June 2023
Volume 64, Issue 7
Open Access
Retina  |   June 2023
Sortilin Inhibition Protects Neurons From Degeneration in the Diabetic Retina
Author Affiliations & Notes
  • Thomas Stax Jakobsen
    Department of Biomedicine, Aarhus University, Aarhus, Denmark
    Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
  • Jakob Appel Østergaard
    Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
    Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
  • Mads Kjolby
    Department of Biomedicine, Aarhus University, Aarhus, Denmark
    Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
    Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
  • Elisa Lund Birch
    Department of Biomedicine, Aarhus University, Aarhus, Denmark
  • Toke Bek
    Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
  • Anders Nykjaer
    Center for Proteins in Memory (PROMEMO) and Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
  • Thomas J. Corydon
    Department of Biomedicine, Aarhus University, Aarhus, Denmark
    Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
  • Anne Louise Askou
    Department of Biomedicine, Aarhus University, Aarhus, Denmark
    Department of Ophthalmology, Aarhus University Hospital, Aarhus, Denmark
  • Correspondence: Thomas J. Corydon, Department of Biomedicine, Aarhus University, Hoegh Guldbergs Gade 10, 8000 Aarhus C, Denmark; [email protected]
  • Anne Louise Askou, Department of Biomedicine, Aarhus University, Hoegh Guldbergs Gade 10, 8000 Aarhus C, Denmark; [email protected]
  • Footnotes
     TJC and ALA contributed equally.
Investigative Ophthalmology & Visual Science June 2023, Vol.64, 8. doi:https://doi.org/10.1167/iovs.64.7.8
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Thomas Stax Jakobsen, Jakob Appel Østergaard, Mads Kjolby, Elisa Lund Birch, Toke Bek, Anders Nykjaer, Thomas J. Corydon, Anne Louise Askou; Sortilin Inhibition Protects Neurons From Degeneration in the Diabetic Retina. Invest. Ophthalmol. Vis. Sci. 2023;64(7):8. https://doi.org/10.1167/iovs.64.7.8.

      Download citation file:


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

      ×
  • Supplements
Abstract

Purpose: To investigate the level and localization of the multifunctional receptor sortilin in the diabetic retina, as well as the effect of sortilin inhibition on retinal neurodegeneration in experimental diabetes.

Methods: The localization of sortilin and colocalization with the p75 neurotrophin receptor (p75NTR) and Müller cell (MC) markers were determined using immunofluorescence on retinal sections from human patients with diabetes and streptozotocin-induced diabetic C57BL/6J male mice. In the diabetic mice, levels were further quantified using Western blot and quantitative PCR. Therapeutic studies were performed on diabetic mice using intravitreally injected anti-sortilin antibodies. Neuroprotection was evaluated in vivo by optical coherence tomography and by quantification of retinal ganglion cells (RGCs) in flat mounts.

Results: Increased levels of sortilin were observed in human and murine diabetic retinas compared with nondiabetic control retinas. Sortilin was highly localized to retinal MCs, and, notably, colocalization with p75NTR was only seen in diabetic retinas. A remarkable protective effect of sortilin inhibition on inner retinal cells was observed in diabetic mice. At eight weeks after diabetes induction, inner retinal thickness was reduced by 9.7% (−12.7%, −6.6%; P < 0.0001; n = 11−12) in the PBS-injected control group compared with the anti-sortilin injected group. Similarly, the count of RGCs was reduced by 20.5% (−30.8%, −10.2%; P = 0.0009) in the PBS-injected control group compared with the anti-sortilin–injected group.

Conclusions: Sortilin is upregulated in the diabetic retina, and sortilin inhibition effectively protects against neuronal loss. Thus sortilin emerges as a novel pharmacological target in diabetic retinal neurodegeneration—an important early event in the pathogenesis of diabetic retinopathy.

