Investigative Ophthalmology & Visual Science Cover Image for Volume 44, Issue 8
August 2003
Volume 44, Issue 8
Free
Retinal Cell Biology  |   August 2003
Novel Role for a Complement Regulatory Protein (CD46) in Retinal Pigment Epithelial Adhesion
Author Affiliations
  • Barbara J. McLaughlin
    From the Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky; and the
  • Wei Fan
    From the Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky; and the
  • Jing Juan Zheng
    From the Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky; and the
  • Hui Cai
    Department of Ophthalmology, Columbia University, New York, New York.
  • Lucian V. Del Priore
    Department of Ophthalmology, Columbia University, New York, New York.
  • Nalini S. Bora
    From the Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky; and the
  • Henry J. Kaplan
    From the Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, Kentucky; and the
Investigative Ophthalmology & Visual Science August 2003, Vol.44, 3669-3674. doi:https://doi.org/10.1167/iovs.02-0813
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Barbara J. McLaughlin, Wei Fan, Jing Juan Zheng, Hui Cai, Lucian V. Del Priore, Nalini S. Bora, Henry J. Kaplan; Novel Role for a Complement Regulatory Protein (CD46) in Retinal Pigment Epithelial Adhesion. Invest. Ophthalmol. Vis. Sci. 2003;44(8):3669-3674. https://doi.org/10.1167/iovs.02-0813.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

purpose. There is increasing evidence that the complement system may play a significant role in one of the leading diseases causing blindness in the elderly population, age-related macular degeneration. In this study, a novel role in the retina for a regulatory protein in the complement system, CD46, is proposed.

methods. The retinal pigment epithelium (RPE) was obtained from human donor eyes as well as human immortalized RPE cell lines (ARPE19). Immunohistochemistry and confocal microscopy were used to immunolocalize CD46 and β1 integrin. Immunoprecipitation experiments with antibodies to either CD46 or β1 integrin were performed on RPE cell lysates. A cell adhesion assay was used to determine the proportion of RPE cells that adhere to Bruch’s membrane explants from donor eyes.

results. Immunohistochemistry and confocal microscopy demonstrated that CD46 was polarized to the basal surface of the RPE along with β1 integrin, shown previously to be involved in RPE adhesion. Immunoprecipitation experiments demonstrated that CD46 and β1 integrin coprecipitated from RPE cell lysates when either protein was used as the precipitating antibody. The adhesion assay showed that antibodies to either CD46 or β1 integrin reduced RPE adhesion to the surface of Bruch’s membrane compared with the control.

conclusions. These findings suggest that this complement regulatory protein, which protects host cells from autologous complement attack, may have a functional interaction with β1 integrin in the eye that is related to RPE adhesion to its basement membrane and Bruch’s membrane.

