Abstract
Synthetic high-density lipoprotein (sHDL) and rapamycin (Rap) have both been shown to be potential treatments for age-related macular degeneration (AMD). The low aqueous solubility of Rap, however, limits its therapeutic utility. Here we used an Apolipoprotein A-I mimetic peptide and phospholipid-based sHDL for the intravitreal delivery of Rap. By incorporation of Rap in sHDL nanoparticles (sHDL-Rap), we achieve 125-fold increase in drug aqueous concentration. When applied in vitro to retinal pigment epithelium cells, sHDL-Rap exhibited the abilities to efflux cholesterol, neutralize endotoxin, and suppress NF-κB activation. As an mTOR inhibitor, Rap induced autophagy and inhibited NF-κB-mediated pro-inflammatory signaling. Additionally, a greater reduction in lipofuscin accumulation and increased anti-inflammatory effects were achieved by sHDL-Rap relative to free drug or sHDL alone. In vivo studies demonstrated that sHDL reached the target retina pigment epithelium (RPE) layer following intravitreal administration in rats. These results suggest that sHDL-Rap holds potential as a treatment for AMD.
Keywords: high-density lipoprotein, drug delivery, rapamycin, autophagy, AMD
Graphical Abstract
Rapamycin loaded synthetic HDL (sHDL-Rap) was made by co-lyophilization and thermocycling, which resulted in uniform, 10 nm sized particles with the highest drug encapsulation efficiency (40%). These sHDL-Rap particles displayed little toxicity in ARPE-19 RPE cells and were able to modulate cholesterol homeostasis, autophagy and inflammation in RPE cells. sHDL nanoparticle was also able to target RPE layer in vivo following single intravitreal administration using fluorescently labelled DiO-sHDL nanoparticles in rats.
Introduction
Age-related macular degeneration (AMD) is the most common cause of visual impairment in developed countries (1). AMD is characterized by the loss of central vision, as the result of cell dysfunction and the death of cone photoreceptors in the macula. Photoreceptor degeneration is thought to be secondary to the dysfunction and death of underlying retinal pigment epithelium (RPE) cells (2). RPE cells serve three main functions in the visual cycle: 1) transport nutrients from the choroid to photoreceptors, 2) phagocytose outer segments shed from photoreceptors, and 3) recycle the visual pigment necessary for visual transduction. As an essential part of the visual cycle and photoreceptor health, dysfunction or loss of any of these processes performed by RPE cells may underlie the development of AMD (3).
Currently the only approved treatments for AMD are for the neovascular, or wet-AMD, with no treatments available for nonexudative, or dry-AMD. Given this lack of treatment options, there is a clear need to research potential therapies for dry-AMD. While it remains challenging to develop effective dry-AMD therapies, recent advances in the understanding of mechanisms underlying disease progression are providing novel avenues for treatment development. Increasing evidence has revealed that impaired autophagy, increased protein aggregation and inflammasome activation are all indicative of an AMD pathological phenotype.
For example, RPE cells phagocytose up to 10% of the photoreceptor outer segment (POS) length on a daily basis and process the vast amounts of lipids contained within these structures (3, 4). Therefore, dysfunctional RPE cells may have decreased autophagy and impaired lysosomal POS clearance. This could increase lipofuscin accumulation, leading to enhanced oxidative stress, protein aggregation, and drusen formation (5). Dysfunction in this pathway may also lead to exocytosis of damaged proteins which can activate an inflammatory response (6). Thus, therapeutic approaches regulating autophagy and suppressing chronic inflammation might offer some relief to patients by slowing or preventing AMD progression.
One such therapeutic option is rapamycin (Rap), a classic immunosuppressant approved by the FDA to prevent organ rejection following transplantation (7). Rap is a specific inhibitor of the mTOR (mechanistic target of rapamycin) protein which controls cell metabolism and cell growth through the regulation of protein synthesis and autophagy (8). Inhibition of mTOR in ARPE-19 cells leads to the activation of autophagy which increases autophagic flux and reduces lipofuscin-like aggregates (9). Rap can also inhibit NF-κB signaling, reducing inflammatory cytokine production (10, 11). Taken together, Rap has exciting therapeutic potential for the treatment and prevention of retinopathy in AMD.
Due to its broad therapeutic mechanism of action, Rap had been tested for the treatment of a variety of ocular diseases including AMD, diabetic macular edema, choroidal neovascularization and dry eye in multiple Phase 1/2 clinical trials (NCT00814944, NCT00656643, NCT00712491 and NCT00711490). However, it is difficult to safely administer therapeutic doses of rapamycin to the eye due to the extremely low aqueous solubility of the drug (~2μg/mL). Thus, in previous clinical studies Rap was administered in non-aqueous solutions containing 96% PEG 400 and 4% ethanol (12, 13). In these trials, Rap was administered by either a subconjunctival or an intravitreal injection at 0.3–1.3mg per dose. The high levels of solvents resulted in irritation of the eye as well as drug precipitation upon injection, leading to patient compliance issues and limiting the levels of Rap reaching the retina (14). Therefore, techniques to improve the solubility of Rap in an aqueous solution will greatly enhance its clinical utility. Nanoparticle-based systems for the effective delivery of therapeutically relevant doses of drugs to the retina are a promising technique for achieving this goal.
Compared to other nanocarriers, such as liposomes, micelles, and polymeric nanoparticles, synthetic high-density lipoprotein (sHDL) is uniquely positioned for the treatment of AMD (15–17). sHDL nanoparticles have been shown to enhance the removal of excess cholesterol from macrophages in atheroma, reduce vascular inflammation, and improve endothelial function in cardiovascular disease (18–20). In the context of AMD, recent studies found that intravitreal administration of the ApoA-1 mimetic peptide, 4F (Peptide sequence: Ac--DWFKAFYDKVAEKFKEAF-NH2), reduced lipid deposition in murine Bruch’s membrane, while topical administration of sHDL reduced the size of in mice (21, 22). Reduction of lipid deposition is critical for AMD because drusen, or accumulated lipid molecules, are a hallmark of dry-AMD. Similarly, decreasing the extent of a choroidal neovascularization injury, thus decreasing the potential for vision loss, is vital for slowing the progression of AMD. Another benefit in using sHDL as the delivery vehicle is that it is readily able to incorporate water-insoluble drugs and deliver them to target tissues (15). Combining the therapeutic utility of sHDL for reducing lipid deposition via enhanced lipid export with the anti-inflammatory and pro-autophagic properties of Rap may prove to be a novel treatment method for dry-AMD.
To test this hypothesis, we present efficacy and safety data on Rap-loaded sHDL nanoparticles tested in vitro and in vivo. We designed an sHDL-Rap compound optimized to have enhanced lipid clearance, autophagic activation, and anti-inflammatory effects. The optimized lipid composition of sHDL was made by co-lyophilization and thermocycling, which resulted in uniform, 10nm sized particles. We found the optimal drug encapsulation efficiency to be 40%. These sHDL-Rap particles displayed very low toxicity in ARPE-19 cells (>90% ARPE-19 viability) and were able to modulate cholesterol homeostasis, autophagy and inflammation in RPE cells (Schematic Design). We also showed that sHDL is targeted to RPE in vivo following single intravitreal (IVT) administration using fluorescently labeled DiO-sHDL nanoparticles in rats.
