Molecular Vision 1999; 5:4 <http://www.molvis.org/molvis/v5/p4/>
Received 30 December 1998 | Accepted 13 April 1999 | Published 19 April 1999
Download
Reprint


The golden age of retinal cell culture

Gail M. Seigel
 
 

Department of Neurobiology & Anatomy and Center for Visual Science, University of Rochester School of Medicine and Dentistry, Rochester, NY

Correspondence to: Gail M. Seigel, Ph.D., Box 603, Department of Neurobiology & Anatomy and Center for Visual Science, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642; Phone: (716) 273-4844; FAX: (716) 442-8766; email: gail_seigel@urmc.rochester.edu


Abstract

In the late 1950s, the study of retinal cells in vitro was in its infancy. Today, retinal cell and tissue culture is routinely used for studies of cell growth, differentation, cytotoxicity, gene expression, and cell death. This review discusses the major classifications of retinal cell and tissue culture, including primary cell/explant models, retinoblastoma cell lines, and genetically engineered cell lines. These topics are addressed in an historical perspective, coupled with present-day applications for this continually-developing technology.


Introduction

The culture of retinal cells and tissues is now a widely used tool, with broad applications in the field of Ophthalmology. As with all in vitro systems, both advantages and potential limitations exist. Advantages include highly controllable conditions which allow for measurements on a cell-by-cell basis, isolation from confounding systemic/ocular effects, time course flexibility, and a reduction in the number of animals used for research. Possible limitations include selective loss of specific cell phenotypes/functions, changes in tissue architecture, and the potentially questionable relevance of in vitro findings.

For many applications, the advantages of in vitro studies outweigh the potential limitations. With a balanced approach, in conjunction with animal studies, in vitro studies can provide important basic information about normal retinal functions. Retinal cell and tissue culture exists in three basic forms: primary cultures (reaggregate, explant, and dispersed), retinoblastoma cultures, and genetically engineered cultures.


Primary Retinal Cell Culture

Reaggregate cultures

Any cell that is isolated from a tissue, grown in vitro (prior to subculture) is considered a primary cell culture. The modern era of primary retinal cell culture began in the late 1950's with the work of A. A. Moscona [1,2] in which he elegantly demonstrated the ability of enzymatically dissociated chick neuroretinal cells to reassociate into multicellular, histotypic reaggregates under controlled conditions allowing replicable results [3]. Using this system, Moscona and Linser were able to use reaggregates to study the regulation of retinal enzymes, such as carbonic anhydrase and glutamine synthetase [4-6]. From these early studies, it became clear that cells of the neuroretina were readily isolated, were able to form cell-cell associations, and maintained many of the characteristics of the retina in situ. With this reaggregate system, important intrinsic properties of retinal cells were established. Retinal reaggregates were able to form rosettes, organized cellular structures comprised of a central lumen, surrounded by a circular array of photoreceptors with connecting cilia and external limiting membrane. In the chick reaggregate culture, rosettes formed that were characteristic of the age of origin of the cells [7,8], while the retinal cells therein were able to continue some degree of development with regard to tissue organization, synaptogenesis, photoreceptor differentiation, and enzyme function [3,9-11]. As seen in Figure 1, the cellular organization of retinal reaggregate cultures, feature rosette formation which is visible with light microscopy.

Retinal reaggregate culture has recently undergone a "pellet culture" modification [12], in which slow-speed centrifugation of dissociated retinal cells promotes reaggregation. Reaggregate/pellet cultures are used for studies of differentiation [12], cell-cell interactions [13], and even the disaggregating effects of retroviruses [14]. Importantly, the potential significance of the retinal pigment epithelium (RPE) for the proper cellular stratification of the neuroretina was revealed with this technique, using chimaeric reaggregates of neuroretina and RPE [15,16].

The establishment of the retinal reaggregate cell culture system led to the evolution of other types of primary retinal culture. For the purposes of this discussion, they will be classified as explant cultures and dispersed cell cultures.

Explant cultures

Explant cultures of whole or partial retina were established in an attempt to preserve the tissue architecture characteristic of the retina in situ. With retinal explants, as with other explanted tissues, cells retain many histological and biochemical features and can be maintained for several days, or even weeks. These types of retinal cultures cannot be propogated, exhibit greater degrees of experimental variation among sister cultures, and are difficult to use for some quantitative studies due to variations in geometry and composition [17]. Retinal explant cultures have been ideal for studies of synaptic organization [18], cell-cell interactions [19], outgrowth of cells and their neurites [20-22], and retinal differentiation [23,24]. Retinal explants retain the highest degree of tissue preservation of all retinal cell culture systems and continue to be the method of choice for in vitro studies that require intact cell-cell associations.

Retinal explant cultures have been especially helpful in addressing a fundamental question in the evolution of retinal cell culture, namely, how do cultured retinal cells differ from their intact counterparts in vivo? Cultured explants from neonatal rabbit exhibit markers of the GABAergic phenotype for several days in a manner consistent with that seen in vivo [24]. In a comparative study by Germer and colleagues [25], development of an outer plexiform layer, as well as ribbon and conventional synapses of the neonatal rabbit retina in vitro followed a similar pattern to that seen in vivo. The photoreceptor cells in their preparation grew well-developed inner segments and cilia, but not mature outer segments. In a second study of these neonatal rabbit retinal explants [26], glutamine synthetase expression in Müller cells was lower in vitro by an order of magnitude. The Müller cells of the rabbit retinal explant cultures could be stimulated to express additional glutamine synthetase by addition of glucocorticoid hormones or retinal pigment epithelial cell-conditioned medium. Therefore, loss of systemic, circulating factors may account for some of the changes seen when isolated retinal tissue is grown in culture.

Chick retinal explants have been useful in identifying developmental changes characteristic of in vitro conditions. Influences of the substratum (collagen, poly L-lysine, etc.) have been noted with regard to intermediate filament expression in chick embryonic retinal explants [27]. Although neurofilament L was not detected in vitro, the early expression of neurofilament subunits M and H during the maturation process was dependent upon the type of substratum present on the dish (27). In an autoradiographic study of developing chick retina [28], the major differences observed were that the retinal explants grown in culture contained cells that were morphologically more spindle-shaped and the retina, as a whole, was thinner than in vivo. Importantly, cultured chick retinal explant development lagged behind age-matched in vivo retinae by approximately 24 h. These differences can be considered when assessing the use of retinal explants to address experimental problems.

Dispersed cell cultures

Dissociated-dispersed retinal cells, from embryonic, postnatal, and adult animals, lack the cell-cell organization of explant cultures, but can be more useful for some transfection studies, patch-clamp recordings, videomicroscopy, and quantitative analyses where access to individual cells is important or where intercellular communication must be limited.

