Introduction
The Prominin-1 (Prom1) gene encodes a transmembrane glycoprotein [
1,
2], which is widely recognized as an antigenic marker for stem cells and cancer stem cells [
3,
4]. Prom1 is expressed in differentiated epithelial and non-epithelial cells [
5], glial cells [
5], and the adult retina [
6], suggesting that Prom1 plays a general role beyond stemness and differentiation status and is not limited to specific cell types. In the retina, Prom1 is present in the photoreceptor outer segments [
7] and plays an essential role in photoreceptor disk morphogenesis [
8]. The loss of function Prom1 mutations result in inherited retinal dystrophies, including autosomal dominant and autosomal recessive retinitis pigmentosa [
7,
9], cone-rod dystrophies [
10,
11,
12], and macular dystrophies [
8,
13]. Prom1-associated macular dystrophy, also known as Stargardt disease 4 (STGD4), has clinical and pathophysiological features similar to ABCA4-related Stargardt disease 1 (STGD1) and the atrophic (dry) form of age-related macular degeneration (AMD), where abnormal cytotoxic lipofuscin bisretinoid accumulation triggers degeneration of macular rods, cones, and retinal pigment epithelial (RPE) cells [
14,
15,
16]. This indicates that Prom1 dysfunction causes photoreceptor and RPE degeneration, primarily in the macula, but that disease progression may or may not involve increased bisretinoid lipofuscin. Understanding how Prom1 regulates RPE health and homeostasis is crucial for developing effective therapies.
We have previously shown that Prom1 is expressed in human and mouse RPE cell cultures in vitro. Somewhat surprisingly, Prom1 is predominantly a cytoplasmic protein in human RPE cells, and Prom1-knockout (K.O.) in human RPE cells activates mTORC1/2 activities and impairs the trafficking of autophagosomes to lysosomes [
17]. Our studies also showed that cytosolic Prom1 interacts with p62 and HDAC6 in the developing autophagosome in human RPE cells, confirming its role in autophagy regulation [
17]. We have recently shown that Prom1-KO activates mTORC1, reduces TFEB activity, and induces epithelial-mesenchymal transition (EMT) in mouse RPE (mRPE) cells, demonstrating that Prom1-mTORC1-TFEB signaling is a central driver of cell-autonomous mRPE homeostasis and suggesting a possible role in the development of geographic atrophy (G.A.) [
18].
Prom1-related retinopathies are associated with various pathogenic Prom1 variants and heterogeneous phenotypical characteristics. The main phenotypic distinction lies between recessive and dominant forms of the disease [
19]. While the recessive disease is associated with early-onset retinal degeneration, the dominant disease is associated with late-onset dystrophy, predominantly involving the macula, demonstrating that Prom1 mutations and inheritance patterns differentially impact multiple cell types in the outer retina. It is unclear whether Prom1 dysfunction primarily affects the photoreceptor cells with secondary involvement of the RPE or whether the RPE is a primary origin of the disease. Increasing evidence suggests that loss of function Prom1 mutations, including c.400C>T, p.R373C, and c.869delG mutants, cause RPE granular mottling, thinning of the outer retina, and parafoveal RPE atrophy in the macula [
20,
21,
22]. In some younger patients with a mean age of 42 years, SD-OCT showed thinning of the RPE/Bruch’s membrane, indicative of RPE cell loss and early progression to GA [
23]. Consistent with these observations, ophthalmic findings in younger patients with Prom1 R373C mutation exhibit distinct macular phenotypes, including central G.A., multifocal G.A., and bull’s eye maculopathy [
24]. In subsequent follow-up studies of these young patients, the G.A. area was significantly enlarged in a time-dependent manner. Profound degeneration of the outer retinal layer, accompanied by extensive loss of RPE cells, was consistent with the longitudinal progression of Prom1-associated retinal degeneration. While these findings suggest that Prom1 dysfunction primarily impacts the RPE, additional investigations are necessary to understand how Prom1 function differentially regulates photoreceptor versus RPE homeostasis.