Schematic design:
Rapamycin-loaded synthetic HDL (sHDL-Rap) delivered to the retina, inducing autophagy, removing excess cholesterol, and resolving inflammation through synergistic actions.
Methods and Methods
Materials
22A peptide (PVLDLFRELLNELLEALKQKLK) was custom synthesized by GenScript (Piscataway, NJ). Rapamycin was purchased from Cayman (Ann Arbor, MI). Phospholipids, 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC), egg sphingomyelin (SM), 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), and 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) were purchased from Avanti Polar Lipids (Alabaster, AL) and NOF America Corporation (White Plains, NY). Fluorescent label, 3,3′-dioctadecyloxacarbocyanine perchlorate (DiO), was purchased from Invitrogen (Carlsbad, CA). All other reagents were analytical grade and were purchased from commercial suppliers.
Preparation and characterizations of rapamycin-loaded sHDL (sHDL-Rap)
sHDL nanoparticles were prepared by co-lyophilization followed by thermocycling (16). Briefly, 22A peptide and various phospholipids (DMPC, DPPC, POPC or DOPC) were dissolved in about 1mL acetic acid at a 22A:lipid ratio of 1:2 by weight (Equivalent molar ratios are presented in Table 1). Rapamycin was dissolved in methanol as a stock solution at 10mg/mL. Components (lipid, 22A, rapamycin) were then mixed together and subjected to lyophilization overnight to remove the organic solvent. sHDL-DiO was generated by adding 10μg DiO per 1mg peptide directly into the acetic acid mixture of peptide and DMPC. The lyophilized powder was hydrated with PBS, vortexed and subjected to heating/cooling cycles to form a clear, homogeneous solution of sHDL nanoparticles.
Table 1.
The effect of phospholipid composition and rapamycin loading on sHDL size and drug encapsulation efficiency
| Lipid (mg) | Peptide (mg) | Mole ratio (peptide: lipid) | Rapamycin (μg) | EE% | Particle size (nm) | Appearance |
|---|---|---|---|---|---|---|
| DMPC (10) | 22A (5) | 1: 7.15 | 500 | 43.4 ± 2.3 | 9.1. ± 0.2 | clear |
| 100 | 43.4 ± 1.1 | 9.8 ± 0.2 | clear | |||
| 25 | 38.4 ± 2.7 | 9.8 ± 0.2 | clear | |||
| SM (10) | 22A (5) | 1: 7.38 | 100 | 23.1 ± 1.8 | 11.0 ± 0.3 | clear |
| DPPC (10) | 22A (5) | 1: 7.15 | 100 | 23.5 ± 1.8 | 10.1 ± 0.2 | clear |
| POPC (10) | 22A (5) | 1: 6.90 | 100 | 43.5 ± 2.6 | 18.8 ± 0.6 | cloudy |
| DOPC (10) | 22A (5) | 1: 7.15 | 100 | 31.8 ± 1.8 | 20.6 ± 1.2 | cloudy |
Size distribution was determined by dynamic light scattering (DLS) on a Malvern Zeta sizer (Westborough, MA). Particle purity was determined by gel permeation chromatography (GPC) with UV detection at 220 nm using a Tosoh TSK gel column (Tosoh Bioscience, King of Prussia, PA) separated by a Waters HPLC, as previously described (16).
Encapsulation efficiency (EE) and drug-loading (DL) capacities were determined after removing un-encapsulated rapamycin using Zeba Spin Desalting columns (5 mL7K MWCO, Thermo Fisher). The sHDL-Rap nanoparticles collected before and after desalting were analyzed by UPLC detection for drug content. The UPLC measurement was carried out with an Acquity UPLC BEH C18 column (1.7μm, 2.1×100mm, Waters, Milford, MA, USA) at 40°C and the mobile phase was methanol:water (with 0.1% formic acid) at a ratio of 80:20 at 0.3mL/min with an injection volume of 10μL. The concentration of rapamycin was detected by UV absorbance at a wavelength of 278 nm. EE and DL were calculated according to the following formulas: EE (%) = rapamycin encapsulated in sHDL/total amount of rapamycin×100 %, DL (%) = Theoretical loading × EE%.
For stability evaluations, sHDL-Rap (rapamycin 100μg/mL) and Rap solution (10μg/mL) were incubated in PBS (pH 7.4) at 37°C for 72 hours. The particle size of sHDL-Rap at different time points was measured by DLS. Undegraded Rap contents in rapamycin solution or sHDL-Rap were analyzed by UPLC as described above. In a parallel experiment, at different time points during incubation, sHDL-Rap was passed through Zebra Spin Desalting Columns (MWCO 7kD) to remove unencapsulated rapamycin. The encapsulated intact Rap was then quantified using UPLC as described above to calculate drug retention percentage.
Culture of RPE cells
ARPE-19 cell line was purchased from ATCC (CRL-2302, Manassas, VA) and routinely maintained in DMEM/F12 medium supplemented with 10% FBS (Gibco, Grand Island, NY). Highly-polarized RPE monolayers were established by culturing in DMEM/F12 medium for 1 month prior to experiments. For transepithelial electrical resistance (TEER) measurements, cells were grown on Transwell® cell culture inserts (6.5mm diameter, 0.4mm pore size, polyester, Corning, Tewksbury, MA, USA). TEER was measured using the Millicell® ERS-2 voltohmmeter (MilliporeSigma).
Cytotoxicity assay
A cytotoxicity assay was conducted using slight deviations from the existing MTT assays (23). Briefly, ARPE-19 cells were seeded on a 96-well plate at a density of 1×104/per well. After 1 month of culture, cells were incubated with sHDL, Rap or sHDL-Rap at different concentrations for 24 hours. MTT solution (Sigma, 5mg/mL in PBS) was added into each well and incubated for 4 hours. The media was removed, and cells were dissolved in dimethyl sulfoxide. Absorbance was measured at 550 nm using a microplate reader (SynergyTM NEO HTS Multi-Mode Microplate Reader, Bio-Tek). PBS-treated cells were set as the baseline for 100% viability.