Since dispersed cell cultures are even further removed from the orderly structure of the intact retina, identification of these dispersed cells is paramount. This problem can be illustrated in the dispersed rat embryonic retinal cell monolayer culture shown in Figure 2. Flat cells and neuronal-like cells are evident, however, morphology alone is not enough to positively identify each of the six major phenotypes of the neuroretina: ganglion, bipolar, photoreceptor, amacrine, horizontal, and Müller cells. With the advent of retina specific antibodies, such as those for rat from the lab of Barnstable [29,30] and those for chicken from Lemmon [31,32], specific retinal cell types could be identified in vitro on the basis of retina-specific marker expression, rather than on less-reliable morphological characteristics. In addition to the development of specific antibodies, retinal cell identification evolved to include tetanus-toxin binding of retinal neurons [33], autoradiographic localization of labelled neurotransmitters [34], and electron microscopic identification of synaptic elements [35]. For particular retinal cell types of interest, highly specific means of identification were introduced. Ganglion cells are identified by pre-labelling in vivo (prior to cell culture) by intraxonal retrograde transport of horseradish peroxidase [36]. Horizontal cells have been identified based on specific glutamate-modulated K+ current conductance, in conjunction with immunoreactivity to parvalbumin [37]. For amacrine cells, autoradiographic evidence of acetylcholine [38,39], GABA immunocytochemistry [40] and autoradiographic demonstration of 3H-muscimol uptake [40] have proven useful for identification purposes.

Retinal cell identification procedures were also adapted to enrich cultures for specific retinal cell populations. As with neural cell culture, enrichment procedures involved retrograde labelling, gradient separation, both positive and negative selection procedures (chemical and immunological), and mechanical means of separation. Retrograde labelling [41] coupled with metrizamide gradient separation [42] has been used to select for ganglion cell types. Another ganglion cell enrichment procedure has relied upon the Thy-1 marker and other ganglion-specific antibodies for immunological panning [43]. For positive selection of different cell types, retina-specific cell surface markers used for fluorescence-activated cell sorting [44] can separate mixed retinal preparations into nearly purified cultures. Selective destruction, such as anti-opsin/complement treatment of cell cultures, can help exclude photoreceptors from a final cell population [45]. Photoreceptor enrichment has also been possible through the chemical destruction of undesired retinal neurons by the photoreceptor-sparing toxins kainic acid and ß-bungarotoxin [46] and by the non-ionic detergent Triton X-100 [47,48]. Mechanical selection through the adept use of a vibratome has been used as a physical means of photoreceptor purification [49]. Sequential papain and DNAse enzymatic digestion coupled with density gradient centrifugation has allowed for pure Müller cell cultures [50]. Enzymatic dissociation in combination with velocity sedimentation has been reported for isolation of purified horizontal cells [51]. For more general applications, rat retinal sublayers can be separated by mild enzymatic treatment with trypsin followed by physical separation of retinal tissue into the outer nuclear layer, inner nuclear layer, inner plexiform layer, and ganglion cell/nerve fiber layer [52].

The origin of retinal tissue used for monolayer and explant cell culture has included a diverse assortment of creatures, including human [53-59], rabbit [24,25], rat [30], mouse [60], pig [61] chicken [46], drosophila [62], moth [63], zebrafish [64], carp, [65], goldfish [66,67], frog [68,69], turtle [70], gecko [71] and even camel [72,73]. As a rule, embryonic/postnatal tissue has been easier to propogate than adult tissue. Survival, as well as the differentiation of many primary retinal cell cultures, has been enhanced with the addition of specific factors to the culture medium, such as basic fibroblast growth factor [74], acidic fibroblast growth factor [75], ciliary neurotrophic factor [76,77], neurotrophin-3 [78], glial-derived neurotrophic factor [79] and retinoids [80,81]. RPE conditioned medium has been shown to possess potent survival and maturation factors for neural retinal cells in vitro [82,83], including pigment epithelium-derived factor (PEDF) [84,85].

Retinal Cell Differentiation in Primary Culture

Primary retinal cell culture models have been ideal for retinal developmental studies. Isolated from confounding ocular/systemic influences, retinal cells and explants have been used to address basic questions as to the intrinsic vs. extrinsic nature of retinal cell differentiation. Retinal progenitor cell fate was shown to be affected by cell-cell interactions [86] and was dependent upon retinal progenitor cell "competence" to respond to environmental cues [87]. In the absence of extrinsic signals, murine retinal precursor cells grown in clonal density cultures (as single cells) could generate both neurons and glia [88]. In the chicken, the majority of similarly isolated retinal progenitor cells tended to develop into photoreceptors in the absence of intraretinal influences, a "photoreceptor default" pathway [89]. Furthermore, retinal cell fate could be determined after the time of terminal mitosis [90]. All of these developmental findings relied upon coordination between in vivo and in vitro experimentation to identify some of the powerful differentiation influences of the retina.

With the advent of improved purification procedures, such as a recently modified vibratome-mediated purification of photoreceptors [91], primary retinal cell culture methods continue to be refined and re-defined. More detailed technical advice beyond the scope of this overview is available [92], and continues to evolve. Primary retinal cell and tissue culture remains the most widely-used form of retinal cell culture.


Retinoblastoma cells

The major drawbacks of primary retinal cell cultures are their limited supply and finite lifespan. Investigators have addressed this problem by establishing proliferative retinal cell cultures of virtually infinite lifespan from retinoblastoma tumors. These retinoblastoma cell lines are not only immortal, but can also be stored frozen in liquid Nitrogen for later use, unlike primary cell cultures. The first retinoblastoma cell line described was the Y79 cell line, derived from the intraocular tumor of a 2.5 year old human [93]. Shortly thereafter, the WERI-Rb1 [94], WERI-Rb24, and WEI-Rb27 [95] cell lines were also established from human intraocular tumors. Other human retinoblastoma cell lines include FMC-Rb1 [96], Rb 355-7 [97], SO-Rb50 [98], and HXO-Rb44 [99]. Through analysis of genetic polymorphisms, it was determined that Y79, Rb 355-7, and WERI-Rb27 are genetically related [100]. These human retinoblastoma tumor cell cultures have several characteristics in common, such as growth in suspension as "grape-like" clusters (Figure 3), immunoreactivity to glial and/or neuronal markers [101-103], and the ability to form tumors in vivo [94,99,104]. Retinoblastoma cells were found to exhibit alterations in the expression of the retinoblastoma susceptibility gene, Rb [105]. Alterations in Rb expression sparked significant interest in the use of retinoblastoma cells as tools to gain insight into mutational mechanisms of Rb inactivation.

