry HM, Pidwell DJ. Transplantation of intact sheets of fetal neural retina
with its retinal pigment epithelium in retinitis pigmentosa patients. Am J Ophthalmol 2002; 133 (4): 544-50.
11. Radtke ND, Aramant RB, Seiler MJ, Petry HM, Pidwell D. Vision change after sheet transplant of fetal retina with
retinal pigment epithelium to a patient with retinitis pigmentosa. Arch Ophthalmol 2004; 122 (8): 1159-65.
12. Radtke ND, Aramant RB, Petry HM, Green PT, Pidwell DJ, Seiler MJ. Vision Improvement in Retinal Degeneration
Patients by Implantation of Retina Together with Retinal Pigment Epithelium. Am J Ophthalmol 2008.
13. Angenieux B, Schorderet DF, Arsenijevic Y. Epidermal growth factor is a neuronal differentiation factor for retinal
stem cells in vitro. Stem Cells 2006; 24 (3): 696-706.
14. Klassen H, Kiilgaard JF, Zahir T, et al. Progenitor cells from the porcine neural retina express photoreceptor
markers after transplantation to the subretinal space of allorecipients. Stem Cells 2007; 25 (5): 1222-30.
15. Klassen HJ, Ng TF, Kurimoto Y, et al. Multipotent retinal progenitors express developmental markers, differentiate
into retinal neurons, and preserve light-mediated behavior. Invest Ophthalmol Vis Sci 2004; 45 (11): 4167-73.
16. Canola K, Angenieux B, Tekaya M, et al. Retinal stem cells transplanted into models of late stages of retinitis
pigmentosa preferentially adopt a glial or a retinal ganglion cell fate. Invest Ophthalmol Vis Sci 2007; 48 (1): 446-54.
17. Merhi-Soussi F, Angenieux B, Canola K, et al. High yield of cells committed to the photoreceptor fate from
expanded mouse retinal stem cells. Stem Cells 2006; 24 (9): 2060-70.
18. Zhao S, Thornquist SC, Barnstable CJ. In vitro transdifferentiation of embryonic rat retinal pigment epithelium to
neural retina. Brain Res 1995; 677 (2): 300-10.
19. Binder S, Krebs I, Hilgers RD, et al. Outcome of transplantation of autologous retinal pigment epithelium in age-
related macular degeneration: a prospective trial. Invest Ophthalmol Vis Sci 2004; 45 (11): 4151-60.
20. Joussen AM, Heussen FM, Joeres S, et al. Autologous translocation of the choroid and retinal pigment epithelium
in age-related macular degeneration. Am J Ophthalmol 2006; 142 (1): 17-30.
21. MacLaren RE, Uppal GS, Balaggan KS, et al. Autologous transplantation of the retinal pigment epithelium and
choroid in the treatment of neovascular age-related macular degeneration. Ophthalmology 2007; 114 (3): 561-70.
22. MacLaren RE, Bird AC, Sathia PJ, Aylward GW. Long-term results of submacular surgery combined with macular
translocation of the retinal pigment epithelium in neovascular age-related macular degeneration. Ophthalmology
2005; 112 (12): 2081-7.
23. Perron M, Harris WA. Retinal stem cells in vertebrates. Bioessays 2000; 22 (8): 685-8.
24. Tropepe V, Coles BL, Chiasson BJ, et al. Retinal stem cells in the adult mammalian eye. Science 2000; 287 (5460):
2032-6.
25. Ahmad I, Tang L, Pham H. Identification of neural progenitors in the adult mammalian eye. Biochem Biophys Res
Commun 2000; 270 (2): 517-21.
26. MacNeil A, Pearson RA, MacLaren RE, Smith AJ, Sowden JC, Ali RR. Comparative analysis of progenitor cells
isolated from the iris, pars plana, and ciliary body of the adult porcine eye. Stem Cells 2007; 25 (10): 2430-8.