To address this knowledge gap, this study focused on understanding Prom1’s expression and its significance in RPE biology. To validate Prom1 expression in the pigmented RPE in situ, we used chromogenic and fluorescent RNAscope assays and immunogold electron microscopy. To demonstrate that loss of Prom1 in the mouse leads to RPE degeneration in vivo and recapitulates clinical features of atrophic AMD, we used AAV2/1-mediated Prom1 gene knockdown (K.D.). Our studies demonstrate that Prom1 plays distinct roles in the photoreceptors vs. RPE and begin to demonstrate how Prom1 preserves the physiologic functions of the RPE.
Materials and Methods
Materials
The backbone replication-deficient all-in-one purified experimental viral particles AAV2/1.CMV.saCas9.U6.Prom1gRNA (titer of 6.76x1022 Genome Copies/ml) and control viral particles AAV2/1.CMV.saCas9.U6.scrambledgRNA (titer of 7.96x1012 Genome Copies/ml) gRNA were commercially obtained from GeneCopoeia (Rockville, MD). The AAV particles were generated following a standardized protocol using highly purified plasmids and Endofectin-AAV reagents. The mouse Prom1-transcript variant 4 mRNA-RNAscope probe (catalog # 412221) and RNAscope assay kit were purchased from ACDBio (Newark, CA). The cleaved caspase-3 (Asp175) (D3E9) rabbit mAb- Alexa Fluor 647 conjugate (catalog# 9602), goat serum (catalog # 5425), and Prolong Gold AntiFade Reagent with DAPI (catalog #8961), anti-p62 antibody (catalog# 23214), and anti-LC3-I/II antibody (catalog# 12741) were obtained from Cell Signaling Technology (Danvers, MA).
Mice and Colony Management
C57BL/6J mice were obtained from the Jackson Laboratory (JAX, stock #000664), and the albino sentinel mice were from Charles River (CD-1 strain code 022). Mice were housed, maintained on a 12-h light-dark cycle, and provided food and water ad libitum. The Institutional Animal Care and Use Committee of Vanderbilt University Medical Center (VUMC) approved all experiments. All animal procedures followed the guidelines of the Association for Research in Vision and Ophthalmology Statement on the Use of Animals in Ophthalmic and Vision Research. Both male and female mice (4-6 weeks old) were used for this project.
Subretinal Injections
Both male and female C57BL/6J mice (4-6 weeks of age) were anesthetized by intraperitoneal injection with a mixture of 12.5 mg/Kg xylazine and 62.5 mg/kg ketamine. Topical ocular anesthesia was performed using 0.5% proparacaine, and the pupils were dilated with 0.5% tropicamide. Following dilation, the animals were placed under a dissecting microscope (Nikon), and the fundus was visualized with a drop of 2.5% methylcellulose. The sclera was punctured posterior to the limbus with a 33-gauge hypodermic Hamilton needle, avoiding trauma to the iris and the lens. The needle was placed at the inferior site of the ora serrata and advanced transsclerally into the subretinal space, as described earlier [
25]. The contents of the syringe, 1ml of the viral vector solution (AAV2/1-saCas9-Prom1-gRNA or AAV2/1-saCas9-scrambled-gRNA) were slowly released into the subretinal space.
Electroretinogram (ERG) in Mice
Mice were dark-adapted overnight, dilated with 1% tropicamide, and anesthetized with 20/8/0.8 mg/kg ketamine/xylazine/urethane according to previously published methods [
26]. To prevent hypothermia in anesthetized mice, they were placed on the heated surface of the ERG system. Corneal electrodes with integrated stimulators (Celeris System, Diagnosys LLC, Lowell, MA) were placed on eyes lubricated with GenTeal drops. The subdermal platinum needle electrodes were placed in the snout and back of the head at the location of the visual cortex. A ground electrode was placed in the back of the mouse. For ERGs, mice were exposed to 50 flashes of 1 Hz, 0.05 cd s/m
2 white light with a pulse frequency of 1, as published earlier [
26]. Each experimental group had 13-15 eyes.