Diabetic retinopathy (DR) is a leading cause of visual impairment in the Western world.1 The underlying pathology is complex2 and remains to be elucidated in detail. The hallmark of DR is changes in the microvasculature with loss of pericytes and formation of microaneurysms as early events.3,4 However, already decades ago, it was compellingly argued also to consider DR as a neurosensory disorder.5 Mounting evidence now supports that degeneration and dysfunction of retinal neurons occur in the course of DR and contribute to its development.6 Interestingly, dysfunction and loss of neuronal cells seem to be early events that may precede the vascular lesions characterizing the disease.7,8 
Sortilin9 is a multifunctional receptor belonging to the Vps10p domain receptor family. In addition to its important role as a sorting receptor, sortilin has emerged as an essential regulator of neurotrophin signaling.10 Neurotrophins are growth factors essential for neuronal development, integrity, and functionality. These growth factors are commonly secreted as pro-neurotrophins that as opposed to their mature counterparts are proapoptotic.11 As an example, the mature nerve growth factor (NGF) targets the tyrosine receptor kinase A in conjunction with the p75 neurotrophin receptor (p75NTR) for trophic signaling, whereas the immature pro-nerve growth factor (proNGF) binds with high affinity to a receptor complex comprising sortilin and p75NTR for induction of proapoptotic10 or proinflammatory12 pathways. Without the presence of sortilin, the proNGF/p75NTR interaction is of low affinity, and there is no proapoptotic signaling because the p75NTR/sortilin complex is not formed.9,10 ProNGF-induced apoptotic signaling through the p75NTR/sortilin complex has been shown to play a role in several neurodegenerative pathologies.13,14 In addition, the pleiotropic effect of sortilin as a sorting receptor has implicated it in diverse pathologies such as cardiovascular disease15 and carcinogenesis.16 
The potential role of sortilin in DR has not been investigated. However, diabetes mellitus (DM) has been shown to cause an imbalance of proNGF and NGF in the human retina,17 and animal studies have implicated disturbed neurotrophin signaling in retinal ganglion cell (RGC) degeneration in DR,18,19 optic nerve lesions and glaucoma20 through proNGF/p75NTR-mediated mechanisms. In addition, proNGF itself induces RGC death.12 Furthermore, postmitotic RGCs express sortilin13,21 and p75NTR13,22 when programmed RGC death peaks in the developing mouse retina. 
Thus we hypothesized that sortilin is upregulated in the diabetic retina and involved in the diabetes-induced loss of RGCs: The localization of sortilin and p75NTR was studied by immunofluorescence (IF) in retinal tissue obtained postmortem from six human donors with DR and five controls. Subsequently, sortilin expression and localization were evaluated in the retina of streptozotocin (STZ)-induced diabetic mice. Therapeutic studies using intravitreally (IVT)-injected anti-sortilin polyclonal blocking antibodies in diabetic mice were used to evaluate the neuroprotective effect of sortilin inhibition. 
Methods
Human Tissues
Six eyes from six patients with a verified history of DR and five eyes from five normal controls were studied (Table). The eyes had been selected among eyes obtained by autopsy between 1991 and 1998 to study vascular changes in DR.23 The retinal vascular system had been cast using the technique previously described.24,25 The eyes had been divided by horizontal section through the fovea and the optic disk, and one half had been embedded in paraffin as previously described.26 Clinical data at the time of casting are shown in the Table. The study using the previously collected tissue material was approved by the Regional Committee for Scientific Ethics, approval 1-10-72-299-12. 
Table.
 
Relevant Data of the Eyes Included in This Study Obtained at the Time of Examination
Table.
 