In the flurry of Rb research in the late 1980s and early 1990s, retinoblastoma cells were used to elucidate the potential role of Rb as a tumor suppressor. Although replacement of a wild-type Rb gene into Rb-defective or deficient cells decreased growth rate, colony-formation in soft agar, and tumorigenicity [106], Rb reconstitution alone was not enough to completely prevent retinoblastoma cells from forming tumors in vivo [107,108]. The Rb gene was shown to be an important component in the coordination of normal retinal growth and differentiation [109] though it did not appear to be a strong tumor suppressor. Elements of the Rb gene family were differentially expressed during mammalian embryogenesis [110] and mice deficient in the Rb gene were non-viable, with defects in neurogenesis and hematopoiesis [111]. Significant strides were made in the study of Rb gene expression with the aid of retinoblastoma cell culture systems.

The undifferentiated nature of the Y79 retinoblastoma cells also made them very useful in bioassays to test putative retinal differentiating agents. Morphological observations could be made, once these cells were attached to suitable matrices [112,113]. Differentiating agents, such as dibutyryl cAMP, retinoids, and sodium butyrate [114-118] were found to affect the morphology, growth rate, and biochemistry of Y79 cells in vitro. Our own studies showed that the growth inhibitor succinylated concanavalin A (SCA) could reversibly decrease the expression of the N-MYC protein in Y79 cells and induce expression of the photoreceptor marker interphotoreceptor retinoid-binding protein [119]. Profound morphological effects were seen in adherent Y79 cells upon treatment with PEDF, including the formation of large, organized rosettes and significant neurite outgrowth [84,85]. However, even this differentiating treatment was not enough to block the tumor-forming abilities of this cell line [120]. Interestingly, sodium butyrate, a less neuritogenic compound than PEDF, inhibited tumor-formation in vivo [121], presumably due to its growth-inhibiting effects.

Recently, Y79 cells have been utilized for studies on the efficacy of anti-cancer drugs [122-125], radiation [126], and photodynamic therapies [127]. Under some of these treatment conditions, Y79 cells undergo apoptotic cell death through a p53-dependent mechanism [128]. These findings suggest a potential role for the manipulation of pro-apoptotic genes in the treatment of retinoblastoma. In addition to the human retinoblastoma cell lines, mouse [129] and rat [130] cell lines exist that continue to be useful in studies of retinal cell differentiation, tumor biology, and anti-cancer therapeutics.


Genetically engineered retinal cultures

In rare instances, continuously-growing cell lines arise spontaneously from primary cultures [131-134]. More commonly, cell-cycle-related genes that induce cell proliferation, such as SV40 large T antigen, E1A/E1B gene of adenovirus, or src are introduced by investigators to promote an immortalized, growth stimulated culture. In genetically engineered retinal cultures, some resulting cell lines will lack the tumor-forming potential of the aforementioned retinoblastoma cell lines, and possibly retain more differentiated features. Like the retinoblastoma cell lines, the genetically engineered immortalized retinal cell cultures can withstand long-term freezer storage.

In the late 1980s, human retinoblasts were transformed with SV40 virus, adenovirus, and ras genes for the purpose of studying oncogene activity, efficiency of transformation, and tumorigenicity [135]. In these studies, retinal cell lines were used to demonstrate that stable, malignant transformation of primary human cells required the cooperation of more than one oncogene [136]. As with retinoblastoma cells, these genetically engineered cells, called Ad 12 HER 10, responded to dibutyryl cAMP in a differentiation process characterized by cell rounding and neuritic-like process formation [137].

KGLDMSM, another human retinal cell line, developed by Dutt and colleagues [138], was induced to progress through the cell cycle by expression of SV40 Large T Antigen. KGLDMSM cells were shown to express neuron-specific enolase, neurofilament, glycine receptor, synaptophysin and secretogranin [139]. These cells also responded to cAMP and the phorbol ester TPA by undergoing morphological changes characterized by extension of neuritic processes and c-fos expression [139]. A retinal Müller cell line, of rat origin, was also established by the introduction of SV40 large T antigen [140]. The Müller cell line, termed rMC-1, was shown to express GFAP, a marker for reactive gliosis and cellular retinaldehyde binding protein (CRALBP), a marker for Müller cells in the adult retina. Similarly, a rat retinal Müller cell line established by introduction of the human papilloma virus E6/E7 gene [141] retained expression of Müller cell markers, including S-100, carbonic anhydrase C, CRALBP, and GFAP, but not glutamine synthetase.

In the early 1970s, Pessac, Calothy, and colleagues developed a retinal cell culture system to study the effects of oncogenes, such as v-src, on the proliferative and differentiating capacity of chicken and quail neuroretinal cells [142,143]. These Rous sarcoma virus transformed avian retinal cells were still able to retain retinal characteristics, such as choline acetyl transferase, glutamic acid decarboxylase, tetanus-toxin binding [144], and A2B5 immunoreactivity [145]. Retrovirally or oncogenically transformed avian retinal cells have been used recently for the identification of new retina-specific genes, such as the melanocyte specific gene QNR-71 [146], and the characterization of new avian cell lines expressing mature retinal markers [147].

Rous sarcoma virus-transformed chick neuroretinal cells do not always remain growth-stimulated. In the case of LA29NR, a chick neuroretinal cell line infected with Rous sarcoma virus, cells were proliferative for 15 passages, but then succumbed to a senescent process [148]. The senescence was associated with a loss of transformed phenotype and eventually cell death. Nevertheless, even 15 passages represented a prolonged lifespan for these cells and allowed for an additional 4 months of experimentation, not possible with primary cultures.

Retinal cells induced to progress through the cell cycle often maintain features of retinal precursor cells, rather than a mature retinal phenotype. Such retinal precursor features are characteristic of the cell lines developed in our own laboratory, namely the E1A-NR.3 and the clonally-derived R28 cell cultures [149]. These cells were established from postnatal day 6 Sprague-Dawley rat retinae and were forced to remain active in the cell cycle by a replication-incompetent retrovirus containing the 12S portion of the adenovirus E1A gene. The 12S portion of the E1A gene was used to promote cell growth, while avoiding tumor-forming potential seen in SV40 large T antigen-expressing cells [150,151]. E1A-NR.3 and R28 cells exhibit contact-inhibited growth [149] and were transplanted intraocularly into rats with no tumor-formation seen in 42 eyes [152]. The E1A-NR.3 and R28 retinal cells have been useful for retinal transplantation experiments [152], and as targets for cytotoxicity and neuroprotection studies [153-155]. These cells have been shown to express markers of photoreceptor [149,156], Müller [157], and ganglion cell phenotypes (Figure 4).

Genetically engineered retinal cell lines continue to be used for studies requiring large numbers of cells, and can be cloned by limiting dilution if homogeneity is desired. Additionally, these cell lines can be custom-designed from isolated retinal cell types and offer a great deal of flexibility in experimental design.