Chromogenic and Fluorescent Prom1 RNAscope Assays in Mouse Retina Sections
The freshly isolated eyeballs from the euthanized C57BL6J and sentinel mice were fixed in 10% neutral-buffered formalin (NBF) overnight, embedded in paraffin, cut into 3.5-4mm sections, and placed on glass slides. In some cases, the ocular posterior cup was dissected from the mouse eye for in-situ localization of Prom1 in mouse RPE. The slides were then placed on the Leica Bond-RX IHC stainer, and all steps besides coverslipping were performed on the Bond IHC stainer. Slides were baked and deparaffinized, and heat-induced antigen retrieval was performed using the Lecia Epitope Retrieval 2 solution at 95oC and the ACD 2.5 LSx protease enzyme using the ACD RNAscope® protocol. Slides were hybridized with the Prom1 probe (Cat#412228, 23214, ACD-Bio-Techne (Newark, CA) for 2 hours. Both negative (Dapb) and positive (PPIB) control slides were used alongside the target probe. The ACD RNAscope® 2.5 LSx Reagent Kit- chromogenic RED detection system was used to visualize RPE and P.R.s of the sentinel albino mouse retina sections. The slides were counterstained with hematoxylin, a Leica Bond detection system component. Slides were dehydrated, coverslipped, and used for 40X brightfield microscopy.
To visualize Prom1 expression in pigmented RPE in the C57/BL6J mice, we used the ACD RNAscope® Multiplex Fluorescent Reagents Kit system containing OPAL570 and Cy5 fluorescent markers. DAPI was used as a counterstain to visualize nuclei, an ACD/Leica RNAscope kit component. Slides were coverslipped, and images were taken using the Nikon widefield microscope (100X) and the LSM 880 confocal microscope (60X) at the Cell Imaging Shared Resource at Vanderbilt. Images were also captured using a 40X Leica fluorescent whole slide imager at Vanderbilt’s Digital Histology Shared Resource.
H&E Staining and Histology
Mice subretinally injected with scrambled or Prom1-gRNA were housed and fed ad libitum at the Vanderbilt University Department of Animal Care. After 10-11 weeks post-injection, mice were euthanized by CO2 asphyxiation followed by thoracotomy, and eyes were collected and fixed in 4% PFA. Eyes were subsequently embedded in paraffin, cut into 3.5-4mm sections, and stained with hematoxylin-eosin (H&E). The stained sections were analyzed using the Translational Pathology Shared Resource (TPSR), and images were captured using a high throughput Leica SCN400 40X Brightfield slide scanner automated digital image system from Leica Microsystems at Vanderbilt’s Digital Histology Shared Resource (DHSR). All whole slides were imaged at 40X magnification to a 0.25 mm/pixel resolution. The slides were viewed and annotated, and images were analyzed using the Aperio ImageScope (v.12.4.6.5003) available through the Digital Slide Archive (DSA) at VUMC.
Immunohistochemistry
The formalin-fixed-paraffin-embedded mouse retinal sections were placed on slides, and all steps involving immunohistochemistry (IHC) were performed on the Bond IHC stainer at Vanderbilt’s TPSR. Slides were deparaffinized, and heat-induced antigen retrieval was performed using the Epitope Retrieval 1 solution for 20 minutes. Slides were then incubated with anti LC3A/B (Cat#23214, Cell Signaling, Danvers, MA) and anti-p62 (Cat#23214, Cell Signaling, Danvers, MA) for 1 hour at a dilution of 1:2500 and 1:500, respectively. The Bond Polymer Refine Detection system was used for chromogenic visualization. The slides were dehydrated, coverslipped with hematoxylin as a counterstain, and scanned using the Leica 40X SCN400 Brightfield scanner at DHSR. Images were analyzed using the Aperio ImageScope available through DSA at VUMC.
To detect active caspase-3 levels in mouse retinal sections, antigen retrieval was performed, and the slides were blocked with goat serum. Subsequently, the slides were incubated overnight with an anti-cleaved-caspase3-Alexa Fluor 647 conjugate antibody at 40C. The slides were washed three times with 1X PBS and coverslipped with an antifade mounting medium containing DAPI. The fluorescent immunostained tissue slides were imaged on an Aperio Versa 200 automated slide scanner (Leica Biosystems) at 40X magnification to a 0.162 mm/pixel resolution. The images were analyzed using the Aperio ImageScope as Vanderbilt’s DHSR.