Evidence is growing that the complement system may play a significant but as yet undefined role in age-related macular degeneration (AMD), the leading cause of blindness in the elderly population. One of the hallmarks of this disease is the formation of extracellular deposits or drusen between the retinal pigment epithelium (RPE), its basement membrane, and the remaining Bruch’s membrane. The disease progression leads to RPE dysfunction, detachment, and, eventually, degeneration that adversely affects the sensory photoreceptors and results in visual loss. Some investigators have suggested on the basis of immunolocalization of terminal complement complexes in drusen that their formation involves complement activation and that a dysfunctional condition in the RPE is an initiating event in AMD. 1 Although some components of the complement system are present in the retina 2 and RPE, 3 very little is known about their role, other than that they serve a protective function in innate immunity. The purpose of the present study was to explore the role of one complement regulatory protein, CD46, in RPE cell adhesion. 
CD46 (membrane cofactor protein [MCP]) acts as a serum protease cofactor that degrades C3b and prevents activation of the complement cascade that serves to protect the host cell against autologous attack. 4 5 6 It is a transmembrane glycoprotein that is present on most nucleated cells, and it serves as a receptor for measles virus, as well as C3b, C4b, and two other human pathogens. 7 8 9 10 11 12 In addition, it is polarized on the basolateral membrane of epithelial cells from nonocular tissue 13 14 15 16 and is highly expressed at the blood–brain barrier. 17 In this study, CD46 was also preferentially localized to the basolateral membrane surface of the RPE in situ, along with β1 integrin. In addition, CD46 coprecipitated with β1 integrin, which has been shown to mediate RPE attachment to the basement membrane. 18 Function-blocking experiments with antibodies demonstrated that RPE adhesion to Bruch’s membrane extracts can be reduced by anti-CD46 antibodies. Although it has been shown that CD46 associates directly with multiple β1 integrins in nonocular tissue, 19 this is the first report in ocular tissue of a functional interaction between a complement regulatory protein and a β1 integrin, which may be significant not only in adhesive mechanisms in the retina but in a constellation of functional interactions associated with integrin signaling pathways. Furthermore, by studying the role of this protein in normal RPE, we may gain new insights into the relationship of the complement system and complement regulatory proteins to the dysfunctional RPE observed in AMD. 
Materials and Methods
Immunohistochemistry
All research on tissue obtained from human subjects adhered to the tenets of the Declaration of Helsinki. Posterior globes of donor eyes (ages, 54–74 years) obtained from the Kentucky Lions Eye Bank were prepared for immunohistochemistry by fixing in 4% paraformaldehyde overnight, dehydrating, and embedding in paraffin. Consecutive 5-μm sections were hydrated, incubated for 2 hours with 0.3% H2O2 in phosphate-buffered saline to quench endogenous peroxidase followed by incubation with 5% goat normal serum. The sections were then incubated with 1:100 mouse anti-human CD46 antibody (BD PharMingen, San Diego, CA) and washed and incubated with biotinylated goat anti-mouse antibody and a buffered aqueous solution (ExtrAvidin-HRP; Sigma-Aldrich, St. Louis, MO) and a stain (NovaRed Substrate Kit for Peroxidase; Vector Laboratories, Burlingame, CA) that produces a red color in contrast to the gold-brown of the melanin granules in the RPE. Control sections were incubated either with isotype-matched nonimmune serum or without the primary antibody. 
The RPE-choroid from one donor eye was prepared as a flatmount for confocal microscopy and in situ staining with CD46 antibody (as above) and visualized by incubation with a Cy3-conjugated goat anti-mouse antibody (Sigma-Aldrich). RPE cell nuclei were counterstained with 4′,6′-diamino-2-phenylindole (DAPI; Vector Laboratories). 
RPE cells, harvested from donor eyes and maintained as primary cultures in DMEM/F12 with 10% fetal bovine serum (FBS), were also prepared for immunohistochemistry and confocal microscopy by fixing in 4% paraformaldehyde for 20 minutes. Cultured RPE were immunostained for CD46 as just described and, in addition, for the β1 integrin antibody (Chemicon International, Temecula, CA) and then visualized by a Cy3-labeled antibody. 
In addition, ARPE19 cells (ATCC, Manassas, VA) derived from human RPE were cultured on chamber slides (Nalge Nunc International, Naperville, IL) in DMEM-F12 containing 10% FBS with 100U/mL penicillin and 100 μg/mL streptomycin and maintained in culture conditions for 4 weeks before preparing for CD46 and β1 integrin immunohistochemistry and confocal microscopy as described for cultured cells. 
Reverse Transcription–Polymerase Chain Reaction
ARPE19 monolayers maintained in culture for 4 weeks or confluent RPE cell cultures obtained from human eyes (ages, 35–65 years) and established after two to six passages were used to extract total RNA (RNeasy Mini Kit; Qiagen, Valencia, CA) according to the manufacturer’s specification. The yield and purity of RNA were estimated by optical density at 260/280 nm. After DNase treatment, cDNAs were synthesized from RNAs with reverse transcriptase (Superscript II; Invitrogen/Gibco, Gaithersburg, MD) with oligo dT as the primer, according to the manufacturer’s specifications. Polymerase chain reactions were performed in an automatic sequencer (Gene-Amp PCR System 2400; Applied Biosystems Inc., Foster City, CA) with advantage cDNA polymerase mix (Clontech, Palo Alto, CA). The following primer for CD46 was used: forward, 5′-CCT GCA AAT GGG ACT TAC G-3′; reverse, 5′-AAA AAC CCT TAT CGC ATT CAA AC-3′. 
PCR products were sequenced by DNA autosequencing (CEQ 2000; Beckman Instruments, Fullerton, CA) and the sequence identity verified by using a BLAST search of the Genome Systems Data Bank, available at http://www.ncbi.nlm.nih.gov/blast/. 
Immunoblot Analysis
To detect the expression of the complement regulatory protein CD46 in RPE, Western blot analysis was performed with RPE cells from donor eyes (ages, 60, 73, and 76 years) and ARPE19 cell lines. Posterior globes were frozen within 12 hours after death and were stored at −80°C, thawed, and dissected. RPE cells were collected by adding Hanks’ medium into the eyecups and gentle pipetting. Total protein extract of RPE cells (collected from donor eyes or ARPE19) was prepared with lysis buffer containing 50 mM Tris-HCl (pH 8.0), 1% Triton-X-100, 150 mM NaCl, 0.02% sodium azide, 10 μg/mL antipain, 10 μg/mL leupeptin, 10 μg/mL peptain A, 2 μg/mL aprotinin, and 100 μM phenylmethylsulfonyl fluoride (PMSF). Protein concentration was determined using a bicinchoninic acid (BCA Protein Assay Reagent Kit; Pierce, Rockford, IL). Fifty micrograms of each protein sample was separated by SDS-PAGE on 10% gels under nonreducing conditions. Proteins were then transferred to polyvinylidene difluoride (PVDF) membranes. The membranes were blocked overnight at 4°C in TBS-T solution containing 5% nonfat dry milk and incubated with mouse anti-human antibodies against CD46 (BD PharMingen International, San Diego, CA) in TBS-T solution. The antibody binding was detected with horseradish peroxidase-conjugated anti-mouse (Dako, Glostrup, Denmark) secondary antibody and enhanced chemiluminescence (ECL) Western blot detection reagents (Amersham Life Science, Buckinghamshire, UK). 