27. Das AV, James J, Rahnenfuhrer J, et al. Retinal properties and potential of the adult mammalian ciliary epithelium
stem cells. Vision Res 2005; 45 (13):1653-66.
28. Coles BL, Angenieux B, Inoue T, et al. Facile isolation and the characterization of human retinal stem cells. Proc Natl
Acad Sci USA 2004;101 (44): 15772-7.
29. Nishiguchi KM, Kaneko H, Nakamura M, Kachi S, Terasaki H. Identification of photoreceptor precursors in the
pars plana during ocular development and after retinal injury. Invest Ophthalmol Vis Sci 2008; 49 (1): 422-8.
30. Nickerson PE, Emsley JG, Myers T, Clarke DB. Proliferation and expression of progenitor and mature retinal
phenotypes in the adult mammalian ciliary body after retinal ganglion cell injury. Invest Ophthalmol Vis Sci 2007;
48 (11): 5266-75.
31. Jomary C, Jones SE. Induction of functional photoreceptor phenotype by exogenous Crx expression in mouse
retinal stem cells. Invest Ophthalmol Vis Sci 2008; 49 (1): 429-37.
32. Haruta M, Kosaka M, Kanegae Y, et al. Induction of photoreceptor-specific phenotypes in adult mammalian iris
tissue. Nat Neurosci 2001; 4 (12): 1163-4.
33. Sun G, Asami M, Ohta H, Kosaka J, Kosaka M. Retinal stem/progenitor properties of iris pigment epithelial cells.
Dev Biol. 2006;289 (1): 243-52.
34. Semkova I, Kreppel F, Welsandt G, et al. Autologous transplantation of genetically modified iris pigment epithelial
cells: a promising concept for the treatment of age-related macular degeneration and other disorders of the eye.
Proc Natl Acad Sci USA 2002; 99 (20):13090-5.
27
Textbook of Vitreoretinal Diseases and Surgery
35. Thumann G, Bartz-Schmidt KU, El Bakri H, et al. Transplantation of autologous iris pigment epithelium to the
subretinal space in rabbits. Transplantation 1999; 68 (2):195-201.
36. Thumann G, Aisenbrey S, Schraermeyer U, et al. Transplantation of autologous iris pigment epithelium after
removal of choroidal neovascular membranes. Arch Ophthalmol 2000; 118 (10):1350-5.
37. Kano T, Abe T, Tomita H, Sakata T, Ishiguro S, Tamai M. Protective effect against ischemia and light damage of iris
pigment epithelial cells transfected with the BDNF gene. Invest Ophthalmol Vis Sci 2002; 43 (12): 3744-53.
38. Hojo M, Abe T, Sugano E, et al. Photoreceptor protection by iris pigment epithelial transplantation transduced with
AAV-mediated brain-derived neurotrophic factor gene. Invest Ophthalmol Vis Sci 2004; 45 (10): 3721-6.
39. Zhao X, Das AV, Thoreson WB, et al. Adult corneal limbal epithelium: a model for studying neural potential of non-
neural stem cells/progenitors. Dev Biol. 2002; 250 (2): 317-31.
40. Zhao X, Das AV, Bhattacharya S, et al. Derivation of neurons with functional properties from adult limbal epithelium:
implications in autologous cell therapy for photoreceptor degeneration. Stem Cells. 2008; 26 (4): 939-49.
41. Seigel GM, Sun W, Salvi R, Campbell LM, Sullivan S, Reidy JJ. Human corneal stem cells display functional neuronal
properties. Mol Vis 2003; 9: 159-63.
42. Hirano M, Yamamoto A, Yoshimura N, et al. Generation of structures formed by lens and retinal cells differentiating
from embryonic stem cells. Dev Dyn 2003; 228 (4): 664-71.
43. Aoki H, Hara A, Nakagawa S, et al. Embryonic stem cells that differentiate into RPE cell precursors in vitro develop
into RPE cell monolayers in vivo. Exp Eye Res 2006; 82 (2): 265-74.