Transmission Electron Microscopy (TEM)
TEM was performed using methods described earlier with modifications [
27]. Briefly, eyecups were fixed in 4% PFA and 0.5% glutaraldehyde. After fixation, the samples were cryoprotected by gradual equilibrium with 30% glycerol followed by plunge freezing in liquid ethane. Samples were freeze-substituted in 1.5% uranyl acetate in methanol for 48 hours at -80
0C, followed by infiltration with HM20 at -30
0C. The HM20 was polymerized with UV light for 48 hours at -30
0C. Samples were sectioned at 100 nm nominal thickness of a UC7 ultramicrotome and collected onto 300 mesh Ni grids. For immunogold labeling, the grids were fixed, and antigens were retrieved using 0.1% sodium borohydride with 50 mM glycine and then blocked in 10% goat serum. Samples were incubated with rabbit polyclonal primary antibodies against Prom1 from Origene (Rockville, MD) (catalog number TA354470) and Abcam (Waltham, MA) (catalog number ab19898) at 1:50 dilution for 2 hours, followed by the secondary antibody at 1:20 for 1 hour. The grids were poststained with 2% uranyl acetate. TEM imaging was performed on a JEOL 2100+ equipped with an AMT nanosprint15 MKII CMOS camera using AMT acquisition software.
Analysis of Single-Cell RNA-Sequencing RPE and Retina Datasets
All published single-cell RNA-sequencing and ATAC mouse and human RPE/retina datasets were analyzed using Spectacle, an interactive online resource for single-cell RNA data analysis (
https://singlecell-eye.org/app/spectacle/). Spectacle uses retina, RPE, and choroid datasets from human and mouse samples to identify which cell types express a gene of interest and characterize gene expression changes across regions or disease states [
28].
Statistical Analysis
All data were analyzed using the GraphPad Prism 9 program (GraphPad Software Inc., San Diego, CA). Data are expressed as mean ± S.E. An unpaired 2-tailed Student’s t-test and Bonferroni post-hoc testing were used to assess statistical significance. Unless otherwise stated, *P<0.05, **P<0.01, ****P<0.0001 values were considered significant.
Discussion
Despite emerging robust data demonstrating that Prom1 regulates RPE homeostasis [
17,
18], direct evidence of its presence in mouse RPE in situ has been lacking until now. This study convincingly shows that mouse RPE expresses the Prom1 gene in situ, at least to a sufficient level, to impact key RPE processes, including waste removal by autophagy and stabilizing lysosomal activity. While Prom1 mRNA expression is significantly higher in the mouse PR inner segments, the substantially greater number of PRs than RPE cells in the mouse retina [
47] contributes partly to this observation. Prom1’s function in RPE is distinct from its role in PRs. In PRs, Prom1 is primarily involved in the structural organization of the OS and is localized to the disk membrane [
6,
48]. This localization supports its role in maintaining PR integrity and function. In contrast, in RPE cells, Prom1 is not a membrane-bound protein; its mRNA localizes to the perinuclear region, and we have shown that Prom1 functions as a signaling molecule within the cytoplasm to regulate autophagy and other intracellular processes integral to mouse RPE cell survival. These observations are consistent with our previous findings of cytoplasmic localization of Prom1 in human RPE cells [
17]. Prom1 has been observed translocating from the membrane to the cytoplasm in response to high glucose levels, indicating that the cellular environment and metabolic state significantly influence Prom1’s localization [
3]. Prom1 traffics to lysosomes occurs through its physical interactions with cytosolic histone deacetylase 6 (HDAC6) [
49]. Our previous studies in human RPE cells revealed that cytosolic Prom1 interacts with autophagy-related proteins such as p62 and HDAC6, playing a crucial role in autophagosome biogenesis, followed by its trafficking to lysosomes [
17]. Furthermore, our recent findings show that the loss of Prom1 in mouse RPE cultures disrupts autophagy and induces epithelial-mesenchymal transition, underscoring Prom1’s pivotal role as a regulator of cell-autonomous RPE homeostasis [
18]. Collectively. these studies establish that Prom1 predominantly localizes in the cytoplasm under resting conditions in RPE cells and is essential for RPE waste removal, a function markedly distinct from its well-known role at the cell surface.
This study’s novel finding is the presence of Prom1 in RPE mitochondria. This suggests a role in mitochondrial dynamics, energy metabolism, and apoptosis. This finding could have implications for understanding metabolic disorders and degenerative diseases affecting the RPE. The localization studies expand our knowledge of Prom1’s functions and interactions within RPE cells, shedding light on RPE cellular processes and diseases.