Immunoprecipitation
RPE cells harvested from human donor eyes or ARPE19 cells were washed with ice-cold PBS and lysed for 30 minutes at 4°C in nonionic detergent (Brij 98; Sigma-Aldrich; 20 mM Tris [pH 7.5], 150 mM NaCl, 1% Brij 98, 1 mM CaCl2, 1 mM MgCl2, 10 μg/mL antipain, 10 μg/mL leupeptin, 10 μg/mL peptain A, 2 μg/mL aprotinin, and 100 μM PMSF). The insoluble material was removed by centrifugation at 12,000g for 15 minutes at 4°C, and the supernatants were precleared by incubation with Sepharose protein G (Amersham Pharmacia Biotech) for 1 hour at 4°C. Precleared supernatants were centrifuged and transferred to new tubes and then incubated overnight at 4°C with either anti-CD46 rabbit polyclonal (Santa Cruz Biotechnology, Santa Cruz, CA) or anti-β1 integrin monoclonal (Chemicon International) precipitating antibody. The purified rabbit IgG or isotype-matched IgG served as the control. The immune complex was precipitated by adding 1:10 volume of Sepharose protein G and incubating at 4°C. After a 1-hour incubation under constant rotation, samples were centrifuged and washed five times with lysis buffer containing 0.5% Brij 98. Bound proteins were eluted with two times lysis buffer and boiling for 10 minutes. Immunoprecipitated proteins were resolved in a 7.5% SDS gel under nonreducing conditions and transferred to PVDF membranes. Coprecipitated β1 integrin or CD46 were probed either by a combination of biotin-labeled mAb (Ancell, Bayport, MN) plus horseradish peroxidase-conjugated streptavidin (Amersham Pharmacia Biotech) or a combination of unlabeled mouse mAb (BD PharMingen International) plus HRP-conjugated rabbit anti-mouse secondary antibody (Chemicon International). 
Cell Adhesion Assay
Explants of human Bruch’s membrane were prepared from human donor eyes (60–70-year-old donors), as described previously. 20 21 22 After a full-thickness circumferential incision was made posterior to the ora serrata and the vitreous and anterior segment removed, the posterior poles were inspected and discarded if there was any evidence of subretinal blood, drusen, or irregular pigmentation of the macular RPE. The neural retina was removed and 0.02 N ammonium hydroxide was pipetted into the eyecup to remove adherent RPE, followed by washing with PBS three times. A 6.5-mm diameter corneoscleral trephine was used to punch out explants of human Bruch’s membrane from the macula and periphery of the eyecups. Six to eight explants were harvested per eye. 
Second-passage human RPE cells were harvested from donor eyes (53–65-year-old donors) by incubating in 0.25% trypsin-0.25% edetic acid in Hanks’ balanced salt solution for 20 minutes. Ten milliliters of MEM/15 was added for quenching and the cell suspension was centrifuged for 5 minutes at 800 revolutions per minute. The cell pellet was incubated on a shaker table at room temperature for 1 hour in one of the following antibodies: 0.1 to 25 μg/mL of mouse anti-human CD46 monoclonal antibody (Accurate Chemical & Scientific Corp., New York, NY), 10 μg/mL of mouse anti-human β1 integrin monoclonal antibody (Chemicon International), or 1:500 mouse non-specific IgG1κ MOPC21 monoclonal antibody (Sigma-Aldrich). The cell pellet was washed three times and resuspended in MEM without serum, and aliquots of 5 × 104 viable cells were applied to each Bruch’s membrane explant. Cells were allowed to attach for 24 hours. Unattached cells were removed by picking up the tissue with fine forceps and dipping three times in Hanks’ balanced salt solution before placing the explants in a new well of a 96-well plate. Three explant buttons were used for each experimental condition. Data were analyzed by one-way analysis of variance followed by a multiple comparison test or a two-tailed t-test. 
Cell adhesion assays were performed using an established MTT-based cell assay, as described previously. 18 23 MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; Sigma-Aldrich) is a dye with characteristics that change when it is dehydrogenated by cellular mitochondrial dehydrogenase, and the activity of the latter enzyme is proportional to the number of live cells exposed to the dye. 20 The amount of yellow-reduced tetrazolium was quantified with an enzyme-linked immunosorbent assay reader with a 570-nm filter after the solid tissue was removed from the wells containing explants and 96-well plates read. The number of cells attached to the surface was calculated by comparing the enzyme-linked immunosorbent assay readings obtained on the wells with an unknown number of cells with a standardized curve obtained by using control wells containing up to 50,000 live cells as an internal standard for each experiment. The attachment proportion was defined and calculated as the proportion of cells that had attached to the surface during the experimental period. Cell adhesion experiments were performed in triplicate seven times. 
Results
Immunolocalization and Expression of CD46
In histologic sections of the normal human eye, CD46 staining showed a preferential distribution on the basolateral surface of the RPE, but not in Bruch’s membrane (Fig. 1) . Confocal microscopy of horizontal sections of RPE harvested directly from human donor eyes and prepared as a flatmount demonstrated a complete absence of CD46 staining on the apical RPE membranes in situ (Fig. 2A) , as well as an absence of autofluorescent pigment granules (Fig. 2B) . CD46 staining was present in sections of the basolateral RPE surface (Fig. 2C) , and some autofluorescence was present, due to pigment granules in the basal RPE cytoplasm (Fig. 2D) . Confocal microscopy of cultures immunolabeled for CD46 and β1 integrin also revealed a basolateral membrane localization of both proteins in horizontal and vertical views of the RPE monolayer in primary cultured RPE from donor eyes (Fig. 3) and in RPE cell lines (Fig. 4) . When the RPE monolayer was viewed at different confocal planes from apical to basolateral surfaces, immunolabeled CD46 and β1 integrin were present only on the basolateral membrane surfaces. When viewed in vertical sections, antibody staining for CD46 and β1 integrin clearly labeled the basal RPE surface. 
Immunoblot analysis of RPE obtained from donor eyes and the ARPE19 cell line demonstrated the presence of a protein doublet at 55 and 65 kDa, which corresponds to the lighter and heavier isoforms of CD46, respectively (Fig. 5) . Primary RPE cultures established from donor eyes and cultured ARPE19 cells grown to confluence and maintained for 4 weeks or longer demonstrated the presence of mRNA for CD46 (Fig. 6) . A PCR product was obtained in the expected base pair range of 448 bp for CD46, and sequencing confirmed a 99% sequence identity with its appropriate cDNA. 