Relevant Data of the Eyes Included in This Study Obtained at the Time of Examination
Immunofluorescence in Human Retinal Sections
Tissue sections were cut 2 µm thick from the equator region of the eye and were incubated at 60°C for 60 minutes. Sections were deparaffinized in xylene overnight (ON) before rehydration with graded ethanol washes. Antigens were retrieved by incubation in Tris-EDTA buffer in a microwave oven for three minutes (maximum temperature 60°C), left to cool for >30 minutes at room temperature on a rocking table, and washed in PBS. Unspecific binding was reduced by incubation in 2% BSA in PBS for 10 minutes. The sections were incubated with the primary antibodies (see Supplementary Table S1) in 1% BSA in PBS ON at 4°C. The following day, sections were washed in PBS before incubation with the secondary antibodies (see Supplementary Table S1) in 1% BSA in PBS at room temperature for 30 minutes. Cell nuclei were stained with DAPI (Sigma-Aldrich, St. Louis, MO, USA), washed in water, dipped in 70% ethanol, and left to dry. Cover glasses were mounted using ProLong Gold Antifade mounting medium (ThermoFisher Scientific, Waltham, MA, USA). A control stain for smooth muscle actin (SMA), a protein found in retinal vessels, was used to assess the quality of the tissue (Supplementary Fig. S3). Negative controls were performed, where the primary antibody was omitted (Supplementary Fig. S4A). 
Fluorescence staining and colocalization were examined using a Zeiss LSM 710 confocal laser scanning microscope (Zeiss, Jena, Germany) fitted with a Plan-Apochromat 20 × 63 × 1.4 objective. Excitation and emission wavelengths for Alexa Fluor 488 were λex = 488 nm and λem 525 nm. Correspondingly for TRITC: λex = 532 nm and λem = 576 nm. SMA staining was visualized by fluorescence microscopy (Leitz DM RB; Leica Microsystems GmbH, Wetzlar, Germany) with a 40 × 1.0 objective, and images were captured with a Leica DFC 360 FX camera and associated software (Leica Application Suite, version 3). Images were captured using the same settings for Alexa Fluor 488 and Alexa Fluor 568; fluorescence intensities can be compared among images in the same figure. 
Quantification of Sortilin and p75NTR Levels in the Human Retina
Vertical densitometric profile plots representing the mean fluorescent signal intensity across the retinas in each group were generated using the Plot Profile function of ImageJ27 for both sortilin and p75NTR using the DAPI nuclear stain to determine the location in the retina. Three linear plot profiles were analyzed for each retinal image, and the mean fluorescent signal intensity was calculated. In each of the retinal images, eight different retinal layers were identified: Nerve fiber layer (NFL), ganglion cell layer (GCL), inner plexiform layer (IPL), inner nuclear layer, outer plexiform layer, outer nuclear layer, outer limiting membrane (OLM), and inner and outer segments of the photoreceptors. For each layer, the mean fluorescent signal intensity was calculated. Total fluorescence intensity of sortilin and p75NTR across the retina was calculated as the area under the linear plot profiles of the respective fluorescence intensities. For the SMA-stained sections, the mean fluorescent signal intensity from the NFL to the OLM were similarly determined using the Plot Profile function in ImageJ. 
Animals
The use of animals was approved by the Danish Animal Inspectorate (authorization no. 2012-15-2934-00113 and 2016-15-0201-00947). For diabetes induction, eight-week-old C57BL/6J male mice were purchased from Janvier Labs (Le Genest-Saint-Isle, France). Animals were maintained under a 12-/12-hour light/dark cycle at the Animal Facilities at the Department of Biomedicine, Aarhus University, Denmark with free access to autoclaved tap water and standard chow (Altromin, Brogaarden, Denmark). The animals were handled in accordance with the Association for Research in Vision and Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research. 
Before intravitreal injections or optical coherence tomography (OCT), mice were anesthetized with an intraperitoneal injection of a mixture of ketamine and medetomidine hydrochloride (Ketador 60–100 mg/kg [Richter Pharma AG, Wels, Austria] and Cepetor/Domitor 0.5–1 mg/kg [ScanVet Animal Health A/S, Fredensborg, Denmark]). Pupils were dilated with a drop of 1% tropicamide (Mydriacyl; Alcon Nordic A/S, Copenhagen, Denmark). During anesthesia, the eyes were lubricated with carbomer eye gel (Viscotears 2 mg/mL, Alcon Nordic). Immediately after procedures, anesthesia was reversed by atipamezole 0.5–1 mg/kg (Antisedan, Orion Pharma, Copenhagen, Denmark), and animals were placed on a warming plate until they moved spontaneously. 
Induction of Diabetes
Diabetes was induced in eight-week-old C57BL/6J male mice by five intraperitoneal STZ injections on five consecutive days using doses of 55 mg/kg body weight in 10 mM citrate buffer.28 Nondiabetic control mice were injected with citrate buffer only. Animals were fasted for four to six hours before the STZ injection. Body weight and blood glucose were measured weekly throughout the study. Blood glucose was measured in tail-capillary blood by Contour Next (Ascensia Diabetes Care, Copenhagen, Denmark). Animals were considered diabetic when blood glucose was >15 mmol/L. Mice were reinjected with two doses of 55 mg STZ/kg body weight intraperitoneally until diabetes was achieved. When blood glucose reached >15 mmol/L for all animals in the diabetic groups, the experimental period started. Animals with signs of illness or ketonuria (i.e., fast weight loss) were excluded. 