Cholesterol efflux assay
Phenol red-free media was used to avoid any interference with physiological cholesterol pathways. RPE cells were seeded in 24-well transwell insert with 0.4 μm pore size (Corning, NY) and cultured for one month (transepithelial electrical resistance (TEER) value reached around 50Ω/cm2). They were then labeled with [1,2-3H]-cholesterol at a concentration of 1μCi/mL (Perkin Elmer) in DMEM containing 0.5% fatty acid-free bovine serum albumin for 24 hours. Cells were then washed twice with PBS and equilibrated in fresh DMEM containing 0.5% fatty acid-free bovine serum albumin supplemented with 5μg/ml of the ACAT inhibitor Sandoz 58–035 (Sigma) for 24 hours. Rapamycin (1μg/mL), sHDL (50μg/mL) or sHDL-Rap (1μg/mL of Rap and 50μg/mL of sHDL) was then added into the medium for 16 hours. To measure the cholesterol efflux, the media were collected and centrifuged to remove cell debris while the cells were lysed with 0.1% SDS/0.1M NaOH solution for 2 hours. Radioactivity in the media and cell lysate were counted on a PerkinElmer scintillation counter. Cholesterol efflux in the media was expressed as a percentage of total cell [3H]-cholesterol content.
Western blot
After different treatments, ARPE-19 cells were washed twice with cold PBS and lysed in RIPA buffer containing an EDTA-free protease inhibitor cocktail (Sigma) and kept on ice for 30 min. Protein concentration in the supernatant was measured with the Bradford assay reagent (BioRad, Cressier, Switzerland). Protein lysates (50μg/lane) were separated by SDS-PAGE then transferred to PVDF membranes. Membranes were blocked in 5% non-fat blocking milk (BioRad) for 1 hour at room temperature and incubated overnight with the following primary antibodies: rabbit anti-LC3 (CST 4108S, 1:1000 dilution), anti-P65 (CST 8242S, 1:1000 dilution) and anti-GAPDH (CST 5174S, 1:1000 dilution). After washing, membranes were incubated with appropriate HRP conjugated secondary antibodies and their signal was developed using an enhanced chemiluminescence (ECL, Amersham Biosciences) substrate. Images were acquired on a Protein Simple FluorChem M imaging system (San Jose, CA). Intensity of bands was quantified using ImageJ and normalized to GAPDH levels.
Lipofuscin images by confocal microscope
For confocal microscopy imaging, ARPE-19 cells were cultured on gelatin-coated coverslips in a 6-well plate and treated with POS (4μg protein per cm2 cell growth area, Invision Bioresources, Seattle, WA) daily for 7 days. Cells were also treated with Rap (1μg/mL), sHDL (50μg/mL) or sHDL-Rap (1μg/mL of Rap) on days 6 and 7. Cells were then washed with PBS to remove non-internalized POS, fixed with 4% paraformaldehyde, and nuclei were stained with 5μg/mL DAPI. Finally, cells were imaged by a Nikon A-1 confocal microscope.
Anti-inflammation effects
Confluent ARPE-19 cells were treated with PBS, Rap, sHDL or sHDL-Rap for 18 hours, followed by lipopolysaccharide (LPS, 10μg/ml; Sigma–Aldrich) and 4-hydroxynonenal (HNE, 15μM; Sigma–Aldrich) for 24 hours. Media samples were collected, and the concentrations of soluble pro-inflammatory cytokines IL-6, TGF-β, and IL-1β were measured in triplicate using commercial ELISA kits (Bio-legend) according to the manufacturer’s protocols.
Biodistribution in vivo
The biodistribution of sHDL following intravitreal (IVT) injection was examined using 3,3′-dioctadecyloxacarbocyanine perchlorate (DiO)-loaded sHDL nanoparticles. Male, Sprague-Dawley rats (age ~2–3 months) were purchased from Charles River (Wilmington, MA, USA). 3–4 animals per group were used. 2μL of DiO-sHDL was injected into the left eye of each animal and free DiO dissolved in DMSO (DMSO-DiO) was injected into the right eye. Rats were euthanized 24 hours and 72 hours post-injection, and eyes were enucleated and immediately flash frozen in OCT (optimal cutting temperature) media. 10μm thick sections were prepared and coverslipped using ProLong™ Gold Antifade Mountant with DAPI (Thermo Fisher Scientific, Waltham, MA, USA Cat# P36930) and DiO fluorescence in the outer retina was evaluated using a Leica DM6000 wide field fluorescent microscope with a 40x objective. Additional frozen sections were prepared and subjected to H&E staining as previously described (24).
Statistical analysis
All data are presented as mean ± standard deviation. Statistical comparisons were performed by one-way ANOVA for multiple groups. p < 0.05, 0.01, and 0.001 were considered statistically significant and marked with *, **, ***, respectively.
Results
Optimizing sHDL nanoparticles for rapamycin delivery
In this study, we made sHDL consisting of the ApoA-I mimetic peptide, 22A, and phosphatidylcholines that were previously utilized by our lab to deliver a variety of drugs (16, 25, 26). This ApoA-I mimetic peptide retains many biological functions of endogenous ApoA-I protein, easily forms homogeneous sHDL nanoparticles, and demonstrated favorable safety and pharmacokinetic profiles in human clinical trials (27, 28). The selection of phospholipid is critical as fatty acid chain lengths and degrees of saturation impact the fluidity of the sHDL membrane, which, in turn, may affect the efficiency of drug molecule incorporation (17). We developed a panel of sHDL formulations containing the 22A peptide with various lipids to optimize the loading of Rap (Fig. 1A). Phospholipids with low transition temperature (Tm) (for example, DOPC with Tm = −17°C and POPC with Tm = −2°C, respectively) made sHDL suspensions murky, whereas phospholipids with high Tm (for example, DPPC and DMPC with Tm of 41°C and 24°C, respectively) formed clear sHDL solutions. sHDL nanoparticles with clear solutions had an average diameter of 10nm, as determined by dynamic light scattering (Table 1). Transmission electron micrographs of sHDL particles containing DMPC phospholipids also showed uniform spherical shapes (Fig. 1B) with a purity >95% as evidenced by GPC (Fig. 1C). To determine encapsulation efficiency of Rap, unencapsulated rapamycin was removed via a desalting column, and sHDL-Rap particles were assayed. UPLC was used to quantify encapsulation efficiency. The highest rapamycin encapsulation efficiency was achieved using DMPC-based sHDL (43%), followed by SM (23%) and DPPC (23%) based sHDL. DMPC-sHDL encapsulation efficiency did not change significantly when generating sHDL-Rap particles using 25, 100 or 500μg/ml or rapamycin, showing ample capacity to incorporate rapamycin. When compared to the low aqueous solubility of rapamycin (2μg/mL), incorporation of rapamycin into sHDL showed an effective solubility of 250μg/mL resulting in 125-fold improvement in solubility without using organic solvents. Based on sHDL appearance, particle size homogeneity and encapsulation efficiency, we chose DMPC as the lipid component of our sHDL-Rap formulation for further investigation.
Figure 1.
Schematic representation of the sHDL nanoparticle preparation and characteristics (A) Schematic representation of protocol for generating rapamycin-encapsulated sHDL nanoparticles. These particles are comprised of DMPC, 22A peptide and rapamycin. (B) Representative TEM image of sHDL-Rap nanoparticles. The scale bar is set to 20 nm. (C) Size distribution measured by DLS. (D) The purity analysis analyzed by GPC. (E) Particle sizes of sHDL-Rap at different time points under 37°C incubation. (F) Percentage of the intact rapamycin in sHDL-Rap and rapamycin PBS solution. (G) Percentage of encapsulated and intact rapamycin in sHDL-Rap. Data are reported as mean ± SD, n = 3.