Summary

Retinal cell culture has proven to be a powerful tool in Ophthalmic research. While no cell culture described can replace the intact eye, these cultures do provide controllable experimental systems for the examination of fundamental retinal processes. The major characteristics of retinal culture systems are summarized in Table 1. The relevancy of retinal cell culture experiments remains a subject of debate. This debate will continue as we seek to improve upon retinal cell culture systems and create cell lines that more closely approximate the retina in situ.


Acknowledgements

The author wishes to thank Anna Paxhia, Thurma V. McDaniel, and Lois Chiu for technical support. G. M. S. is supported by EY10676 from the National Eye Institute.

All animal experimentation was accomplished in accordance with the ARVO statement on the use of animals in Ophthalmic research.


References

1. Moscona AA. Formation of lentoids by dissociated retinal cells of the chick embryo. Science 1957; 125:598-9.

2. Moscona AA. Patterns and mechanisms of tissue reconstruction from dissociated cells. In: Rudnick D, editor. Developing systems and their control. New York: Ronald Press; 1960. p. 45-70.

3. Moscona AA. Rotation-mediated histogenetic reaggregation of dissociated cells: a quantifiable approach to cell interactions in vitro. Exp Cell Res 1961; 22:455-75.

4. Linser P, Moscona AA. Induction of glutamine synthetase in embryonic neural retina: localization in Muller fibers and dependence on cell interactions. Proc Natl Acad Sci U S A 1979; 76:6476-80.

5. Linser P, Moscona AA. Carbonic anhydrase C in the neural retina: transition from generalized to glia-specific cell localization during embryonic development. Proc Natl Acad Sci U S A 1981; 78:7190-4.

6. Linser PJ, Moscona AA. Induction of glutamine synthetase in embryonic neural retina: its suppression by the gliatoxic agent alpha-aminoadipic acid. Brain Res 1981; 227:103-19.

7. Sheffield J, Li HP. Interactions among cells of the developing retina in vitro. Am Zool 1987; 27:145-59.

8. Moscona AA. Recombination of dissociated cells and the development of cell aggregates. In: Wilmer EN, editor. Cells and tissues in culture: methods, biology, and physiology. London: Academic Press; 1965. p. 489-529.

9. Sheffield JB, Moscona AA. Electron microscopic analysis of aggregation of embryonic cells: the structure and differentiation of aggregates of neural retina cells. Dev Biol 1970; 23:36-61.

10. Stefanelli A, Zacchei AM, Caravita S, Cataldi A, Ieradi LA. New-forming retinal synapses in vitro. Experientia 1967; 23:199-200.

11. Vogel Z, Daniels MP, Nirenberg M. Synapse and acetylcholine receptor synthesis by neurons dissociated from retina. Proc Natl Acad Sci U S A 1976; 73:2370-4.

12. Watanabe T, Voyvodic JT, Chan-Ling T, Sagara H, Hirosawa K, Mio Y, Matsushima S, Uchimura H, Nakahara K, Raff MC. Differentiation and morphogenesis in pellet cultures of developing rat retinal cells. J Comp Neurol 1997; 377:341-50.

13. Pearlman EM, Seigel GM, Notter MF. Induction of c-fos by exctitatory amino acids in developing chick retina is affected by changes in cellular interactions. J Neurosci Res 1993; 36:252-9.

14. Notter MF, Navon SE, Fung BK, Balduzzi PC. Infection of neuroretinal cells in vitro by avian sarcoma viruses UR1 and UR2: transformation, cell growth stimulation, and changes in transducin levels. Virology 1987; 160:489-93.

15. Vollmer G, Layer PG, Gierer A. Reagregation of embryonic chick retinal cells: pigment epithelial cells induce a high order of stratification. Neurosci Lett 1984; 48:191-6.

16. Vollmer G, Layer PG. An in vitro model of proliferation and differentiation of the chick retina: coaggregates of retinal and pigment epithelial cells. J Neurosci 1986; 6:1885-96.

17. Freshney RI, editor. Animal cell culture: a practical approach. 2nd ed. New York: Oxford University Press; 1992.

18. Sassoe-Pognetto M, Feigenspan A, Bormann J, Wassle H. Synaptic organization of an organotypic slice culture of the mammalian retina. Vis Neurosci 1996; 13:759-71.

19. Feigenspan A, Bormann J, Wassle H. Organotypic slice culture of the mammalian retina. Vis Neurosci 1993; 10:203-17.

20. Yamagata M, Sanes JR. Lamina-specific cues guide outgrowth and arborization of retinal axons in the optic tectum. Development 1995; 121:189-200.

21. Carri NG, Perris R, Johansson S, Ebendal T. Differential outgrowth of retinal neurites on purified extracellular matrix molecules. J Neurosci Res 1988; 19:428-39.

22. Carri NG, Ebendal T. Organotypic cultures of neural retina: neurite outgrowth stimulated by brain extracts. Brain Res 1983; 282:219-29.

23. Veroman SA. [Retinal differentiation of turkey embryos of various ages in organotypical cultures]. Arkh Anat Gistol Embriol 1981; 81:50-7.

24. Rowe-Rendleman C, Mitchell CK, Habrecht M, Redburn DA. Expression and downregulation of the GABAergic phenotype in explants of cultured rabbit retina. Invest Ophthamol Vis Sci 1996; 37:1074-83.

25. Germer A, Kuhnel K, Grosche J, Friedrich A, Wolburg H, Price J, Reichenbach A, Mack AF. Development of the neonatal rabbit retina in organ culture. 1. Comparison with histogenesis in vivo, and the effect of a gliotoxin (alpha-aminoadipic acid). Anat Embryol (Berl) 1997; 196:67-79.

26. Germer A, Jahnke C, Mack A, Enzmann V, Reichenbach A. Modification of glutamine synthetase expression by mammalian Muller (glial) cells in retinal organ cultures. Neuroreport 1997; 8:3067-72.

27. Torelli S, Sogos V, Marzilli MA, D'Atri M, Gremo F. Developmental expression of intermediate filament proteins in the chick embryo retina: in vivo and in vitro comparison. Exp Biol 1989; 48:187-96.

28. Spence SG, Robson JA. An autoradiographic analysis of neurogenesis in the chick retina in vitro and in vivo. Neuroscience 1989; 32:801-12.

29. Barnstable CJ, Hofstein R, Akagawa K. A marker of early amacrine cell development in rat retina. Brain Res 1985; 352:286-90.

30. Akagawa K, Barnstable CJ. Identification and characterization of cell types in monolayer cultures of rat retina using monoclonal antibodies. Brain Res 1986 383:110-20.