44. Zhao X, Liu J, Ahmad I. Differentiation of embryonic stem cells into retinal neurons. Biochem Biophys Res Commun
2002; 297 (2): 177-84.
45. Ikeda H, Osakada F, Watanabe K, et al. Generation of Rx+/Pax6+ neural retinal precursors from embryonic stem
cells. Proc Natl Acad Sci USA 2005; 102 (32):11331-6.
46. Banin E, Obolensky A, Idelson M, et al. Retinal incorporation and differentiation of neural precursors derived from
human embryonic stem cells. Stem Cells 2006; 24 (2): 246-57.
47. Lamba DA, Karl MO, Ware CB, Reh TA. Efficient generation of retinal progenitor cells from human embryonic
stem cells. Proc Natl Acad Sci USA 2006;103 (34):12769-74.
48. Ueno M, Matsumura M, Watanabe K, et al. Neural conversion of ES cells by an inductive activity on human
amniotic membrane matrix. Proc Natl Acad Sci USA 2006; 103 (25): 9554-9.
49. Osakada F, Ikeda H, Mandai M, et al. Toward the generation of rod and cone photoreceptors from mouse, monkey
and human embryonic stem cells. Nat Biotechnol 2008; 26 (2): 215-24.
50. Watanabe K, Kamiya D, Nishiyama A, et al. Directed differentiation of telencephalic precursors from embryonic
stem cells. Nat Neurosci 2005; 8 (3): 288-96.
51. Meyer JS, Katz ML, Maruniak JA, Kirk MD. Embryonic stem cell-derived neural progenitors incorporate into
degenerating retina and enhance survival of host photoreceptors. Stem Cells 2006; 24 (2): 274-83.
52. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Viable offspring derived from fetal and adult mammalian
cells. Nature 1997; 385 (6619): 810-3.
53. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures
by defined factors. Cell 2006; 126 (4): 663-76.
54. Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature 2007;
448 (7151): 313-7.
55. Arnhold S, Klein H, Semkova I, Addicks K, Schraermeyer U. Neurally selected embryonic stem cells induce tumor
formation after long-term survival following engraftment into the subretinal space. Invest Ophthalmol Vis Sci
2004;45(12):4251-5.
56. Sanchez-Ramos J, Song S, Cardozo-Pelaez F, et al. Adult bone marrow stromal cells differentiate into neural cells
in vitro. Exp Neurol 2000;164(2):247-56.
57. Chan-Ling T, Baxter L, Afzal A, et al. Hematopoietic stem cells provide repair functions after laser-induced Bruch’s
membrane rupture model of choroidal neovascularization. Am J Pathol 2006;168(3):1031-44.
58. Atmaca-Sonmez P, Li Y, Yamauchi Y, et al. Systemically transferred hematopoietic stem cells home to the subretinal
space and express RPE-65 in a mouse model of retinal pigment epithelium damage. Exp Eye Res 2006;83(5):1295-
1302.
59. Kicic A, Shen WY, Wilson AS, Constable IJ, Robertson T, Rakoczy PE. Differentiation of marrow stromal cells into
photoreceptors in the rat eye. J Neurosci 2003;23(21):7742-9.
60. Arnhold S, Heiduschka P, Klein H, et al. Adenovirally transduced bone marrow stromal cells differentiate into
pigment epithelial cells and induce rescue effects in RCS rats. Invest Ophthalmol Vis Sci 2006;47(9):4121-9.
61. Asahara T, Masuda H, Takahashi T, et al. Bone marrow origin of endothelial progenitor cells responsible for
postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res 1999;85(3):221-8.
28
Cell Therapy in Retinal Diseases
62. Otani A, Kinder K, Ewalt K, Otero FJ, Schimmel P, Friedlander M. Bone marrow-derived stem cells target retinal
astrocytes and can promote or inhibit retinal angiogenesis. Nat Med 2002;8(9):1004-10.