Coimmunoprecipitation of CD46 and β1 Integrin in RPE Cells
To characterize the association of CD46 with β1 integrin, immunoprecipitation experiments were performed on cell lysates of RPE harvested from human donor eyes and the ARPE19 cell line and immunoblotted with antibodies to CD46 and β1 integrin. β1 integrin coprecipitated with CD46 from both human RPE (Fig. 7A) and RPE cell lines. Reciprocally, CD46 coimmunoprecipitated with β1 integrin in both human RPE (Fig. 7B) and ARPE19 cell lysates. Control samples incubated with purified rabbit IgG or isotype-matched IgG failed to precipitate either CD46 or β1 integrin. 
RPE Cell Adhesion Assay
We used the cell adhesion assay previously described in which RPE attachment to their native physiological substrate, the basal lamina of human Bruch’s membrane, is determined (Fig. 8) . RPE cells that were preincubated with an irrelevant IgG (mouse IgG1κ MOPC21 monoclonal antibody) before seeding onto Bruch’s membrane served as the control for all the adhesion studies. Anti-CD46 reduced RPE adhesion to human Bruch’s membrane by approximately 40% to 50% compared with the control in a dose-dependent manner (Fig. 8A) , with half-maximum inhibition at a concentration of approximately 1 μg/mL of anti-CD46. We have demonstrated that anti-β1 integrin antibodies decrease RPE adhesion to human Bruch’s membrane and RPE-derived extracellular matrix. 18 In the present study, 10 μg/mL of anti-β1 integrin, previously shown to be a saturating dose, produced inhibition comparable to anti-CD46 alone (Fig. 8B) . Incubation with both anti-CD46 and anti-β1 integrin did not produce further inhibition compared with either antibody alone (Fig. 8B)
Discussion
A complement regulatory protein, CD46, preferentially localized to the basolateral membrane of the RPE from human donor eyes in situ and in primary cultures of human RPE, as well as in ARPE19 cell lines. Immunoprecipitation experiments of RPE lysates from the same sources demonstrated that CD46 coprecipitated with β1 integrin and, in the reverse immunoprecipitation protocol, β1 integrin coprecipitated with CD46, indicating a physical relationship between the two proteins. Functional blocking of RPE adhesion with antibodies to CD46 confirms that anti-CD46 reduces RPE cell adhesion, similar to the effect that has been shown with anti-β1 integrin antibodies. 18 Incubation with antibodies to both CD46 and anti-β1 integrin inhibited RPE adhesion to the same extent as either antibody alone, thus suggesting that both antibodies affect the same site. 
CD46 (membrane cofactor protein [MCP]) is expressed on all nucleated human cells and acts to protect the host cell against autologous complement attack by degrading C3b. 24 25 It is also present on the basolateral surface of polarized epithelial cells, 13 14 15 16 similar to the localization shown in the current study in the RPE. Further studies of the basolateral targeting mechanism have shown that there is a functional interaction between CD46 and DLG4, a member of the guanylate kinase family and that the polarized expression of CD46 in epithelial cells requires the DLG4-binding domain. 16  
One of the proteins belonging to the DLG4 family may also have a functional interaction with CD46 in RPE cells. DLG4 is one of a family of four human proteins that share a single homologue with a tumor-suppressor called disc large (DLG) 26 27 28 from Drosophila. DLG4 is also called postsynaptic density (PSD) or synapse-associated protein (SAP) because of its localization in the postsynaptic density region of neurons. These proteins have multiple protein–protein interaction motifs, including three PDZ domains, which derive their acronym from the three proteins first characterized as having these recognition domains (PSD from neurons, DLG from Drosophila, ZO1 from tight junctions). These proteins are polarized and interact with other PDZ domain-containing proteins, membrane receptors, cell adhesion molecules, and the cytoskeleton to regulate epithelial cell polarization and assemble signaling cascades. 29 30 31 32 In view of this, it is of interest that recent studies of rat RPE have shown that one of these PDZ domain-containing proteins, SAP-97, localizes to the basolateral surface of RPE and may have a direct interaction with ezrin, 33 an actin-binding protein associated with the morphogenesis of RPE apical microvilli and basal infoldings. 34 Ezrin belongs to the ERM family of proteins, which collectively defines three highly homologous proteins (ezrin, radixin, moesin) that constitute a group of plasma membrane–cytoskeleton linkers that regulate cell adhesion and morphogenesis of the actin-rich cell cortex. 35 Future work in our laboratory will investigate the relationship of CD46 to ezrin and SAP-97 and whether there are any functional associations with RPE adhesion. 
Cell adhesion to the extracellular matrix is a crucial regulator of cell behavior and the large protein complexes of signaling proteins and cytoskeleton are assembled into functional units at the sites of integrin–matrix adhesion. 36  
We have shown that the β1 subunit of integrins partially mediate the adherence of human RPE cells to RPE-derived extracellular matrix and the basal lamina layer of human Bruch’s membrane. 18 Recently, it has been shown by immunoprecipitation experiments in carcinoma-derived cell lines that CD46 associates with multiple β1 integrins 19 and indirectly with a superfamily of surface molecules, 37 tetraspanin, known also to associate with a subset of β1 integrins 38 39 40 41 and to form a web with common functions related to migration, proliferation, intracellular signaling and adhesion. 42 43 44 45 46 CD46 is a newly discovered component of this web. 19 The relevance of these protein associations to RPE cells is that they may form the functional units underlying normal adhesion mechanisms, maintaining a healthy RPE phenotype that is not proliferative or migratory. When there is disease, these functional units may become disrupted, the RPE may lose attachment to Bruch’s membrane, and RPE cells may break away from the monolayer and undergo apoptosis. 47 Therefore, the loss of RPE cells, which is one of the first signs of AMD, 48 may be preceded by the loss of RPE attachment to Bruch’s membrane through a dysfunctional CD46–β1 integrin complex. A recent study, in which flatmount preparations of human cadaveric eyes were stained with the TUNEL technique, provides direct evidence that human RPE undergoes age-related apoptosis in situ, with apoptotic human RPE confined mainly to the macula of older human eyes. 49 Another similar report suggests that human RPE die by apoptosis around the edges of geographic atrophy. 50 Secondary atrophy of the underlying choriocapillaris and overlying photoreceptors would then follow and signal the clinical recognition of AMD. More studies are needed to determine how specific molecular interactions of integrin with CD46 and other protein partners play a role as a functional unit in maintaining RPE adhesion and phenotype. 
 