31. Lemmon V. Monoclonal antibodies specific for glia in the chick nervous system. Brain Res 1985; 355:111-20.

32. Lemmon V. A monoclonal antibody that binds to the surface of photoreceptors. Brain Res 1988; 467:117-23.

33. Beale R, Nicholas D, Neuhoff V, Osborne NN. The binding of tetanus toxin to retinal cells. Brain Res 1982; 248:141-9.

34. Oka MS, Frederick JM, Landers RA, Bridges CD. Adult human retinal cells in culture. Identification of cell types and expression of differentiated properties. Exp Cell Res 1985; 159:127-40.

35. Sparrow JR, Hicks D, Barnstable CJ. Cell commitment and differentiation in explants of embryonic rat neural retina. Comparison with the developmental potential of dissociated retina. Brain Res Dev Brain Res 1990; 51:69-84.

36. Nurcombe V, Bennett MR. Embryonic chick retinal ganglion cells identified "in vitro". Their survival is dependent on a factor from the optic tectum. Exp Brain Res 1981; 44:249-58.

37. Picaud S, Hicks D, Forster V, Sahel J, Dreyfus H. Adult human retinal neurons in culture: Physiology of horizontal cells. Invest Ophthalmol Vis Sci 1998; 39:2637-48.

38. Masland RH, Mills JW. Autoradiographic identification of acetylcholine in the rabbit retina. J Cell Biol 1979; 83:159-78.

39. Masland RH, Mills JW, Hayden SA. Acetylcholine-synthesizing amacrine cells: identification and selective staining by using radioautography and fluorescent markers. Proc R Soc Lond B Biol Sci 1984; 223:79-100.

40. Huba R, Hofmann HD. Identification of GABAergic amacrine cell-like neurons in chick retinal monolayer cultures. Neurosci Lett 1990; 117:37-42.

41. Simon P, Thanos S. Combined methods of retrograde staining, layer-separation and viscoelastic cell stabilization to isolate retinal ganglion cells in adult rats. J Neurosci Methods 1998; 83:113-24.

42. Sarthy PV, Curtis BM, Catterall WA. Retrograde labeling, enrichment, and characterization of retinal ganglion cells from the neonatal rat. J Neurosci 1983; 3:2532-44.

43. Barres BA, Silverstein BE, Corey DP, Chun LL. Immunological, morphological, and electrophysiological variation among retinal ganglion cells purified by panning. Neuron 1988; 1:791-803.

44. Dyer SA, Derby MA, Cole GJ, Glaser L. Identification of sub-populations of chick neural retinal cells by monoclonal antibodies: a fluorescence activated cell sorter screening technique. Brain Res 1983; 285:197-203.

45. Politi L, Adler R. Selective destruction of photoreceptor cells by anti-opsin antibodies. Invest Ophthamol Vis Sci 1987; 28:118-25.

46. Politi LE, Adler R. Generation of enriched populations of cultured photoreceptor cells. Invest Ophthalmol Vis Sci 1986; 27:656-65.

47. Cahill GM, Besharse JC. Light-sensitive melatonin synthesis by Xenopus photoreceptors after destruction of the inner retina. Vis Neurosci 1992; 8:487-90.

48. Cahill GM, Besharse JC. Circadian clock functions localized in xenopus retinal photoreceptors. Neuron 1993; 10:573-7.

49. Wang X, Silverman M. [A new technique for isolation of photoreceptor layer]. Chung Hua Yen Ko Tsa Chih 1996; 32:304-6.

50. Guidry C. Isolation and characterization of porcine Muller cells. Myofibroblastic dedifferentiation in culture. Invest Ophthalmol Vis Sci 1996; 37:740-52.

51. Van Buskirk R, Dowling JE. Isolated horizontal cells from carp retina demonstrate dopamine-dependent accumulation of cyclic AMP. Proc Natl Acad Sci U S A 1981; 78:7825-9.

52. Shiosaka S, Kiyama H, Tohyama M. A simple method for the separation of retinal sublayers from the entire retina with special reference to application for cell culture. J Neurosci Methods 1984; 10:229-35.

53. Kim SU, Takahashi H. Tissue culture study of adult human retina neurons. Invest Ophthalmol Vis Sci 1988; 29:1372-9.

54. Hicks D, Forster V, Dreyfus H, Sahel J. Survival and regeneration of adult human photoreceptors in vitro. Brain Res 1994; 643:302-5.

55. Aotaki-Keen AE, Harvey AK, de Juan E, Hjelmeland LM. Primary culture of human retinal glia. Invest Ophthalmol Vis Sci 1991; 32:1733-8.

56. Kelley MW, Turner JK, Reh TA. Regulation of proliferation and photoreceptor differentiation in fetal human retinal cell cultures. Invest Ophthalmol Vis Sci 1995; 36:1280-9.

57. Gaudin C, Forster V, Sahel J, Dreyfus H, Hicks D. Survival and regeneration of adult human and other mammalian photoreceptors in culture. Invest Ophthalmol Vis Sci 1996; 37:2258-68.

58. Hu DN, Ritch R. Tissue culture of adult human retinal ganglion cells. J Glaucoma 1997; 6:37-43.

59. Arroyo JG, Ghazvini S, Char DH. An immunocytochemical study of isolated human retinal Muller cells in culture. Graefes Arch Clin Exp Ophthalmol 1997; 235:411-4.

60. Politi LE, Lehar M, Adler R. Development of neonatal mouse retinal neurons and photoreceptors in low density cell culture. Invest Ophthalmol Vis Sci 1988; 29:534-43.

61. Picaud S, Pattnaik B, Hicks D, Forster V, Fontaine V, Sahel J, Dreyfus H. GABAA and GABAC receptors in adult porcine cones: evidence from a photoreceptor-glia co-culture model. J Physiol (Lond) 1998; 513:33-42.

62. Ziemba SE, Saks S, Janviriya Y, Stephenson RS. Dissociation of photoreceptors from whole heads of the fruit fly, Drosophila melanogaster. Cell Tissue Res 1995; 280:473-7.

63. Woolever P, Pipa RL. Eye disk differentiation in the wax moth: induction in vitro. J Exp Zool 1975; 191:359-64.

64. McMahon DG. Modulation of electrical synaptic transmission in zebrafish retinal horizontal cells. J Neurosci 1994; 14:1722-34.

65. Young LH, Dowling JE. Monoclonal antibodies distinguish subtypes of retinal horizontal cells. Proc Natl Acad Sci U S A 1984; 81:6255-9.

66. Wagner EC, Raymond PA. Muller glial cells of the goldfish retina are phagocytic in vitro but not in vivo. Exp Eye Res 1991; 53:583-9.