The stability of optimized sHDL-Rap was evaluated in terms of particle size and chemical stability of rapamycin. As shown in Fig 1E, the particle size of sHDL-Rap remained stable in prolonged incubation under 37°C without signs of aggregation. As rapamycin is prone to degradation under physiological pH (29), the chemical stability of rapamycin in sHDL-Rap was evaluated in comparison with free rapamycin solution. Results showed that less than 20% rapamycin remained undegraded in PBS solution within 24 hours. However, the chemical stability of rapamycin was greatly increased in sHDL-Rap formulation, with ~80% rapamycin, either encapsulated in sHDL particles or released as free form, staying intact at 24 hour time point (Fig 1F). The percentage of the intact rapamycin encapsulated in sHDL-Rap was determined after removing released drug molecules using desalting columns. Results showed that around 90% drug was released or degraded within 24 hours of incubation, and a complete drug release/degradation was observed in the 72 hour time point (Fig 1G).
The effect of sHDL-Rap on cell viability and cholesterol efflux
We next tested for any cytotoxicity of our sHDL and sHDL-Rap formulations in RPE-like cells (ARPE-19). sHDL has been widely tested in various cell types and is considered nontoxic over a range of concentrations (16, 30). In ARPE-19 cells, drug-free DMPC-based sHDLs show little cytotoxicity at all concentrations tested (10–100μg/mL of 22A peptide) (Fig. 2A). Treatment with rapamycin displayed clear cytotoxicity in ARPE-19 cells at the concentration of 10μg/mL (73.01% cell viability ). Concentrations of rapamycin lower than 5μg/mL exhibited no significant toxic effects on ARPE-19 cells. (Fig. 2B). Similar results were obtained by treating cells with sHDL-Rap. We chose an effect-concentration of 1μg/mL for rapamycin in the following in vitro studies based on previously published research (31–33). Recent studies have suggested that the pathophysiology of AMD is related to the dysregulation of cholesterol metabolism (34–36). In order to evaluate the ability of sHDL-Rap particles to enhance cholesterol efflux, ARPE-19 cells were assayed for their ability to retain labeled cholesterol (Fig. 2C). Rap alone had no effect on cholesterol efflux as expected. More importantly, incorporation of Rap into sHDL did not attenuate sHDL’s efflux capacity. sHDL and sHDL-Rap exhibited similar cholesterol efflux in RPE cells (~40%) (Fig. 2D). This efflux capacity was similar to values shown for sHDL treated atheroma macrophages (16).
Figure 2.
In vitro cell viability and cholesterol efflux assays (A) MTT assay on ARPE-19 cells treated with sHDL for 24 hours. ARPE-19 cells displayed no significant decrease in cell viability when treated with a wide range of sHDL concentrations. (B) MTT assay on ARPE-19 cells treated with Rap or sHDL-Rap for 24 hours. (C) Schematic representation of the cholesterol efflux assay. (D) sHDL and sHDL-Rap induced significant efflux of [3H]-cholesterol efflux from ARPE-19. Radioactivity counts were measured in cell lysate and media. Calculated % efflux = (media counts)/(media + cell counts)*100. Data is reported as mean ± SEM, n = 3.
Effects of Autophagy Induction or Inhibition on RPE Cell Autofluorescence
Recent evidence indicates that decreased autophagic flux is associated with RPE impairment and AMD pathology (37, 38). Autophagy decline in RPE cells can result in the accumulation of aggregation-prone proteins, cellular degeneration and ultimately cell death. This presents itself in the form of accumulated auto-fluorescent lipid-protein aggregates, called lipofuscin, in the lysosomes of RPE cells (6). Thus, regulation of autophagy signaling pathways is a potential therapeutic target for AMD treatment. The signaling of mechanistic target of rapamycin (mTOR) has proven to be the key regulator for autophagy activity. Recent findings demonstrated that rapamycin can inhibit mTOR complex 1 (mTORC1), and, in turn, activate autophagy and slow the aging and neurodegenerative processes in mice (39, 40). A commonly accepted criterion to estimate autophagic flux is to measure the ubiquitin-like process. The microtubule-associated protein 1 light chain (LC3-I) will notably become LC3-II after lipidation. LC3-II is then inserted into the inner and outer membranes of the autophagosomes and eventually degraded (41). We measured activation of autophagy after treating ARPE-19 cells with Rap, sHDL, or sHDL-Rap. We used the ratio of LC3-II relative to LC3-I as a quantitative index of autophagy—with an increased ratio representing autophagy activation. Our results show that ARPE-19 cells displayed strong activation of autophagy after rapamycin treatment (Fig. 3A), and displayed a similar response following treatment with sHDL-Rap, suggesting that encapsulation in an sHDL nanocarrier did not alter the effect of rapamycin. sHDL alone had little effect on autophagic flux (Fig. 3A).
Figure 3.
Treatment with sHDL-Rap induces autophagy and reduces lipofuscin formation. (A) Western blot results for the marker of autophagy-microtubule associated protein 1 light chain 3 (LC3). Upper band: LC3-I (cytoplasmic form); lower band: LC3-II (autophagosomal form). ARPE-19 cells were treated for 24 hours with PBS, Rap (1μg/mL), sHDL, or sHDL-Rap (1μg/mL). LC3-II/LC3-I ratios were calculated (mean ± SEM, n = 3, *** p < 0.001). (C) ARPE-19 cells were examined by confocal microscopy following 7 days of incubation with: PBS without POS, PBS with POS, Rap (1μg/mL) with POS, sHDL with POS, or sHDL-Rap (1μg/mL) with POS. Lipofuscin-like cellular autofluorescence (green) was detected using a confocal microscopy (excitation, 480nm; detection, 535nm). Nuclei were visualized by DAPI staining (blue) (Scale bar: 20μm). Fluorescence intensity levels were quantified by Image J (B).