63. Otani A, Dorrell MI, Kinder K, et al. Rescue of retinal degeneration by intravitreally injected adult bone marrow-
derived lineage-negative hematopoietic stem cells. J Clin Invest 2004;114(6):765-74.
64. Nishida A, Takahashi M, Tanihara H, et al. Incorporation and differentiation of hippocampus-derived neural stem
cells transplanted in injured adult rat retina. Invest Ophthalmol Vis Sci 2000;41(13):4268-74.
65. Sakaguchi DS, Van Hoffelen SJ, Theusch E, et al. Transplantation of neural progenitor cells into the developing
retina of the Brazilian opossum: an in vivo system for studying stem/progenitor cell plasticity. Dev Neurosci
2004;26(5-6):336-45.
66. Goodwin HS, Bicknese AR, Chien SN, Bogucki BD, Quinn CO, Wall DA. Multilineage differentiation activity by cells
isolated from umbilical cord blood: expression of bone, fat, and neural markers. Biol Blood Marrow Transplant
2001;7(11):581-8.
67. Buzanska L, Machaj EK, Zablocka B, Pojda Z, Domanska-Janik K. Human cord blood-derived cells attain neuronal
and glial features in vitro. J Cell Sci 2002;115(Pt 10):2131-8.
68. Koike-Kiriyama N, Adachi Y, Minamino K, et al. Human cord blood cells can differentiate into retinal nerve cells.
Acta Neurobiol Exp (Wars) 2007;67(4):359-65.
29
Textbook of Vitreoretinal Diseases and Surgery
Introduction
As the diabetic population burgeons globally, so will the unusual presentations of the disease become
increasingly manifest. The retinopathy of diabetes will be no exception to this rule. This essay attempts
to chronicle the various atypical manifestations of diabetic retinopathy (DR) hitherto reported. The
protean manifestations can be due to the inherent variability of DR, systemic and ocular diseases
that may modify its course or as a result of exceptional novel features.
FEATURELESS RETINA
Featureless retina is a unique, rare type of PDR wherein retinal neovascularization may present
without the characteristic preproliferative background retinal lesions such as microaneurysms,
hemorrhages, cotton-wool spots and intraretinal microvascular abnormalities. The absence of these
features may be explained by the presence of extensive areas of capillary non-perfusion seen in these
cases which leads to the disappearance of these lesions in these severely ischemic zones.4 Although
it may appear atrophic at first glance, fluorescein angiography may reveal undetected areas of
neovascularization with extensive capillary non-perfusion (Figures 3-1A and B).
FIGURES 3-1A AND B: (A) Fundus photograph of an eye that apparently looks like a case of mild nonproliferative
diabetic retinopathy. (B) Fluorescein angiogram of the eye reveals extensive capillary nonperfusion and leakage from
neovascular tissue
— Voi voi, enhän minä saata isää pettää, vaikeroi Plotina, en ole
vielä koskaan pettänyt häntä.
Alma juoksi kotiin noutamaan lankaa ja neulaa, sillä Plotina oli niin
hämmennyksissään kaikesta siitä väkivaltaisuudesta, mitä hänen
kodissaan harjoitettiin, ettei löytänyt omia ompeluvehkeitäänkään.
Palatessaan Alma toi mukanaan pari pöytäliinaa ja puhtaan
päällyspeitteen.
— On.
— Voi, Kaarina, kuinka sinä olet hyvä. Kyllä minä olisin iloinen, jos
tulisit, pelkään, ettei Sohvi-täti ole niin vaatimaton ruuan suhteen
kuin isä. Minulta usein ruoka palaa pohjaan tai unohtuu suola.
Tohtorinna Holstin mielestä oli välttämätöntä, että tytöt rinnan
lukujen kanssa oppivat käytännöllisiä toimia. Kaarina oli siis tottunut
liikkumaan keittiössä, vaikkei hän mikään ensiluokan kokki ollutkaan.
Kaarina niiasi.