67. Boucher SE, Hitchcock PF. Insulin-related growth factors stimulate proliferation of retinal progenitors in the goldfish. J Comp Neurol 1998; 394:386-94.

68. Hale IL, Fisher SK, Matsumoto B. Effects of retinal detachment on rod disc membrane assembly in cultured frog retinas. Invest Ophthalmol Vis Sci 1991; 32:2873-81.

69. Kaplan MW, Iwata RT, Sterrett CB. Retinal detachment prevents normal assembly of disk membranes in vitro. Invest Ophthalmol Vis Sci 1990; 31:1-8.

70. Lasater EM, Witkovsky P. Membrane currents of spiking cells isolated from turtle retina. J Comp Physiol 1990; 167:11-21.

71. Moyer RW, Firth BT, Kennaway DJ. Effect of constant temperatures, darkness and light on the secretion of melatonin by pineal explants and retinas in the gecko Christinus marmoratus. Brain Res 1995; 675:345-8.

72. al-Jafari AA, Kamal MA, Alhomida AS. On the inhibition of camel retina acetylcholinesterase activity by cycloheximide in vitro. Cell Biol Toxicol 1998; 14:167-74.

73. al-Jafari A, al-Khwyter F, Kamal MA, Alhomida AS. Kinetics for camel (Camelus dromedarius) retina acetylcholinesterase inhibition by methotrexate in vitro. Jpn J Pharmacol 1996; 72:49-55.

74. Hicks D, Courtois Y. Fibroblast growth factor stimulates photoreceptor differentiation in vitro. J Neurosci 1992; 12:2022-33.

75. Guillemot F, Cepko CL. Retinal fate and ganglion cell differentiation are potentiated by acidic FGF in an in vitro assay of early retinal development. Development 1992; 114:743-54.

76. Fuhrmann S, Kirsch M, Hofmann HD. Ciliary neurotrophic factor promotes chick photoreceptor development in vitro. Development 1995; 121:2695-706.

77. Ezzeddine ZD, Yang X, DeChiara T, Yancopoulos G, Cepko CL. Postmitotic cells fated to become rod photoreceptors can be respecified by CNTF treatment of the retina. Development 1997; 124:1055-67.

78. de la Rosa EJ, Arribas A, Frade JM, Rodriguez-Tebar A. Role of neurotrophins in the control of neural development: neurotrophin-3 promotes both neuron differentiation and survival of cultured chick retinal cells. Neuroscience 1994; 58:347-52.

79. Carwile ME, Culbert RB, Sturdivant RL, Kraft TW. Rod outer segment mainentance is enhanced in the presence of bFGF, CNTF and GDNF. Exp Eye Res 1998; 66:791-805.

80. Stenkamp DL, Gregory JK, Adler R. Retinoid effects in purified cultures of chick embryo retina neurons and photoreceptors. Invest Ophthalmol Vis Sci 1993; 34:2425-36.

81. Kelley MW, Turner JK, Reh TA. Retinoic acid promotes differentiation of photoreceptors in vitro. Development 1994; 2091-102.

82. Sheedlo HJ, Turner JE. Effects of retinal pigment epithelial cell-secreted factors on neonatal rat retinal explant progenitor cells. J Neurosci Res 1996; 44:519-31.

83. Sheedlo HJ, Nelson TH, Lin N, Rogers TA, Roque RS, Turner JE. RPE secreted proteins and antibody influence photoreceptor cell survival and maturation. Brain Res Dev Brain Res 1998; 107:57-69.

84. Steele FR, Chader GJ, Johnson LV, Tombran-Tink J. Pigment epithelium-derived factor: neurotrophic activity and identification as a member of the serine protease inhibitor gene family. Proc Natl Acad Sci U S A 1993; 90:1526-30.

85. Becerra SP, Palmer I, Kumar A, Steele F, Shiloach J, Notario V, Chader GJ. Overexpression of fetal human pigment epithelium-derived factor in Escherichia coli. A functionally active neurotrophic factor. J Biol Chem 1993; 268:23148-56.

86. Reh TA. Cellular interactions determine neuronal phenotypes in rodent retinal cultures. J Neurobiol 1992; 23:1067-83.

87. Cepko CL, Austin CP, Yang X, Alexiades M, Ezzeddine D. Cell fate determination in the vertebrate retina. Proc Natl Acad Sci U S A 1996; 93:589-95.

88. Jensen AM, Raff MC. Continuous observation of multipotential retinal progenitor cells in clonal density culture. Dev Biol 1997; 188:267-79.

89. Repka A, Adler R. Differentiation of retinal precursor cells born in vitro. Dev Biol 1992; 153:242-9.

90. Belecky-Adams T, Cook B, Adler R. Correlations between terminal mitosis and differentiated fate of retinal precursor cells born in vivo and in vitro: analysis with the "window-labeling" technique. Dev Biol 1996; 178:304-15.

91. Fontaine V, Kinkl N, Sahel J, Dreyfus H, Hicks D. Survival of purified rat photoreceptors in vitro is stimulated directly by fibroblast growth factor-2. J Neurosci 1998; 18:9662-72.

92. Finlay D, Wilkinson G, Kypta R, de Curtis I, Reichardt L. Retinal cultures. Methods Cell Biol 1996; 51:265-83.

93. Reid TW, Albert DM, Rabson AS, Russell P, Craft J, Chu EW, Tralka TS, Wilcox JL. Characteristics of an established cell line of retinoblastoma. J Natl Cancer Inst 1974; 53:347-60.

94. McFall RC, Sery TW, Makadon M. Characterization of a new continuous cell line derived from a human retinoblastoma. Cancer Res 1977; 37:1003-10.

95. Sery TW, Lee EY, Lee WH, Bookstein R, Wong V, Shields JA, Augsburger JJ, Donoso LA. Characteristics of two new retinoblastoma cell lines: WERI-Rb24 and WERI-Rb27. J Pediatr Ophthalmol Strabismus 1990; 27:212-7.

96. Seshedri R, Matthews C, Henderson DW, Zola H, Callen D. A human retinoblastoma cell line expressing the common acute lymphoblastic leukemia antigen and displaying an unusual chromosome abnormality. Cancer Genet Cytogenet 1986; 20:345-54.

97. Fournier GA, Sang DN, Albert DM, Craft JL. Electron microscopy and HLA expression of a new cell line of retinoblastoma. Invest Ophthalmol Vis Sci 1987; 28:690-9.

98. Yi YZ, Jie J. [Establishment and characterization of a human retinoblastoma cell line SO-Rb50]. Chung Hua Yen Ko Tsa Chih 1990; 26:355-9.

99. Xu H, Wang C, Zhu H, Liu S, Xu X, Jiang Y. Characteristics of an established retinoblastoma cell line HXO-Rb44. Yen Ko Hsueh Pao 1995; 11:16-21.