Autophagy is critical for the breakdown of photoreceptor outer segments (POS) and a loss of this breakdown can lead to the accumulation of lipofuscin, an early indicator of AMD pathology. ARPE-19 cells were incubated daily with POS for 7 days and cellular autofluorescence of lipofuscin was detected by confocal microscopy (excitation, 480nm; detection, 535nm). POS induced significant formation of lipofuscin, characterized by autofluorescence (green dots, Fig. 3B). A significant reduction of autofluorescence was observed in Rap groups. This suggests that rapamycin suppresses mTOR, which stimulates autophagosome activity, leading to a more complete degradation of material, resulting in decreased lipofuscin accumulation (9). sHDL alone also showed a significant reduction of lipofuscin formation. Between this study and others, sHDL has shown its ability to remove excess cholesterol and photoreceptor outer segment-derived cholesterol (42). However, further experiments are required to study the physiological role and regulation of sHDL in lipofuscin removal. The sHDL-Rap group here had the most noticeable decrease in fluorescence. Our data fit well with the hypothesis that sHDL-Rap has the strongest ability to inhibit lipofuscin formation.
sHDL-Rap suppresses inflammatory cytokines in ARPE-19 cells
We next evaluated the anti-inflammation effects of sHDL-Rap in ARPE-19 cells. Phosphorylated NF-κB p65, a transcription factor, is generally upregulated during inflammation. After incubation with LPS and 4-hydroxynonenal (HNE), p65 protein levels increased significantly, but this effect was inhibited by treatment with Rap (Fig. 4A). sHDL alone was also capable of reducing p65 protein levels after an inflammatory event, consistent with a previous study in macrophages (43). Interestingly, sHDL-Rap further reduces p65 protein levels after treatment with LPS & HNE, suggesting a synergistic effect. Similarly, the levels of the inflammatory cytokines IL-6 and IL-1β were increased after LPS and HNE incubation, but were attenuated by rapamycin (Fig. 4B, 4C). Both rapamycin and sHDL exhibited drastic reductions in IL-6 and IL-1β compared to PBS controls. As expected, sHDL-Rap displayed superior anti-inflammatory effects compared to rapamycin or sHDL alone. Taken together, these results suggest that sHDL is an effective carrier of rapamycin, retaining the anti-inflammatory effects of both rapamycin and sHDL, allowing for better protective effects in vitro.
Figure 4.
sHDL-Rap is effective at preventing pro-inflammatory signaling. (A) NF-κB subunit p65 protein expression in ARPE-19 cells was measured by western blot. Cells were treated with PBS, Rap, sHDL or sHDL-Rap for 18 hours, followed by LPS+HNE for 24 hours. “No treat” indicates cells which received no treatment of sHDLs or LPS+HNE. (B, C) Pro-inflammatory cytokines (IL-1β, IL-6) were measured by ELISA after treatments as previously described (mean ± SEM, n = 3, *p < 0.05, **p < 0.01, ***p < 0.001 compared to PBS, # p < 0.05 compared to sHDL-Rap).
Intraocular distribution of sHDL
The target tissues for sHDL-Rap are in the posterior segment of the eye, and nanoparticles will likely need to be administered by the intravitreal route. Intravitreal injection is routinely used in clinical practice for the administration of anti-VEGF, and in clinical trials for the administration of organic solvent solutions of Rap. The ability of sHDL-Rap to target RPE cells following intravitreal administration was examined by measuring the biodistribution using fluorescently labeled DiO-sHDL nanoparticles in Sprague-Dawley rats. At 24 hours post-injection, DiO labeled sHDLs diffused rapidly from the vitreous to the retina and accumulated within the RPE layer, indicating transretinal movement of the sHDL, whereas DiO delivered in a solution of DMSO did not efficiently target the outer retina or RPE (Fig. 5A). The intensity of the green fluorescence signal in the retina 72 hours post-injection was much lower compared to that of 24 hours post-injection, indicating the elimination of intra-retinal sHDL nanoparticles (Fig. 5A). In addition, there was no observed histological evidence of toxicity at 24 and 72 hours post-dose following IVT administration of sHDL particles (Fig. 5B). These results indicate that sHDL are capable of efficiently and safely delivering drug cargo to RPE cells following IVT administration. This accumulation of sHDL nanoparticles in the posterior segment of the eye is superior to the delivery of the hydrophobic drug by organic solution (DMSO) injection known to elicit local retinal toxicity (44).
Figure 5.

Successful delivery of sHDL nanoparticles to the retina and RPE. (A) Representative images of frozen sections show fluorescent signals in the outer retina and RPE layers of sHDL-DiO at 24 hours post injection. The fluorescent signal from DiO is mostly absent by 72 hours post injection. (B) H&E staining of frozen sections shows no gross morphological defects associated with sHDL-DiO injection. DMSO – Dimethylsulfoxide, sHDL – synthetic High Density Lipoprotein, ONL – Outer Nuclear Layer, IS/OS – Inner Segment/Outer Segment region, RPE – Retinal Pigment Epithelium. 3–4 animals were included per group.
Discussion
In this work, we have optimized the composition of sHDL for the incorporation of Rap. The highest encapsulation efficiency of Rap (~40%) was achieved by preparing sHDL particles using DMPC. sHDL-Rap nanoparticles, consisting of 22A peptide and DMPC, contain 0.27% (w/w) rapamycin. These sHDL-Rap nanoparticles showed homogeneous shape and were approximately 10nm in diameter. The small and stable particle size of sHDL-Rap is particular favorable to achieving efficient retina delivery, as previous studies have suggested that nanoparticles with small particle sizes diffuse faster within the vitreous and have higher efficiency in retina penetration (45, 46). Encapsulating rapamycin to sHDL particles significantly increased the effective aqueous solubility of Rap and greatly decreased the degradation of rapamycin in physiological pH, which improves the bioavailability of Rap after intravitreal injection. We have demonstrated that sHDL-Rap are effective at inducing cholesterol efflux from ARPE-19 cells and are more effective than Rap alone at inducing autophagy to clear POS, preventing the accumulation of lipofuscin. This, combined with the enhanced solubility of sHDL-Rap in aqueous solutions, creates a promising new delivery mechanism for drugs to the retina.
The advantage of our approach is the utilization of the dual functionality of sHDL. It serves as the nanocarrier to deliver a hydrophobic drug, in this case Rap, and as the cholesterol acceptor, removing excess cholesterol from target tissues (16, 47). One of the key features of dry-AMD is the accumulation of drusen on the basal side of RPE cells. These deposits are rich in lipids, making a therapeutic system which can aid in the clearance of these deposits very useful. In addition, retinal macrophages and monocytes display lower cholesterol clearance capacities, exacerbating the progression of AMD (48, 49). We first demonstrated that sHDL alone enhances the efflux of radiolabeled cholesterol from ARPE-19 cells (Fig. 2) and that loading sHDL nanoparticles with Rap did not inhibit this effect. Other studies also demonstrated that both sHDL and ApoA-1 mimetic peptides can reduce lipid deposition under the RPE (50, 51). Federica et al. demonstrated that in human primary RPE cells, 3H-cholesterol-labeled POS was effluxed to ApoA-I (52), although the mechanisms underlying this observation require further study. The dysfunction of proper cholesterol clearance from RPE is a driver of dry-AMD pathology, therefore restoring this function to RPE may be very beneficial for patients (53, 54).