Figure 1.
 
Histologic section of a donor eye immunostained with anti-CD46 (A). Staining was present on the basolateral membrane surface of the RPE. Inset: higher magnification of basolateral staining. Control sections (B) showed little or no specific staining. Bar, 20 μm.
Figure 1.
 
Histologic section of a donor eye immunostained with anti-CD46 (A). Staining was present on the basolateral membrane surface of the RPE. Inset: higher magnification of basolateral staining. Control sections (B) showed little or no specific staining. Bar, 20 μm.
Figure 2.
 
Confocal microscopy of horizontal sections through RPE from a human donor eye, flatmounted and immunolabeled for CD46. (A, B) represent sections from the same apical RPE plane showing the absence of staining for CD46 (A), as well as the absence of autofluorescent pigment granules (B). (C, D) represent sections from the same basolateral RPE plane showing membrane staining for CD46 (C) and autofluorescence due to pigment granules in the basal RPE cytoplasm (D). Bar, 20 μm.
Figure 2.
 
Confocal microscopy of horizontal sections through RPE from a human donor eye, flatmounted and immunolabeled for CD46. (A, B) represent sections from the same apical RPE plane showing the absence of staining for CD46 (A), as well as the absence of autofluorescent pigment granules (B). (C, D) represent sections from the same basolateral RPE plane showing membrane staining for CD46 (C) and autofluorescence due to pigment granules in the basal RPE cytoplasm (D). Bar, 20 μm.
Figure 3.
 
Confocal microscopy of horizontal sections through the RPE monolayer from primary cultures showing the absence of labeling for CD46 and β1 integrin on the apical plane of the section and the presence of basolateral membrane staining only in the basal plane of the monolayer. Polarized labeling on the basal RPE surface was clearly visible in vertical sections. Bar, 20 μm.
Figure 3.
 
Confocal microscopy of horizontal sections through the RPE monolayer from primary cultures showing the absence of labeling for CD46 and β1 integrin on the apical plane of the section and the presence of basolateral membrane staining only in the basal plane of the monolayer. Polarized labeling on the basal RPE surface was clearly visible in vertical sections. Bar, 20 μm.
Figure 4.
 
Confocal microscopy of horizontal sections through the ARPE19 cell monolayer showing the same absence of apical labeling and intense basolateral labeling for both CD46 and β1 integrin in both horizontal and vertical sections. Bar, 20 μm.
Figure 4.
 
Confocal microscopy of horizontal sections through the ARPE19 cell monolayer showing the same absence of apical labeling and intense basolateral labeling for both CD46 and β1 integrin in both horizontal and vertical sections. Bar, 20 μm.
Figure 5.
 
Immunoblots of RPE from donor eyes and ARPE19 cell lines labeled with antibodies to CD46. Two bands are present in each lane: a dense upper band at 65 kDa and a lighter band at 55kDa, which correspond to the heavier and lighter isoforms of CD46, respectively.
Figure 5.
 
Immunoblots of RPE from donor eyes and ARPE19 cell lines labeled with antibodies to CD46. Two bands are present in each lane: a dense upper band at 65 kDa and a lighter band at 55kDa, which correspond to the heavier and lighter isoforms of CD46, respectively.
Figure 6.
 
RT-PCR of RPE from donor eyes and the ARPE19 cell line. Lanes 14: controls without reverse transcriptase; lanes 57: PCR products of RPE harvested from donor eyes; lane 8: PCR product from ARPE19 cells, all within the expected base pair range (448 bp) for CD46.
Figure 6.
 
RT-PCR of RPE from donor eyes and the ARPE19 cell line. Lanes 14: controls without reverse transcriptase; lanes 57: PCR products of RPE harvested from donor eyes; lane 8: PCR product from ARPE19 cells, all within the expected base pair range (448 bp) for CD46.
Figure 7.
 
CD46 coimmunoprecipitated β1 integrin from RPE cell lysates from donor eyes. (A) Immunoprecipitated proteins when anti-CD46 was used as the precipitating antibody. A dense band of the CD46 protein was labeled with anti-CD46 at the expected range of 55 to 65 kDa and a lighter band at the expected 130 kDa was labeled with anti-β1 integrin. (B) When anti-β1 integrin was used as the immunoprecipitating antibody, a dense band was present at 130 kDa that stained with anti-β1 integrin and a fainter band at 60 to 65 kDa that stained with anti-CD46.
Figure 7.
 
CD46 coimmunoprecipitated β1 integrin from RPE cell lysates from donor eyes. (A) Immunoprecipitated proteins when anti-CD46 was used as the precipitating antibody. A dense band of the CD46 protein was labeled with anti-CD46 at the expected range of 55 to 65 kDa and a lighter band at the expected 130 kDa was labeled with anti-β1 integrin. (B) When anti-β1 integrin was used as the immunoprecipitating antibody, a dense band was present at 130 kDa that stained with anti-β1 integrin and a fainter band at 60 to 65 kDa that stained with anti-CD46.
Figure 8.
 
Cell adhesion assay. (A) Inhibition of RPE attachment by anti-CD46 was dose dependent. RPE cells were incubated with various concentrations of anti-CD46 antibodies before seeding onto the basal lamina layer of human Bruch’s membrane. Anti-CD46 inhibited approximately 40% to 50% of cell attachment, with half-maximum inhibition at a concentration of 1 μg/mL. (B) Anti-β1 integrin (10 μg/mL) produced inhibition similar to that of anti-CD46 (10 μg/mL). Incubation with both antibodies did not cause any additional effect. Control RPE cells were incubated with a nonimmune IgG (mouse IgG1κ MOPC21 monoclonal antibody) to the RPE before seeding onto Bruch’s membrane.
Figure 8.
 