100. Madreperla SA, Bookstein R, Jones OW, Lee WH. Retinoblastoma cell lines Y79, RB355 and WERI-Rb27 are genetically related. Ophthalmic Paediatr Genet 1991; 12:49-56.

101. Kyritsis AP, Tsokos M, Triche TJ, Chader GJ. Retinoblastoma-origin from a primitive neuroectodermal cell? Nature 1984; 307:471-3.

102. Jiang Q, Lim R, Blodi FC. Dual properties of cultured retinoblastoma cells: immunohistochemical characterization of neuronal and glial markers. Exp Eye Res 1984; 39:207-15.

103. Terenghi G, Polak JM, Ballesta J, Cocchia D, Michetti F, Dahl D, Marangos PJ, Garner A. Immunocytochemistry of neuronal and glial markers in retinoblastoma. Virchows Arch A Pathol Anat Histopathol 1984; 404:61-73.

104. del Cerro M, Seigel GM, Lazar E, Grover D, del Cerro C, Brooks DH, DiLoreto D Jr, Chader G. Transplantation of Y79 cells into rat eyes: an in vivo model of human retinoblastomas. Invest Ophthalmol Vis Sci 1993; 34:3336-46.

105. Lee WH, Bookstein R, Hong F, Young LJ, Shew JY, Lee EY. Human retinoblastoma susceptibility gene: cloning, identification, and sequence. Science 1987; 235:1394-9.

106. Huang HJ, Yee JK, Shew JY, Chen PL, Bookstein R, Friedmann T, Lee EY, Lee WH. Suppression of the neoplastic phenotype by replacement of the RB gene in human cancer cells. Science 1988; 242:1563-6.

107. Xu HJ, Sumegi J, Hu SX, Banerjee A, Uzvolgyi E, Klein G, Benedict WF. Intraocular tumor formation of RB reconstituted retinoblastoma cells. Cancer Res 1991; 51:4481-5.

108. Muncaster MM, Cohen BL, Phillips RA, Gallie BL. Failure of RB1 to reverse the malignant phenotype of human tumor cell lines. Cancer Res 1992; 52:654-61.

109. Lee WH, Chen PL, Riley DJ. Regulatory networks of the retinoblastoma protein. Ann N Y Acad Sci 1995; 752:432-45.

110. Jiang Z, Zacksenhaus E, Gallie BL, Phillips RA. The retinoblastoma gene family is differentially expressed during embryogenesis. Oncogene 1997; 14:1789-97.

111. Lee EY, Chang CY, Hu N, Wang YC, Lai CC, Herrup K, Lee WH, Bradley A. Mice deficient for Rb are nonviable and show defects in neurogenesis and haematopoiesis. Nature 1992 359:288-94.

112. Kyritsis A, Tsokos M, Chader G. Attachment culture of human retinoblastoma cells: long-term culture conditions and effects of dibutyryl cyclic AMP. Exp Eye Res 1984; 38:411-21.

113. Kato G, Wakabayashi K. Effect of polylysine-bound laminin on human retinoblastoma cell lines. In Vitro Cell Dev Biol 1988; 24:274-80.

114. Kyritsis AP, Wiggert B, Lee L, Chader GJ. Butyrate enhances the synthesis of interphotoreceptor retinoid-binding protein (IRBP) by Y-79 human retinoblastoma cells. J Cell Physiol 1985; 124:233-9.

115. Tsokos M, Kyritsis AP, Chader GJ, Triche TJ. Differentiation of human retinoblastoma in vitro into cell types with characteristics observed in embryonal or mature retina. Am J Pathol 1986; 123:542-52.

116. Herman MM, Perentes E, Katsetos CD, Darcel F, Farnkfurter A, Collins VP, Donoso LA, Eng LF, Marangos PJ, Weichmann AF, et al. Neuroblastic differentiation potential of the human retinoblastoma cell lines Y-79 and WERI-Rb1 maintained in an organ culture system. An immunohistochemical, electron microscopic, and biochemical study. Am J Pathol 1989; 134:115-32.

117. Kyritsis A, Joseph G, Chader GJ. Effects of butyrate, retinol, and retinoic acid on human Y-79 retinoblastoma cells growing in monolayer cultures. J Natl Cancer Inst 1984; 73:649-54.

118. Kyritsis AP, Tsokos M, Chader GJ. Control of retinoblastoma cell growth by differentiating agents: current work and future directions. Anticancer Res 1986; 6:465-74.

119. Seigel GM, Notter MF. Differentiation of Y79 retinoblastoma cells induced by succinylated concanavalin A. Cell Growth Differ 1993; 4:1-7.

120. Seigel GM, Tombran-Tink J, Becerra SP, Chader GJ, Diloreto DA Jr, del Cerro C, Lazar ES, del Cerro M. Differentiation of Y79 retinoblastoma cells with pigment epithelial-derived factor and interphotoreceptor matrix wash: effects on tumorigenicity. Growth Factors 1994; 10:289-97.

121. del Cerro M, Notter MF, Seigel G, Lazar E, Chader G, del Cerro C. Intraretinal xenografts of differentiated human retinoblastoma cells integrate with the host retina. Brain Res 1992; 583:12-22.

122. Inomata M, Kaneko A. Chemosensitivity profiles of primary and cultured human retinoblastoma cells in a human tumor clongenic assay. Jpn J Cancer Res 1987; 78:858-68.

123. Vento R, Giuliano M, Lauricella M, Carabillo M, Di Liberto D, Tesoriere G. Induction of programmed cell death in human retinoblastoma Y79 cells by C2-ceramide. Mol Cell Biochem 1998; 185:7-15.

124. Di Felice V, Lauricella M, Giuliano M, Emanuele S, Vento R, Tesoriere G. The apoptotic effects of cisplatin and carboplatin in retinoblastoma Y79 cells. Int J Oncol 1998; 13:225-32.

125. Lauricella M, Giuliano M, Emanuele S, Vento R, Tesoriere G. Apoptotic effects of different drugs on cultured retinoblastoma Y79 cells. Tumour Biol 1998; 19:356-63.

126. Kondo Y, Kondo S, Liu J, Haqqi T, Barnett GH, Barna BP. Involvement of p53 and WAF1/CIP1 in gamma-irradiation-induced apoptosis of retinoblastoma cells. Exp Cell Res 1997; 236:51-6.

127. Schmidt-Erfurth U, Diddens H, Birngruber R, Hasan T. Photodynamic targeting of human retinoblastoma cells using covalent low-desity lipoprotein conjugates. Br J Cancer 1997; 75:54-61.