Intravitreal Injections of Anti-Sortilin Polyclonal Antibodies
At 2.5 weeks (T2.5) after the onset of diabetes, intravitreal injections were performed under direct visualization with an operating microscope (OPMI 1 FR PRO; Zeiss). A 30G disposable needle was used to puncture the sclera near the limbus, and a 33G blunt-ended needle of a Hamilton syringe (Hamilton Company, Reno, NV, USA) was then inserted into the opening and used to inject either antibody or buffer solution: The left eye of 20 diabetic mice was injected intravitreally with 2 µL of 1 mg/mL goat anti-sortilin polyclonal antibody (AF2934, R&D Systems, Minneapolis, MN, USA) and the right eye with 2 µL PBS. Twenty eyes from 10 age-matched nondiabetic mice were used as noninjected controls. 
OCT Imaging
At 2.5, four, six, and eight weeks after the onset of DM, the Micron IV image-guided OCT 2 system (Phoenix Research Laboratories, Pleasanton, CA, USA) was used to acquire B-scans in the central retina at an equal distance from the optic disc. The average thickness of the NGI (NFL, GCL, and IPL) complex was determined by loading the OCT images into InSight software (Phoenix Research Laboratories) and manually segmenting the retinal layers. 
Western Blotting
After sacrifice at 20 weeks of diabetes, eyes were extracted, and the retinas were processed for Western blotting as described previously.29,30 Membranes were incubated at 4°C ON with an anti-sortilin antibody or an anti-neuronal growth factor receptor antibody, and with an anti-glyceraldehyde-3-phosphate dehydrogenase antibody as loading control (see Supplementary Table S1). Visualization and quantification were done as described previously.29 Complete Western blots are provided as supplementary material (Supplementary Fig. S6). 
RT-qPCR Analyses
After sacrifice at 20 weeks of diabetes, eyes were enucleated and dropped in ice-cold Hanks’ balanced salt solution buffer. Periocular tissue was removed, and the anterior segment and lens were discarded. The neuroretina was dropped in ice-cold lysis buffer (buffer RLT + 40 mM Dithiothreitol). The tissue lysate was loaded onto a QIAshredder for homogenization, and, subsequently, RNA was purified using RNeasy plus kit, according to protocol (QIAGEN, Hilden, Germany). The iScript cDNA synthesis kit (Bio-Rad, Hercules, CA, USA) was used for first-strand cDNA synthesis, with a total input RNA of 50 ng. PCR reactions were performed in triplicate on 1:5 diluted cDNA using Taqman Gene expression assays and analyzed on a LightCyclerW480 (Roche Diagnostics, Basel, Switzerland) according to Taqman protocols. Relative gene expression of sortilin (Mm00490905; ThermoFisher Scientific) and p75NTR (Mm00446296; ThermoFisher Scientific) was calculated using the standard curve method and related relative to an endogenous control, the housekeeping gene hypoxanthine-guanine phosphoribosyl transferase (Mm00446968; ThermoFisher Scientific). 
Immunofluorescence and TUNEL Staining in Murine Retinal Sections
Eyes were enucleated and fixed in 4% paraformaldehyde ON at 4°C. Paraffin embedding and sectioning were performed at the Department of Pathology, Aarhus University Hospital. For immunofluorescence, sagittal sections of 3 µm were prepared, processed, and stained as described above. However, for anti-glial fibrillary acidic protein (GFAP) staining enzymatic retrieval by 10 minutes’ incubation with 20 µg/mL proteinase K solution (Promega, Madison, WI, USA) was performed according to manufacturer's instruction. The antibodies used are listed in Supplementary Table S1. Negative controls were performed, where the primary antibody was omitted (Supplementary Fig. S4B). TUNEL staining was performed using the DeadEnd Fluorometric TUNEL System (Promega). Pretreatment of the paraffin-embedded sections and TUNEL staining were performed according to the supplied protocol. 
Fluorescence signals were visualized by fluorescence microscopy (Leitz DM RB; Leica Microsystems GmbH) with a 16 × 0.5 or 40 × 1.0 objective and images captured with a Leica DFC 360 FX camera and associated software (Leica Application Suite, version 3). Images were captured using the same settings for Alexa Fluor 488 and Alexa Fluor 568; fluorescence intensities can be compared among images in the same figure. 
Quantification of Retinal Ganglion Cells in Flat Mounts