Cell adhesion assay. (A) Inhibition of RPE attachment by anti-CD46 was dose dependent. RPE cells were incubated with various concentrations of anti-CD46 antibodies before seeding onto the basal lamina layer of human Bruch’s membrane. Anti-CD46 inhibited approximately 40% to 50% of cell attachment, with half-maximum inhibition at a concentration of 1 μg/mL. (B) Anti-β1 integrin (10 μg/mL) produced inhibition similar to that of anti-CD46 (10 μg/mL). Incubation with both antibodies did not cause any additional effect. Control RPE cells were incubated with a nonimmune IgG (mouse IgG1κ MOPC21 monoclonal antibody) to the RPE before seeding onto Bruch’s membrane.
The authors thank Gaby Enzmann and Puran Bora for their discussions regarding the immunohistochemistry and Guangyu Li for designing primers and sequence analysis of PCR products. 
Johnson, LV, Leitner, WP, Staples, MK, Anderson, DH. (2001) Complement activation and inflammatory processes in drusen formation and age related macular degeneration Exp Eye Res 73,887-896 [CrossRef] [PubMed]
Bora, NS, Gobleman, CL, Atkinson, JP, Kaplan, HJ. (1993) Differential expression of the complement regulatory proteins in the human eye Invest Ophthalmol Vis Sci 34,3576-3584
Elner, VM, Schaffer, T, Taylor, K, Glagov, S. (1981) Immunophagocytic properties of retinal pigment epithelium cells Science 211,74-76 [CrossRef] [PubMed]
Seya, T, Atkinson, JP. (1989) Functional properties of membrane cofactor protein of complement Biochem J 264,581-588 [PubMed]
Liszewski, MK, Farries, TC, Lublin, DM, Rooney, IA, Atkinson, JP. (1996) Control of the complement system Adv Immunol 61,201-283 [PubMed]
Liszewski, MK, Post, TW, Atkinson, JP. (1991) Membrane cofactor protein (MCP or CD46): newest member of the regulators of complement activation gene cluster Annu Rev Immunol 9,431-455 [CrossRef] [PubMed]
Dorig, RE, Marcil, A, Chopra, A, Richardson, CD. (1993) The human CD46 molecule is a receptor for measles virus (Edmonston strain) Cell 75,295-305 [CrossRef] [PubMed]
Naniche, DG, Varior-Krishnan, G, Cervoni, F, et al (1993) Human membrane cofactor protein (CD46) acts as a cellular receptor for measles virus J Virol 67,6025-6032 [PubMed]
Manchester, M, Liszewski, MK, Atkinson, JP, Oldstone, MBA. (1994) Multiple isoforms of CD46 (membrane cofactor protein) serve as receptors for measles virus Proc Natl Acad Sci USA 91,2161-2165 [CrossRef] [PubMed]
Dorig, RE, Marcil, A, Richardson, CD. (1994) CD46, a primate-specific receptor for measles virus Trends Microbiol 2,312-318 [CrossRef] [PubMed]
Okada, N, Liszewski, MK, Atkinson, JP, Caparon, M. (1995) Membrane cofactor protein (MCP; CD46) is a keratinocyte receptor for the M protein of group A streptococcus Proc Natl Acad Sci USA 92,2489-2493 [CrossRef] [PubMed]
Kallstrom, H, Liszewski, MK, Atkinson, JP, Jonsson, A-B. (1997) Membrane cofactor protein (MCP; CD46) is a cellular pilus receptor for pathogenic Neisseria Mol Microbiol 25,639-647 [CrossRef] [PubMed]
Maisner, A, Liszewski, MK, Atkinson, JP, Schwartz-Albiez, R, Herrler, G. (1996) Two different cytoplasmic tails direct isoforms of the membrane cofactor protein (CD46) to the basolateral surface of Madin-Darby canine kidney cells J Biol Chem 271,18853-18858 [CrossRef] [PubMed]
Maisner, A, Zimmer, G, Liszewski, MK, Lublin, DM, Atkinson, JP, Herrler, G. (1997) Membrane cofactor protein (CD46) is a basolateral protein that is not endocytosed: importance of the tetrapeptide FTSL at the carboxyl terminus J Biol Chem 272,20793-20799 [CrossRef] [PubMed]
Teuchert, M, Maisner, A, Herrler, G. (1999) Importance of the carboxyl-terminal FTSL motif of membrane cofactor protein for basolateral sorting and endocytosis: positive and negative modulation by signals inside and outside the cytoplasmic tail J Biol Chem 274,19979-19984 [CrossRef] [PubMed]
Ludford-Menting, MJ, Thomas, SJ, Crimeen, B, et al (2002) A functional interaction between CD46 and DLG4: a role for DLG4 in epithelial polarization J Biol Chem 277,4477-4484 [CrossRef] [PubMed]
Shusta, EV, Zhu, C, Boado, RK, Pardridge, WM. (2002) Subtractive expression cloning reveals high expression of CD46 at the blood-brain barrier J Neuropathol Exp Neurol 61,597-604 [PubMed]
Ho, TZ, Del Priore, LV. (1997) Reattachment of cultured human retinal pigment epithelium to extracellular matrix and human Bruch’s membrane Invest Ophthalmol Vis Sci 38,1110-1118 [PubMed]
Lozahic, S, Christiansen, D, Manie, S, Gerlier, D, Billard, M, Boucheix, C, Rubinstein, E. (2000) CD46 (membrane cofactor protein) associates with multiple beta1 integrins and tetraspans Eur J Immunol 30,900-907 [CrossRef] [PubMed]
Tezel, TH, Del Priore, LV, Kaplan, HJ. (1999) Fate of human retinal pigment epithelial cells seeded onto layers of human Bruch’s membrane Invest Ophthalmol Vis Sci 40,467-476 [PubMed]
Tezel, TH, Del Priore, LV. (1999) Repopulation of different layers of host human Bruch’s membrane by retinal pigment epithelial cell grafts Invest Ophthalmol Vis Sci 40,767-774 [PubMed]
Del Priore, LV, Tezel, TH. (1998) Reattachment rate of human retinal pigment epithelium to layers of human Bruch’s membrane Arch Ophthalmol 116,335-341 [CrossRef] [PubMed]