Although Prom1 mutations are linked to macular diseases caused by RPE dysfunction, the precise role of Prom1 in RPE homeostasis remains unclear. We show that AAV2/1-Cas9-Prom1-gRNA-mediated targeting of Prom1 in mouse RPE in vivo leads to RPE abnormalities and secondary P.R. degeneration. Retinal section analysis revealed patches of caspase-3 positive RPE cells, confirming RPE degeneration and apoptosis due to loss of Prom1 function, mirroring the RPE degeneration observed in patients with G.A. These findings provide insights into the pathophysiology of RPE degeneration in macular diseases and suggest that an RPE-specific Prom1-KO mouse model will enhance our understanding of Prom1’s cell-autonomous role in maintaining RPE health and homeostasis. The global Prom1-KO mice of the pure C57BL/6J background showed complete P.R. degeneration by P20. However, the retinal degeneration in the C57BL/6xCBA/NSlc mice showed significantly slower retinal degeneration, demonstrating that retinal degeneration in mouse models depends on genetic background [
50]. Although global K.O. mouse models are useful for studying human disease, tissue-specific gene targeting is required to understand the specific contribution of Prom1 in various retinal cell types and the molecular mechanisms associated with diverse disease phenotypes.
Prom1-related-retinal diseases, although relatively rare, significantly impact vision. The prevalence of these diseases varies based on specific phenotypes and genetic variants [
19]. Most studies show early macular involvement, PR degeneration, and RPE atrophy are common features of Prom1-related inherited retinal disease phenotypes [
21]. Prom1 is essential for maintaining the expression of ABCA4 and RDH12 in mouse RPE, which is crucial in regulating the visual cycle and preventing toxic bisretinoid accumulation [
50]. We have shown that loss of Prom1 in mouse RPE in vitro impairs autophagy flux, activates mTORC1, and decreases transcription factor E.B. (TFEB) activity, leading to RPE defects similar to aAMD [
18]. Furthermore, the loss of Prom1 function due to a pathogenic Prom1 mutation, combined with the heterozygous ABCA4 mutation, exacerbates RPE dysfunction, resulting in granular mottling and macula alterations, providing evidence for their shared pathophysiology [
20]. Loss of Prom1 function may lead to lipofuscin accumulation and other toxic RPE metabolites through molecular crosstalk involving ABCA4 and mTORC1. The question remains—how does Prom1 participate in the expression of these genes? Since Prom1 is not a transmembrane protein in RPE, it cannot transduce the extracellular information intracellularly, but as a cytoplasmic protein, we have shown it can regulate numerous RPE cellular processes involving degradative pathways. Therefore, additional studies are needed to explore how their interplay impacts downstream target genes and upstream regulatory pathways, affecting RPE cell fate and disease outcomes.
It remains unclear whether Prom1-associated retinal diseases originate primarily in the RPE or the P.R.s with secondary RPE damage. Patients with the Prom1 R373C mutation (autosomal dominant Stargardt-like) exhibit macular dystrophy with three distinct phenotypes: central GA, multifocal GA, and bull’s eye maculopathy, suggesting an RPE cell-autonomous function of Prom1 in the human retina [
24]. A recently identified loss-of-function Prom1 variant (c.1354dupT) has shown a multi-phenotypic effect linked to cone-rod dystrophy and retinitis pigmentosa (RP) [
51]. This cohort suggests that Prom1-related pathology may also confer an RP phenotype. Interestingly, a cohort of young patients with this newly identified Prom1 variant displayed alterations in the RPE with a preserved PR layer. Older patients with more advanced conditions exhibited both PR and RPE degeneration, suggesting that in these patients, the disease initially affects the RPE [
51]. Additionally, this study reported the early onset of an autosomal recessive form of STGD4, which differs from the late-onset autosomal dominant manifestation of STGD4 dystrophy. This confirms extensive genetic and phenotypic heterogeneity within Prom1-related inherited retinal diseases [
19,
52]. Some mutations lead to a complete loss of function, while others may result in a partially functional protein, leading to different degrees of retinal and RPE dystrophies. [
53,
54]. Modifier genes, epigenetic modifications, and other genetic factors can influence the heterogeneity of disease presentation, making diagnosis and management challenging.