Eight weeks after the onset of diabetes, mice were sacrificed and eyes were enucleated and fixed in 4% paraformaldehyde for two hours at room temperature. The anterior segment and lens were removed and the neuroretina was carefully peeled off the retinal pigment epithelium (RPE)/choroid and immersed in PBS buffer. Retinas were permeabilized and blocked in 0.3% BSA and 1% Triton X-100 in PBS ON at 4°C on a shaker. Retinas were washed and stained with anti-Brn3a dissolved in 1% Triton X-100 in PBS buffer with 1 mM MgCl2, 1 mM CaCl2, 0.1 mM MnCl2 ON at 4°C on a shaker before being stained with secondary antibody dissolved in 0.3% BSA and 1% Triton X-100 in PBS for two hours at RT. Retinas were washed and stained with DAPI. Flat mounts were transferred to Super-FrostPlus glass slides and mounted with the RGC layer facing upward using ProLong Gold antifade reagent (ThermoFisher Scientific). Images for assessment of RGC densities were acquired 1 mm peripheral to the OD by fluorescence microscopy (Leitz DM RB; Leica Microsystems) and captured with a Leica DFC 360 FX camera and associated software (Leica Application Suite version 3, Leica Microsystems). Unfortunately, the quality of the Brn3A stain (Supplementary Fig. S4C) did not allow for quantification but validated the layer in which the RGCs were quantified. The number of RGCs was counted manually as neuronal cell nuclei in the GCL. RGCs were counted in three images from each retina, and a mean was calculated. 
Cell Culture and Immunofluorescence
Mouse neuroblastoma N2A (American Type Culture Collection, Manassas, VA, USA), HEK293 (American Type Culture Collection) and SH-SY5Y (American Type Culture Collection) cells were cultured in Dulbecco's modified Eagle's medium with GlutaMAX (ThermoFisher Scientific) supplemented with 10% fetal calf serum (Sigma-Aldrich) and 1% penicillin/streptomycin (ThermoFisher Scientific). The human Müller cell line Moorfields/Institute of Ophthalmology-Müller 1 (MIO-M1)31 was obtained from the UCL Institute of Ophthalmology, London, UK and cultured in Dulbecco's modified Eagle's medium with high glucose, GlutaMAX and pyruvate (Gibco, ThermoFisher Scientific) supplemented with 10% fetal calf serum (Sigma-Aldrich) and 1% penicillin/streptomycin (ThermoFisher Scientific). All cell lines were maintained in T-75 cell culture flasks (Sarstedt, Nümbrecht, Germany) at 37°C and 5% CO2. At near confluence, cells were seeded into SlideFlasks (ThermoFisher Scientific) and allowed to grow for 24 hours until 50% confluence and fixed using 10% neutral formalin. For transfection, after 24 hours in SlideFlasks, MIO-M1 cells were transfected with 1.5 µg plasmid DNA encoding sortilin (pAAV/apoE-hAAT-Sortilin) using XtremeGENE 9 (Roche) according to manufacturer's protocol and after 72 hours fixed using 10% neutral formalin. For the protein-blocking experiment, slides were washed in PBS followed by incubation for one hour at room temperature with primary antibody that had been preincubated for 30 minutes with or without recombinant murine sortilin (2934-ST; R&D Systems) in a 1:5 ratio. For sortilin immunofluorescence in HEK-293, MIO-M1 and SH-SY5Y, fixed slides were incubated in 70% ethanol followed by incubation with primary antibody for one hour at room temperature. Slides were washed in PBS and incubated with secondary antibody for one hour at room temperature. Finally, the slides were counterstained with DAPI before being mounted with cover glasses. Images were acquired by fluorescence microscopy (Leitz DM RB; Leica Microsystems) and captured with a Leica DFC 360 FX camera and associated software (Leica Application Suite version 3, Leica Microsystems). Fluorescence intensities can be compared among images in the same figure unless exposure times are stated. 
Single-Cell RNA-Seq Analysis
Processed single cell RNA sequencing (scRNA-seq) data from human32 and mouse33 retina samples was downloaded from, respectively, the Human Cell Atlas Data Portal (WongAdultRetina) and GEO (GSM4649095). Analyses were performed using Automated Single Cell Analysis Platform VI.34 The data were normalized using Seurat35 and dimensional reduction performed with UMAP.36 Clusters of cells were identified using Seurat37 and manually annotated using cell-type marker genes from Lukowski et al.32 
Statistical Analysis
Measurements are presented as mean ± SD. Data were evaluated for normality. Statistical differences between two group means were evaluated using a two-tailed Student t-test. Differences between multiple means were evaluated using ANOVA followed by post hoc comparisons using the Tukey method. A mean of the measurements from the two eyes of an animal were used if included in the same group. A significance level of α = 0.05 was used. Confidence intervals are presented accordingly. Statistical analyses were performed using the R statistical software package, version R-4.0.5 (https://www.r-project.org/). 
Results
Elevated Sortilin and p75NTR Levels in Patients With Diabetic Retinopathy
Sortilin and p75NTR were detectable in all human retinas examined using IF (Fig. 1A, Supplementary Fig. S1). Densitometric profile analysis of the fluorescent signals revealed significantly higher levels of sortilin and p75NTR in the retinas of patients with DM compared with controls (Fig. 1B). In human diabetic retinas, the fluorescent sortilin signal was strongest in the NFL, whereas it predominated in the GCL in nondiabetic control retinas (Fig. 1C). The highest p75NTR signal was observed in the OLM in diabetic retinas and in the NFL in nondiabetic control retinas (Fig. 1C). Notably, the sortilin and p75NTR signal covaried across the different layers of the diabetic retinas, with particular high intensities in inner retinal layers and corresponding to the OLM. 
Figure 1.
 