128. Nicolo M, Chiocca EA. Marker gene transfer and oncolysis of human Y79 retinoblastoma cells mediated by herpes simplex virus mutants. Ophthalmic Res 1998; 30:30-6.

129. Bernstein SL, Kutty G, Wiggert B, Albert DM, Nickerson JM. Expression of retina-specific genes by mouse retinoblastoma cells. Invest Ophthamol Vis Sci 1994; 35:3931-7.

130. Nishida T, Mukai N, Solish SP, Pomeroy M. Effects of cyclic AMP on growth and differentiation of rat retinoblastoma-like tumor cells in vitro. Invest Ophthalmol Vis Sci 1982; 22:145-56.

131. McLaren MJ, Sasabe T, Li CY, Brown ME, Inana G. Spontaneously arising immortal cell line of rat retinal pigmented epithelial cells. Exp Cell Res 1993; 204:311-20.

132. Davis AA, Bernstein PS, Bok D, Turner J, Nachtigal M, Hunt RC. A human retinal pigment epithelial cell line that retains epithelial characteristics after prolonged culture. Invest Ophthalmol Vis Sci 1995; 36:955-64.

133. Dunn KC, Aotaki-Keen AE, Putkey FR, Hjelmeland LM. ARPE-19, a human retinal pigment epithelial cell line with differentiated properties. Exp Eye Res 1996; 62:155-69.

134. Sheedlo HJ, Wordinger RJ, Fan W, Turner JE. A transformed neonatal rat retinal pigment epithelial cell line: secreted protein analysis and fibroblast growth factor and receptor expression. Curr Eye Res 1997; 16:116-26.

135. Gallimore PH, Grand RJ, Byrd PJ. Transformation of human embryo retinoblasts with simian virus 40, adenovirus and ras oncogenes. Anticancer Res 1986; 6:499-508.

136. Byrd PJ, Grand RJ, Gallimore PH. Differential transformation of primary human embryo retinal cells by adenovirus E1 regions and combinations of E1A + ras. Oncogene 1988; 2:477-84.

137. Grabham PW, Grand RJ, Byrd PJ, Gallimore PH. Differentiation of normal and adenovirus-12 E1 transformed human embryo retinal cells. Exp Eye Res 1988; 47:123-33.

138. Dutt K, Scott M, Wang M, Semple E, Sharma GP, Srinivasan A. Establishment of a human retinal cell line by transfection of SV40 T antigen gene with potential to undergo neuronal differentiation. DNA Cell Biol 1994; 13:909-21.

139. Dutt K, Ezeonu I, Scott M, Semple E, Srinivasan A. Proto-oncogene expression in cAMP and TPA-mediated neuronal differentiation in a human retinal cell line KGLDMSM. Curr Eye Res 1996; 15:477-85.

140. Sarthy VP, Brodjian SJ, Dutt K, Kennedy BN, French RP, Crabb JW. Establishment and characterization of a retinal Muller cell line. Invest Ophthamol Vis Sci 1998; 39:212-6.

141. Roque RS, Agarwal N, Wordinger RJ, Brun AM, Xue Y, Huang LC, Nguyen LP, Shay JW. Human papillomavirus-16 E6/E7 transfected retinal cell line expresses the Muller cell phenotype. Exp Eye Res 1997; 64:519-27.

142. Pessac B, Calothy G. Transformation of chick embryo neuroretinal cells by Rous sarcoma virus in vitro: induction of cell proliferation. Science 1974; 185:709-10.

143. Crisanti P, Pessac B, Calothy G. Avian neuroretina cells in oncogene studies. Methods Enzymol 1995; 254:77-97.

144. Crisanti-Combes P, Lorinet AM, Girard A, Pessac B, Wasseff M, Calothy G. Expression of neuronal markers in chick and quail embryo neuroretina cultures infected with Rous sarcoma virus. Cell Differ 1982; 11:45-54.

145. Pessac B, Girard A, Romey G, Crisanti P, Wassef M, Privat A, Calothy G. Cells with neuronal properties in permanent cultures of quail embryo neuroretinas infected with Rous sarcoma virus. Brain Res 1983; 275:53-9.

146. Turque N, Denhez F, Martin P, Planque N, Bailly M, Begue A, Stehelin D, Saule S. Characterization of a new melanocyte-specific gene (QNR-71) expressed in v-myc-transformed quail neuroretina. EMBO J 1996; 15:3338-50.

147. Pollerberg GE, Kuschel C, Zenke M. Generation of cell lines from embryonic quail retina capable of mature neuronal differentiation. J Neurosci Res 1995; 41:427-42.

148. Seigel GM, Notter MF. Loss of transformed phenotype upon senescence of Rous sarcoma virus-infected chicken neuroretinal cells. J Virol 1992; 66:6242-7.

149. Seigel GM. Establishment of an E1A-immortalized retinal cell culture. In Vitro Cell Dev Biol Anim 1996; 32:66-8.

150. Hammang JP, Baetge EE, Behringer RR, Brinster RL, Palmiter RD, Messing A. Immortalized retinal neurons derived from SV40 T-antigen-induced tumors in transgenic mice. Neuron 1990; 4:775-82.

151. Howes KA, Lasudry JG, Albert DM, Windle JJ. Photoreceptor cell tumors in transgenic mice. Invest Ophthalmol Vis Sci 1994; 35:342-51.

152. Seigel GM, Takahashi M, Adamus G, McDaniel T. Intraocular transplantation of E1A-immortalized retinal precursor cells. Cell Transplant 1998; 7:559-66.

153. Tezel G, Seigel GM, Wax MB. Autoantibodies to small heat shock proteins in glaucoma. Invest Ophthalmol Vis Sci 1998; 39:2277-87.

154. Seigel GM, Liu L. Inducible apoptosis-promoting activity in retinal cell-conditioned medium. Mol Vis 1997; 3:14 <http://www.molvis.org/molvis/v3/p14/>.

155. Adamus G, Machnicki M, Seigel GM. Apoptotic retinal cell death induced by anti-recoverin autoantibodies of cancer-associated retinopathy. Invest Ophthalmol Vis Sci 1997; 38:283-91.

156. Seigel GM, Mutchler AL, Adamus G, Imperato-Kalmar EL. Recoverin expression in the R28 retinal precursor cell line. In Vitro Cell Dev Biol Anim 1997; 33:499-502.

157. Seigel GM, Mutchler AL, Imperato EL. Expression of glial markers in a retinal precursor cell line. Mol Vis 1996; 2:2 <http://www.molvis.org/molvis/v2/p2/>.


Seigel, Mol Vis 1999; 5:4 <http://www.molvis.org/molvis/v5/p4/>
©1999 Molecular Vision <http://www.molvis.org/molvis/>
ISSN 1090-0535