We next investigated the regulatory function of Rap in autophagy signaling pathways. Impaired autophagy activity in senescent cells is related to numerous diseases, including neurodegenerative disorders, heart diseases and cancer (55, 56). Therefore, enhancing autophagy is expected to be beneficial to AMD therapy. Our studies indicate that treatment of ARPE-19 cells with Rap alone or sHDL-Rap can effectively induce autophagy (Fig. 3). The stimulation of autophagy by either Rap or sHDL-encapsulated Rap leads to a decrease in lipofuscin accumulation. These results demonstrate that the pharmacological efficacy of Rap is not lost after encapsulation in sHDL. Interestingly, sHDL alone also showed a relatively strong ability to decrease lipofuscin accumulation without activating autophagy. This could be explained by the ability of sHDL to efflux the outer segment (OS)-derived cholesterol. Thus, sHDL alone is capable of increasing lipid efflux from ARPE-19 cells, while the addition of Rap facilitates intracellular autophagy activity, followed by lipid clearance, thus decreasing overall cellular lipid accumulation. While additional mechanistic studies are required to elucidate individual contributions of both the sHDL carrier and Rap to overall sHDL-Rap efficacy, these results further highlight the advantage of using therapeutically active lipoprotein carriers relative to other traditionally used polymer or lipid-based nanocarriers. Lastly, sHDL-Rap was chosen for AMD treatment due to the anti-inflammatory potential of both sHDL and Rap. An inflammatory response may further accelerate AMD pathology and can occur when autophagic activity drops. In conjunction with this response, there is an increase in lipofuscin accumulation, leading to elevated protein aggregation and ROS production (57). Similarly, the induction of chronic inflammation can accelerate deficiency of autophagy and AMD progression (58). In addition to its role in autophagy, Rap has also been shown to suppress locally activated mTOR levels and related inflammatory molecules (IL-6, MCP-1) through NF-κB suppression. The anti-inflammatory functions of sHDL have been widely studied. These functions include neutralization of endotoxins and reduction of TLR4 recruitment to the lipid raft (59), inhibiting the activation of NF-κB. Furthermore, HDL can activate transcription factor 3 (ATF3) expression to suppress TLR-induced pro-inflammatory cytokine release (60). Our data show that sHDL-Rap exhibited superior anti-inflammatory activity in RPE cells compared to either Rap or sHDL alone, suggesting a synergistic effect.
Various other nanocarriers for ophthalmic delivery of Rap gained considerable attention due to the poor aqueous solubility of Rap and the toxicity of solvent formulations (61–63). MPEG-PCL micelles loaded with Rap increased drug stability and enhanced bioavailability following systemic administration (61). When this drug was delivered intravitreally in a model of experimental autoimmune uveitis, intraocular inflammation was abolished via the downregulation of the Th1 and Th17 response. Another liposomal-based nanoparticle formulation containing Rap (0.4mg/mL and 1mg/mL) was developed, and shown to improve dry eye pathology in a keratoconjunctivitis sicca dog model (62). These nanoparticle-rapamycin formulations show potential for the treatment of dry eye and ocular inflammation; however, they do not involve using a therapeutically active carrier like sHDL.
Beyond the inherent advantages of being a nanocarrier, sHDL has great potential as a therapeutic delivery option due to its other biological activities. sHDL particles possess unique physicochemical properties, including naturally synthesized components, amphipathic apolipoproteins, lipid-loading, hydrophobic agent-incorporating characteristics, and specific receptor-mediated uptake. Multiple sHDL products, CER-001, CSL-112, ETC-216 and ETC-642, have been proven safe and well-tolerated in both healthy and diseased human patients (27, 64, 65). Moreover, their manufacture under current good manufacturing practices (cGMP) is established and these processes could be easily adapted to incorporate rapamycin (15, 66). In addition, sHDL has been shown to facilitate cholesterol efflux and anti-inflammation efficacy in our previous studies (16, 17, 43), indicating the advantages of using it as a treatment for AMD.
In summary, here we applied a therapeutically active carrier, sHDL, for the delivery of a hydrophobic drug, rapamycin, to the back of the eye. Our future studies will involve the examination of sHDL-Rap’s ability to reduce the pathology of AMD in animal models. The combination of sHDL’s potential therapeutic utility in lipid dysregulation and its ability to effectively reach the posterior segment of the eye following IVT administration might be translatable in the delivery of other hydrophobic ocular therapeutics.
Acknowledgements
This work was supported by NIH R01 HL134569 and R21 NS111191 (A.S.), R01 EY029675 (C.G.B), American Heart Association Postdoctoral Fellowship 20POST35210818 (L.M.).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Reference
- 1.Wong WL, Su X, Li X, Cheung CMG, Klein R, Cheng C-Y, et al. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: a systematic review and meta-analysis. The Lancet Global Health. 2014;2(2):e106–e16. [DOI] [PubMed] [Google Scholar]
- 2.Nowak JZ. Age-related macular degeneration (AMD): pathogenesis and therapy. Pharmacological Reports. 2006;58(3):353. [PubMed] [Google Scholar]
- 3.Strauss O The retinal pigment epithelium in visual function. Physiological reviews. 2005;85(3):845–81. [DOI] [PubMed] [Google Scholar]
- 4.Bok D. The retinal pigment epithelium: a versatile partner in vision. Journal of cell science. 1993;1993(Supplement 17):189–95. [DOI] [PubMed] [Google Scholar]
- 5.Kaarniranta K, Sinha D, Blasiak J, Kauppinen A, Veréb Z, Salminen A, et al. Autophagy and heterophagy dysregulation leads to retinal pigment epithelium dysfunction and development of age-related macular degeneration. Autophagy. 2013;9(7):973–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mitter SK, Song C, Qi X, Mao H, Rao H, Akin D, et al. Dysregulated autophagy in the RPE is associated with increased susceptibility to oxidative stress and AMD. Autophagy. 2014;10(11):1989–2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Li J, Kim SG, Blenis J. Rapamycin: one drug, many effects. Cell metabolism. 2014;19(3):373–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hands SL, Proud CG, Wyttenbach A. mTOR’s role in ageing: protein synthesis or autophagy? Aging. 2009;1(7):586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lei L, Tzekov R, Li H, McDowell JH, Gao G, Smith WC, et al. Inhibition or stimulation of autophagy affects early formation of lipofuscin-like autofluorescence in the retinal pigment epithelium cell. International journal of molecular sciences. 2017;18(4):728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Okamoto T, Ozawa Y, Kamoshita M, Osada H, Toda E, Kurihara T, et al. The neuroprotective effect of rapamycin as a modulator of the mTOR-NF-κB axis during retinal inflammation. PloS one. 2016;11(1):e0146517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhang J, Bai Y, Huang L, Qi Y, Zhang Q, Li S, et al. Protective effect of autophagy on human retinal pigment epithelial cells against lipofuscin fluorophore A2E: implications for age-related macular degeneration. Cell death & disease. 2015;6(11):e1972-e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mudumba S, Dor P, Nivaggioli T, Weber D, Farooq S, inventors; United States Patent, assignee. Liquid formulations for treatment of diseases or conditions 2006. [Google Scholar]