Mossmann, T. (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays J Immunol Methods 65,55-63 [CrossRef] [PubMed]
Liszewski, MK, Atkinson, JP. (1992) Membrane cofactor protein Curr Top Microbiol Immunol 178,45-60 [PubMed]
Seya, T, Hirano, A, Matsumoto, M, Nomura, M, Ueda, S. (1999) Human membrane cofactor protein (MCP, CD46): multiple isoforms and functions Int J Biochem Cell Biol 31,1255-1260 [CrossRef] [PubMed]
Woods, DF, Bryant, PJ. (1991) The discs-large tumor suppressor gene of Drosophila encodes a guanylate kinase homolog localized at septate junctions Cell 66,451-464 [CrossRef] [PubMed]
Fujita, A, Kurachi, Y. (2000) SAP family proteins Biochem Biophys Res Commun 269,1-6 [CrossRef] [PubMed]
Dimitratos, SD, Woods, DF, Stathakis, DG, Bryant, PJ. (1999) Signaling pathways are focused at specialized regions of the plasma membrane by scaffolding proteins of the MAGUK family Bioessays 21,912-921 [CrossRef] [PubMed]
Kim, SK. (1997) Polarized signaling: basolateral receptor localization in epithelial cells by PDZ-containing proteins Curr Opin Cell Biol 9,853-859 [CrossRef] [PubMed]
Fanning, AS, Anderson, JM. (1999) PDZ domains: fundamental building blocks in the organization of protein complexes at the plasma membrane J Clin Invest 103,767-772 [CrossRef] [PubMed]
Fanning, AS, Anderson, JM. (1999) Protein modules as organizers of membrane structure Curr Opin Cell Biol 11,432-439 [CrossRef] [PubMed]
Garner, CC, Nash, J, Huganir, RL. (2000) PDZ domains in synapse assembly and signalling Trends Cell Biol 10,274-280 [CrossRef] [PubMed]
Bonilha, VL, Rodriguez-Boulan, E. (2001) Polarity and developmental regulation of two PDZ proteins in the retinal pigment epithelium Invest Ophthalmol Vis Sci 42,3274-3282 [PubMed]
Bonilha, VL, Finnemann, SC, Rodriguez-Boulan, E. (1999) Ezrin promotes morphogenesis of apical microvilli and basal infoldings in retinal pigment epithelium J Cell Biol 147,1533-1548 [CrossRef] [PubMed]
Mangeat, P, Roy, C, Martin, M. (1999) ERM proteins in cell adhesion and membrane dynamics Trends Cell Biol 9,187-192 [CrossRef] [PubMed]
Schwartz, MA. (2001) Integrin signaling revisited Trends Cell Biol 11,466-470 [CrossRef] [PubMed]
Maecker, HT, Todd, SC, Levy, S. (1997) The tetraspanin superfamily: molecular facilitators FASEB J 11,428-442 [PubMed]
Rubinstein, E, LeNaour, F, Billard, M, Prenant, M, Boucheix, C. (1994) CD9 antigen is an accessory subunit of the VLA integrin complexes Eur J Immunol 24,3005-3013 [CrossRef] [PubMed]
Rubinstein, E, LeNaour, F, Lagaudriere, C, Billard, M, Conjeaud, H, Bousheix, C. (1996) CD9, CD63, CD81, and CD82 are components of a surface tetraspan network connected to HLA-DR and VLA integrins Eur J Immunol 26,2657-2665 [CrossRef] [PubMed]
Berditchevski, F, Zutter, MM, Hemler, ME. (1996) Characterization of novel complexes on the cell surface between integrins and proteins with four transmembrane domains (TM4 proteins) Mol Biol Cell 7,193-207 [CrossRef] [PubMed]
Radford, K J, Thorne, RF, Hersey, P. (1996) CD63 associates with transmembrane 4 superfamily members, CD9 and CD81, and with beta 1 integrins in human melanoma Biochem Biophys Res Commun 222,13-18 [CrossRef] [PubMed]
Ikeyama, S, Koyama, M, Yamaoko, M, Sasada, R, Miyake, M. (1993) Suppression of cell motility and metastasis by transfection with human motility-related protein (MRP-1/CD9) DNA J Exp Med 177,1231-1237 [CrossRef] [PubMed]
Radford, KJ, Thorne, RF, Hersey, P. (1997) Regulation of tumor cell motility and migration by CD63 in a human melanoma cell line J Immunol 158,3353-3358 [PubMed]
Dong, JT, Suzuki, H, Pin, SS, et al (1996) Down-regulation of the KAI1 metastasis suppressor gene during the progression of human prostatic cancer infrequently involves gene mutation or allelic loss Cancer Res 56,4387-4390 [PubMed]
Lagaudriere-Gesbert, C, LeNaour, F, Lebel-Binay, S, et al (1997) Functional analysis of four tetraspans, CD9, CD53, CD81, and CD82, suggests a common role in costimulation, cell adhesion, and migration: only CD9 upregulates HB-EGF activity Cell Immunol 182,105-112 [CrossRef] [PubMed]
Horvath, G, Serru, V, Clay, D, Billard, M, Boucheix, C, Rubinstein, E. (1998) CD19 is linked to the integrin-associated tetraspans CD9, CD81, and CD82 J Biol Chem 273,30537-30543 [CrossRef] [PubMed]
Tezel, TH, Del Priore, LV, Kaplan, HJ. (1997) Reattachment to a substrate prevents apoptosis of human retinal pigment epithelium Graefes Arch Clin Exp Ophthalmol 235,41-47 [CrossRef] [PubMed]
Del Priore, LV, Tezel, TH, Ho, TC, Kaplan, HJ. (1997) Capone, A De Juan, E Del Priore, L Panozzo, G Zarbin, M eds. New Developments in the Treatment of Age-Related Macular Degeneration ,145-151 Progei Editori Verona, Italy.
Del Priore, LV, Kuo, YH, Tezel, TH. (2002) Age-Related changes in human RPE cell density and apoptosis proportion in situ Invest Ophthalmol Vis Sci 43,3312-3318 [PubMed]
Dunaief, JL, Dentchev, T, Ying, GS, Milam, AH. (2002) The role of apoptosis in age-related macular degeneration Arch Ophthalmol 120,1435-1442 [CrossRef] [PubMed]
Figure 1.
 