We examined published human RPE single-cell RNA sequencing datasets using Spectacle and found the expression of the Prom1 gene in human RPE. Prom1 is expressed in the human retina’s macular and peripheral regions, indicating its fundamental role in maintaining RPE function. Importantly, we observed variations in Prom1 expression between distinct clusters of control RPE versus AMD RPE. These variations need further exploration to understand how Prom1 function influences RPE dysfunction and degeneration in aAMD and its potential significance in disease pathogenesis and RPE homeostasis. Analysis of a mouse retinal single-cell RNA sequencing dataset revealed the presence of Prom1 in RPE, amacrine cells, bipolar cells, early and late retinal progenitor cells, and Muller glia (data not shown), suggesting Prom1’s potential involvement in regulating retinal development, regeneration, and disease processes [
55]. The presence of Prom1 in the synaptically active amacrine cells suggests its role in integrating and shaping the visual message presented to retinal ganglion cells. Prom1 in amacrine cells may contribute to retinal circuitry by enhancing the communication between P.R.s, bipolar cells, and ganglion cells, ensuring efficient visual processing and adaptation to changing environmental conditions. Additional studies are warranted to test Prom1’s emerging function in the amacrine cells, retinal progenitor cells, and Muller glia.
We also found Prom1 expression in advanced and proliferating retinal microglia, specialized immune cells critical for surveillance, phagocytosis, and retinal health. Interestingly, activated microglia in the degenerating retina exhibit high levels of the Prom1 gene, particularly during darkness, indicating that Prom1 may be involved in microglial responses to retinal stress. Furthermore, other small microglial populations also express Prom1, suggesting its unique functional roles in regulating phenotypically heterogeneous microglia at different retinal locations. The intricate relationship between Prom1 and microglia underscores the complexity and importance of understanding Prom1-related retinal pathologies.
In summary, our study provides comprehensive insights into the multifaceted role of Prom1 in RPE homeostasis and retinal health. The detailed characterization of Prom1’s localization, function, and implications in RPE/retinal health underscores its significance as a therapeutic target. Future research on elucidating the molecular mechanisms of Prom1’s interactions with other cellular pathways will be essential for developing effective therapies for aAMD and other Prom1-associated retinal diseases. Our findings highlight the importance of Prom1 as a central mediator in maintaining RPE health and offer promising directions for therapeutic advancements in retinal diseases.
Limitations
Our study has certain limitations associated with using subretinal injections of AAV2/1.saCas9.Prom1-gRNA to target Prom1 in the mouse RPE in situ. Subretinal injections typically result in the localized delivery of the vector, meaning the AAV2/1.saCas9.gRNA complex predominantly transduces RPE cells near the injection site. The restricted diffusion of the vector from the injection site can lead to uneven distribution, limiting Prom1 gene knockdown to specific areas. Additionally, the retinal structure and extracellular matrix act as barriers that hinder the widespread dissemination of the vector, resulting in more localized effects. Variability in the subretinal injection procedure, such as differences in injection volume and pressure, can also affect the vector’s spread and the pattern of Prom1 gene knockdown. Notably, the patchy nature of Prom1 knockdown and resulting RPE degeneration more closely mirrors the localized areas of RPE loss and dysfunction seen in human geographic atrophy (GA), a late-stage manifestation of dry AMD. This finding highlights the potential relevance of our model for studying GA progression and therapeutic interventions.
In our study, the eyes injected with Prom1-gRNA exhibited cloudy, patchy, and diffuse yellowish lesions by fundus imaging, consistent with RPE cell pathology and/or dysfunction—this was the only direct evidence of lesions observed. However, H&E staining did not reveal classic atrophic lesions. Nonetheless, the observed RPE degeneration, marked by active caspase-3 labeling, indicates significant RPE cell apoptosis, an early indicator of atrophy. Localized RPE dysfunction may also affect electroretinography (ERG) readings, suggesting photoreceptor (PR) degeneration. Further studies, including quantifying PR layer thickness, are necessary to confirm secondary PR loss. While these additional studies are crucial for fully characterizing the phenotypic changes following Prom1 knockdown in mouse RPE in situ, they were not performed in this study.