— Jätä koko Tullan joukko ja tule pois meille, houkutteli Erkki. Äiti
ottaa sinut avosylin vastaan, meillähän ei ole tyttöjä koko talossa,
kaksi poikaa vain. Pidämme sinua kuin kukkaa kämmenellä.
— Olet ehkä oikeassa, mutta tule sitten, kun olet heistä päässyt.
Illalla hän sai kaksi kirjettä postista. Toinen oli kirjoitettu suurin,
miehekkäin kirjaimin ja kuului seuraavasti:
Äiti ja Isä.
— Jumala, kuinka hyvä sinä olet, kuiskasi hän. Minä saan siis
nähdä isän ja äidin, Lanterin ja kaikki viikon kuluttua.
Hän kavahti ylös ja syöksyi suin päin alas portaita. Siken, joka
arkihuoneessa parhaillaan järjesti vastapestyjä vaatteita kaappiin,
hän oli syleilyllään rutistaa.
*****
Plotina oli kutsuttu toisten mukana. Hänellä oli uusi kaunis puku.
Kaarina arvasi sen olevan Sohvi-tädin lahjoittaman.
— On niin ikävä, kun sinä lähdet, Kaarina, sanoi Sikke. Eikö totta,
Iisa, me olemme paljon pitäneet Kaarinasta.
Updated editions will replace the previous one—the old editions will
be renamed.
1.D. The copyright laws of the place where you are located also
govern what you can do with this work. Copyright laws in most
countries are in a constant state of change. If you are outside the
United States, check the laws of your country in addition to the
terms of this agreement before downloading, copying, displaying,
performing, distributing or creating derivative works based on this
work or any other Project Gutenberg™ work. The Foundation makes
no representations concerning the copyright status of any work in
any country other than the United States.
1.E.6. You may convert to and distribute this work in any binary,
compressed, marked up, nonproprietary or proprietary form,
including any word processing or hypertext form. However, if you
provide access to or distribute copies of a Project Gutenberg™ work
in a format other than “Plain Vanilla ASCII” or other format used in
the official version posted on the official Project Gutenberg™ website
(www.gutenberg.org), you must, at no additional cost, fee or
expense to the user, provide a copy, a means of exporting a copy, or
a means of obtaining a copy upon request, of the work in its original
“Plain Vanilla ASCII” or other form. Any alternate format must
include the full Project Gutenberg™ License as specified in
paragraph 1.E.1.
• You pay a royalty fee of 20% of the gross profits you derive
from the use of Project Gutenberg™ works calculated using the
method you already use to calculate your applicable taxes. The
fee is owed to the owner of the Project Gutenberg™ trademark,
but he has agreed to donate royalties under this paragraph to
the Project Gutenberg Literary Archive Foundation. Royalty
payments must be paid within 60 days following each date on
which you prepare (or are legally required to prepare) your
periodic tax returns. Royalty payments should be clearly marked
as such and sent to the Project Gutenberg Literary Archive
Foundation at the address specified in Section 4, “Information
about donations to the Project Gutenberg Literary Archive
Foundation.”
• You comply with all other terms of this agreement for free
distribution of Project Gutenberg™ works.
1.F.
1.F.4. Except for the limited right of replacement or refund set forth
in paragraph 1.F.3, this work is provided to you ‘AS-IS’, WITH NO
OTHER WARRANTIES OF ANY KIND, EXPRESS OR IMPLIED,
INCLUDING BUT NOT LIMITED TO WARRANTIES OF
MERCHANTABILITY OR FITNESS FOR ANY PURPOSE.
Please check the Project Gutenberg web pages for current donation
methods and addresses. Donations are accepted in a number of
other ways including checks, online payments and credit card
donations. To donate, please visit: www.gutenberg.org/donate.
Our website is not just a platform for buying books, but a bridge
connecting readers to the timeless values of culture and wisdom. With
an elegant, user-friendly interface and an intelligent search system,
we are committed to providing a quick and convenient shopping
experience. Additionally, our special promotions and home delivery
services ensure that you save time and fully enjoy the joy of reading.
ebookultra.com