- 13.Mudumba S, Dor P, Nivaggioli T, Weber D, Farooq S, inventors; United States Patent, assignee. Rapamycin formulations and methods of their use 2018. [Google Scholar]
- 14.Patel S, Barnett JM, Kim SJ. Retinal toxicity of intravitreal polyethylene glycol 400. Journal of Ocular Pharmacology and Therapeutics. 2016;32(2):97–101. [DOI] [PubMed] [Google Scholar]
- 15.Kuai R, Li D, Chen YE, Moon JJ, Schwendeman A. High-density lipoproteins: nature’s multifunctional nanoparticles. ACS nano. 2016;10(3):3015–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Guo Y, Yuan W, Yu B, Kuai R, Hu W, Morin EE, et al. Synthetic high-density lipoprotein-mediated targeted delivery of liver X receptors agonist promotes atherosclerosis regression. EBioMedicine. 2018;28:225–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Yuan W, Yu B, Yu M, Kuai R, Morin EE, Wang H, et al. Synthetic high-density lipoproteins delivering liver X receptor agonist prevent atherogenesis by enhancing reverse cholesterol transport. Journal of Controlled Release. 2020;329:361–71. [DOI] [PubMed] [Google Scholar]
- 18.Nissen SE, Tsunoda T, Tuzcu EM, Schoenhagen P, Cooper CJ, Yasin M, et al. Effect of recombinant ApoA-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial. Jama. 2003;290(17):2292–300. [DOI] [PubMed] [Google Scholar]
- 19.Tardif J-C, Grégoire J, L’Allier PL, Ibrahim R, Lespérance J, Heinonen TM, et al. Effects of reconstituted high-density lipoprotein infusions on coronary atherosclerosis: a randomized controlled trial. Jama. 2007;297(15):1675–82. [DOI] [PubMed] [Google Scholar]
- 20.Riwanto M, Landmesser U. High density lipoproteins and endothelial functions: mechanistic insights and alterations in cardiovascular disease. Journal of lipid research. 2013;54(12):3227–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Suda K, Murakami T, Gotoh N, Fukuda R, Hashida Y, Hashida M, et al. High-density lipoprotein mutant eye drops for the treatment of posterior eye diseases. Journal of controlled release : official journal of the Controlled Release Society. 2017;266:301–9. [DOI] [PubMed] [Google Scholar]
- 22.Rudolf M, Mir Mohi Sefat A, Miura Y, Tura A, Raasch W, Ranjbar M, et al. ApoA-I Mimetic Peptide 4F Reduces Age-Related Lipid Deposition in Murine Bruch’s Membrane and Causes Its Structural Remodeling. Curr Eye Res. 2018;43(1):135–46. [DOI] [PubMed] [Google Scholar]
- 23.Supino R. MTT assays. In vitro toxicity testing protocols: Springer; 1995. p. 137–49. [Google Scholar]
- 24.Wubben TJ, Pawar M, Weh E, Smith A, Sajjakulnukit P, Zhang L, et al. Small molecule activation of metabolic enzyme pyruvate kinase muscle isozyme 2, PKM2, circumvents photoreceptor apoptosis. Scientific reports. 2020;10(1):2990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kuai R, Ochyl LJ, Bahjat KS, Schwendeman A, Moon JJ. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nature materials. 2017;16(4):489–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yuan Y, Wen J, Tang J, Kan Q, Ackermann R, Olsen K, et al. Synthetic high-density lipoproteins for delivery of 10-hydroxycamptothecin. International journal of nanomedicine. 2016;11:6229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Khan M, Lalwani N, Drake S, Crockatt J, Dasseux J, editors. Single-dose intravenous infusion of et al. -642, a 22-Mer ApoA-I analogue and phospholipids complex, elevates HDL-C in atherosclerosis patients. Circulation; 2003: LIPPINCOTT WILLIAMS & WILKINS 530 WALNUT ST, PHILADELPHIA, PA: 19106–3621 USA. [Google Scholar]
- 28.Miles J, Khan M, Painchaud C, Lalwani N, Drake S, Dasseux J-L. P105 single-dose tolerability, pharmacokinetics, and cholesterol mobilization in Hdl-c fraction following intravenous administration of Etc-642, a 22-mer Apoa-i analogue and phospholipids complex, in atherosclerosis patients. Arteriosclerosis, Thrombosis, and Vascular Biology: Journal of the American Heart Association. 2004;24(5). [Google Scholar]
- 29.Sun M, Si L, Zhai X, Fan Z, Ma Y, Zhang R, et al. The influence of co-solvents on the stability and bioavailability of rapamycin formulated in self-microemulsifying drug delivery systems. Drug Dev Ind Pharm. 2011;37(8):986–94. [DOI] [PubMed] [Google Scholar]
- 30.Nofer J-R, Levkau B, Wolinska I, Junker R, Fobker M, von Eckardstein A, et al. Suppression of endothelial cell apoptosis by high density lipoproteins (HDL) and HDL-associated lysosphingolipids. Journal of Biological Chemistry. 2001;276(37):34480–5. [DOI] [PubMed] [Google Scholar]
- 31.Pan T, Rawal P, Wu Y, Xie W, Jankovic J, Le W. Rapamycin protects against rotenone-induced apoptosis through autophagy induction. Neuroscience. 2009;164(2):541–51. [DOI] [PubMed] [Google Scholar]
- 32.Demidenko ZN, Zubova SG, Bukreeva EI, Pospelov VA, Pospelova TV, Blagosklonny MV. Rapamycin decelerates cellular senescence. Cell cycle. 2009;8(12):1888–95. [DOI] [PubMed] [Google Scholar]
- 33.Nührenberg TG, Voisard R, Fahlisch F, Rudelius M, Braun J, Gschwend J, et al. Rapamycin attenuates vascular wall inflammation and progenitor cell promoters after angioplasty. The FASEB journal. 2005;19(2):1–21. [DOI] [PubMed] [Google Scholar]
- 34.Pikuleva IA, Curcio CA. Cholesterol in the retina: the best is yet to come. Progress in retinal and eye research. 2014;41:64–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jun S, Datta S, Wang L, Pegany R, Cano M, Handa JT. The impact of lipids, lipid oxidation, and inflammation on AMD, and the potential role of miRNAs on lipid metabolism in the RPE. Experimental eye research. 2019;181:346–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Xu Q, Cao S, Rajapakse S, Matsubara JA. Understanding AMD by analogy: systematic review of lipid-related common pathogenic mechanisms in AMD, AD, AS and GN. Lipids in Health and Disease. 2018;17(1):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kaarniranta K, Hyttinen J, Ryhanen T, Viiri J, Paimela T, Toropainen E, et al. Mechanisms of protein aggregation in the retinal pigment epithelial cells. Frontiers in bioscience (Elite edition). 2010;2:1374–84. [DOI] [PubMed] [Google Scholar]