Histologic section of a donor eye immunostained with anti-CD46 (A). Staining was present on the basolateral membrane surface of the RPE. Inset: higher magnification of basolateral staining. Control sections (B) showed little or no specific staining. Bar, 20 μm.
Figure 1.
 
Histologic section of a donor eye immunostained with anti-CD46 (A). Staining was present on the basolateral membrane surface of the RPE. Inset: higher magnification of basolateral staining. Control sections (B) showed little or no specific staining. Bar, 20 μm.
Figure 2.
 
Confocal microscopy of horizontal sections through RPE from a human donor eye, flatmounted and immunolabeled for CD46. (A, B) represent sections from the same apical RPE plane showing the absence of staining for CD46 (A), as well as the absence of autofluorescent pigment granules (B). (C, D) represent sections from the same basolateral RPE plane showing membrane staining for CD46 (C) and autofluorescence due to pigment granules in the basal RPE cytoplasm (D). Bar, 20 μm.
Figure 2.
 
Confocal microscopy of horizontal sections through RPE from a human donor eye, flatmounted and immunolabeled for CD46. (A, B) represent sections from the same apical RPE plane showing the absence of staining for CD46 (A), as well as the absence of autofluorescent pigment granules (B). (C, D) represent sections from the same basolateral RPE plane showing membrane staining for CD46 (C) and autofluorescence due to pigment granules in the basal RPE cytoplasm (D). Bar, 20 μm.
Figure 3.
 
Confocal microscopy of horizontal sections through the RPE monolayer from primary cultures showing the absence of labeling for CD46 and β1 integrin on the apical plane of the section and the presence of basolateral membrane staining only in the basal plane of the monolayer. Polarized labeling on the basal RPE surface was clearly visible in vertical sections. Bar, 20 μm.
Figure 3.
 
Confocal microscopy of horizontal sections through the RPE monolayer from primary cultures showing the absence of labeling for CD46 and β1 integrin on the apical plane of the section and the presence of basolateral membrane staining only in the basal plane of the monolayer. Polarized labeling on the basal RPE surface was clearly visible in vertical sections. Bar, 20 μm.
Figure 4.
 
Confocal microscopy of horizontal sections through the ARPE19 cell monolayer showing the same absence of apical labeling and intense basolateral labeling for both CD46 and β1 integrin in both horizontal and vertical sections. Bar, 20 μm.
Figure 4.
 
Confocal microscopy of horizontal sections through the ARPE19 cell monolayer showing the same absence of apical labeling and intense basolateral labeling for both CD46 and β1 integrin in both horizontal and vertical sections. Bar, 20 μm.
Figure 5.
 
Immunoblots of RPE from donor eyes and ARPE19 cell lines labeled with antibodies to CD46. Two bands are present in each lane: a dense upper band at 65 kDa and a lighter band at 55kDa, which correspond to the heavier and lighter isoforms of CD46, respectively.
Figure 5.
 
Immunoblots of RPE from donor eyes and ARPE19 cell lines labeled with antibodies to CD46. Two bands are present in each lane: a dense upper band at 65 kDa and a lighter band at 55kDa, which correspond to the heavier and lighter isoforms of CD46, respectively.
Figure 6.
 
RT-PCR of RPE from donor eyes and the ARPE19 cell line. Lanes 14: controls without reverse transcriptase; lanes 57: PCR products of RPE harvested from donor eyes; lane 8: PCR product from ARPE19 cells, all within the expected base pair range (448 bp) for CD46.
Figure 6.
 
RT-PCR of RPE from donor eyes and the ARPE19 cell line. Lanes 14: controls without reverse transcriptase; lanes 57: PCR products of RPE harvested from donor eyes; lane 8: PCR product from ARPE19 cells, all within the expected base pair range (448 bp) for CD46.
Figure 7.
 
CD46 coimmunoprecipitated β1 integrin from RPE cell lysates from donor eyes. (A) Immunoprecipitated proteins when anti-CD46 was used as the precipitating antibody. A dense band of the CD46 protein was labeled with anti-CD46 at the expected range of 55 to 65 kDa and a lighter band at the expected 130 kDa was labeled with anti-β1 integrin. (B) When anti-β1 integrin was used as the immunoprecipitating antibody, a dense band was present at 130 kDa that stained with anti-β1 integrin and a fainter band at 60 to 65 kDa that stained with anti-CD46.
Figure 7.
 
CD46 coimmunoprecipitated β1 integrin from RPE cell lysates from donor eyes. (A) Immunoprecipitated proteins when anti-CD46 was used as the precipitating antibody. A dense band of the CD46 protein was labeled with anti-CD46 at the expected range of 55 to 65 kDa and a lighter band at the expected 130 kDa was labeled with anti-β1 integrin. (B) When anti-β1 integrin was used as the immunoprecipitating antibody, a dense band was present at 130 kDa that stained with anti-β1 integrin and a fainter band at 60 to 65 kDa that stained with anti-CD46.
Figure 8.
 
Cell adhesion assay. (A) Inhibition of RPE attachment by anti-CD46 was dose dependent. RPE cells were incubated with various concentrations of anti-CD46 antibodies before seeding onto the basal lamina layer of human Bruch’s membrane. Anti-CD46 inhibited approximately 40% to 50% of cell attachment, with half-maximum inhibition at a concentration of 1 μg/mL. (B) Anti-β1 integrin (10 μg/mL) produced inhibition similar to that of anti-CD46 (10 μg/mL). Incubation with both antibodies did not cause any additional effect. Control RPE cells were incubated with a nonimmune IgG (mouse IgG1κ MOPC21 monoclonal antibody) to the RPE before seeding onto Bruch’s membrane.
Figure 8.
 
Cell adhesion assay. (A) Inhibition of RPE attachment by anti-CD46 was dose dependent. RPE cells were incubated with various concentrations of anti-CD46 antibodies before seeding onto the basal lamina layer of human Bruch’s membrane. Anti-CD46 inhibited approximately 40% to 50% of cell attachment, with half-maximum inhibition at a concentration of 1 μg/mL. (B) Anti-β1 integrin (10 μg/mL) produced inhibition similar to that of anti-CD46 (10 μg/mL). Incubation with both antibodies did not cause any additional effect. Control RPE cells were incubated with a nonimmune IgG (mouse IgG1κ MOPC21 monoclonal antibody) to the RPE before seeding onto Bruch’s membrane.
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×