- 38.Mitter SK, Rao HV, Qi X, Cai J, Sugrue A, Dunn WA, et al. Autophagy in the retina: a potential role in age-related macular degeneration. Retinal Degenerative Diseases: Springer; 2012. p. 83–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wilkinson JE, Burmeister L, Brooks SV, Chan CC, Friedline S, Harrison DE, et al. Rapamycin slows aging in mice. Aging cell. 2012;11(4):675–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rodríguez-Muela N, Germain F, Mariño G, Fitze PS, Boya P. Autophagy promotes survival of retinal ganglion cells after optic nerve axotomy in mice. Cell Death & Differentiation. 2012;19(1):162–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mizushima N, Yoshimori T. How to interpret LC3 immunoblotting. Autophagy. 2007;3(6):542–5. [DOI] [PubMed] [Google Scholar]
- 42.Storti F, Raphael G, Griesser V, Klee K, Drawnel F, Willburger C, et al. Regulated efflux of photoreceptor outer segment-derived cholesterol by human RPE cells. Experimental eye research. 2017;165:65–77. [DOI] [PubMed] [Google Scholar]
- 43.Schwendeman A, Sviridov DO, Yuan W, Guo Y, Morin EE, Yuan Y, et al. The effect of phospholipid composition of reconstituted HDL on its cholesterol efflux and anti-inflammatory properties. Journal of lipid research. 2015;56(9):1727–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Galvao J, Davis B, Tilley M, Normando E, Duchen MR, Cordeiro MF. Unexpected low-dose toxicity of the universal solvent DMSO. The FASEB Journal. 2014;28(3):1317–30. [DOI] [PubMed] [Google Scholar]
- 45.Xu QG, Boylan NJ, Suk JS, Wang YY, Nance EA, Yang JC, et al. Nanoparticle diffusion in, and microrheology of, the bovine vitreous ex vivo. Journal of Controlled Release. 2013;167(1):76–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sakurai E, Ozeki H, Kunou N, Ogura Y. Effect of particle size of polymeric nanospheres on intravitreal kinetics. Ophthalmic Res. 2001;33(1):31–6. [DOI] [PubMed] [Google Scholar]
- 47.Lyssenko NN, Haider N, Picataggi A, Cipollari E, Jiao W, Phillips MC, et al. Directional ABCA1-mediated cholesterol efflux and apoB-lipoprotein secretion in the retinal pigment epithelium. Journal of lipid research. 2018;59(10):1927–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ban N, Lee TJ, Sene A, Choudhary M, Lekwuwa M, Dong Z, et al. Impaired monocyte cholesterol clearance initiates age-related retinal degeneration and vision loss. JCI insight. 2018;3(17). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Buschini E, Piras A, Nuzzi R, Vercelli A. Age related macular degeneration and drusen: neuroinflammation in the retina. Progress in neurobiology. 2011;95(1):14–25. [DOI] [PubMed] [Google Scholar]
- 50.Rudolf M, Mir Mohi Sefat A, Miura Y, Tura A, Raasch W, Ranjbar M, et al. ApoA-I mimetic peptide 4F reduces age-related lipid deposition in murine Bruch’s membrane and causes its structural remodeling. Current eye research. 2018;43(1):135–46. [DOI] [PubMed] [Google Scholar]
- 51.Suda K, Murakami T, Gotoh N, Fukuda R, Hashida Y, Hashida M, et al. High-density lipoprotein mutant eye drops for the treatment of posterior eye diseases. Journal of Controlled Release. 2017;266:301–9. [DOI] [PubMed] [Google Scholar]
- 52.Storti F, Raphael G, Griesser V, Klee K, Drawnel F, Willburger C, et al. Regulated efflux of photoreceptor outer segment-derived cholesterol by human RPE cells. Experimental Eye Research. 2017;165:65. [DOI] [PubMed] [Google Scholar]
- 53.Sene A, Apte RS. Eyeballing cholesterol efflux and macrophage function in disease pathogenesis. Trends in Endocrinology & Metabolism. 2014;25(3):107–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Inana G, Murat C, An W, Yao X, Harris IR, Cao J. RPE phagocytic function declines in age-related macular degeneration and is rescued by human umbilical tissue derived cells. Journal of translational medicine. 2018;16(1):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Rami A. Autophagy in neurodegeneration: firefighter and/or incendiarist? Neuropathology and applied neurobiology. 2009;35(5):449–61. [DOI] [PubMed] [Google Scholar]
- 56.Mathew R, Karantza-Wadsworth V, White E. Role of autophagy in cancer. Nature Reviews Cancer. 2007;7(12):961–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Salminen A, Kaarniranta K, Kauppinen A. Inflammaging: disturbed interplay between autophagy and inflammasomes. Aging (Albany NY). 2012;4(3):166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wang S, Wang X, Cheng Y, Ouyang W, Sang X, Liu J, et al. Autophagy dysfunction, cellular senescence, and abnormal immune-inflammatory responses in AMD: from mechanisms to therapeutic potential. Oxidative medicine and cellular longevity. 2019;2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Zhu X, Owen JS, Wilson MD, Li H, Griffiths GL, Thomas MJ, et al. Macrophage ABCA1 reduces MyD88-dependent Toll-like receptor trafficking to lipid rafts by reduction of lipid raft cholesterol. Journal of lipid research. 2010;51(11):3196–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.De Nardo D, Labzin LI, Kono H, Seki R, Schmidt SV, Beyer M, et al. High-density lipoprotein mediates anti-inflammatory reprogramming of macrophages via the transcriptional regulator ATF3. Nature immunology. 2014;15(2):152–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wu W, He Z, Zhang Z, Yu X, Song Z, Li X. Intravitreal injection of rapamycin-loaded polymeric micelles for inhibition of ocular inflammation in rat model. International journal of pharmaceutics. 2016;513(1–2):238–46. [DOI] [PubMed] [Google Scholar]
- 62.Linares-Alba MA, Gómez-Guajardo MB, Fonzar JF, Brooks DE, García-Sánchez GA, Bernad-Bernad MJ. Preformulation studies of a liposomal formulation containing sirolimus for the treatment of dry eye disease. Journal of Ocular Pharmacology and Therapeutics. 2016;32(1):11–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Cholkar K, Gunda S, Earla R, Pal D, Mitra AK. Nanomicellar topical aqueous drop formulation of rapamycin for back-of-the-eye delivery. AAPS PharmSciTech. 2015;16(3):610–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Keyserling CH, Hunt TL, Klepp HM, Scott RA, Barbaras R, Schwendeman A, et al. CER-001, a synthetic HDL-mimetic, safely mobilizes cholesterol in healthy dyslipidemic volunteers. Am Heart Assoc; 2011. [Google Scholar]
- 65.Keyserling CH, Barbaras R, Benghozi R, Dasseux J-L. Development of CER-001: preclinical dose selection through to phase I clinical findings. Clinical drug investigation. 2017;37(5):483–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Dasseux J-l. Peptide/lipid complex formation by co-lyophilization. Google Patents; 2001. [Google Scholar]