Increased levels and colocalization of sortilin and p75NTR in the human diabetic retina. Sortilin and p75NTR levels in retinas from patients with DR compared with control retinas determined by immunofluorescence and confocal microscopy analysis. (A) Paraffin sections from patients with DR D1, D4, and D6, and controls C1, C2, and C5 labeled with anti-sortilin (red) and anti-p75NTR (green). Colocalization is shown in the right column (yellow). Nuclei are stained with DAPI (blue). Scale bars: 20 µm. (B) Bar plots with mean ± SD of total fluorescence intensity of sortilin (red) and p75NTR (green) in retinas from patients with DR and controls. (C) Mean fluorescence intensities of sortilin (red) and p75NTR (green) in different retinal layers in eyes from patients with DR (D) compared with controls (C). Mean ± SD is shown. (D) Detail of the inner retina from patients D6 and C5. White arrows (D6) show sortilin at cell boundaries and protrusions, and gray arrows (C5) show sortilin in intracellular vesicles. Scale bars: 20 µm. INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; PR, inner and outer segments of the photoreceptors; D, patients with DM; C, controls; AU, arbitrary units. Significance levels: * [0.01, 0.05]; ** [0.001, 0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 1.
 
Increased levels and colocalization of sortilin and p75NTR in the human diabetic retina. Sortilin and p75NTR levels in retinas from patients with DR compared with control retinas determined by immunofluorescence and confocal microscopy analysis. (A) Paraffin sections from patients with DR D1, D4, and D6, and controls C1, C2, and C5 labeled with anti-sortilin (red) and anti-p75NTR (green). Colocalization is shown in the right column (yellow). Nuclei are stained with DAPI (blue). Scale bars: 20 µm. (B) Bar plots with mean ± SD of total fluorescence intensity of sortilin (red) and p75NTR (green) in retinas from patients with DR and controls. (C) Mean fluorescence intensities of sortilin (red) and p75NTR (green) in different retinal layers in eyes from patients with DR (D) compared with controls (C). Mean ± SD is shown. (D) Detail of the inner retina from patients D6 and C5. White arrows (D6) show sortilin at cell boundaries and protrusions, and gray arrows (C5) show sortilin in intracellular vesicles. Scale bars: 20 µm. INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; PR, inner and outer segments of the photoreceptors; D, patients with DM; C, controls; AU, arbitrary units. Significance levels: * [0.01, 0.05]; ** [0.001, 0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Sortilin Colocalizes With p75NTR and GFAP in Patients With Diabetic Retinopathy
In controls without DM, sortilin was predominately localized in the inner retina but was observed across retinal layers (Fig. 1A, Supplementary Fig. S1). Characteristically, sortilin showed a staining pattern compatible with localization to intracellular vesicles in retinal cells (Fig. 1D). In most cases, the p75NTR signal was low, and no colocalization was observed in any part of the nondiabetic retinas. In eyes from patients with DM, a clear shift in sortilin and p75NTR signal was observed. Both the sortilin and p75NTR signal were increased, and the signal was colocalized to thread-like structures spanning the retina (Fig. 1A, Supplementary Fig. S1). The sortilin signal was no longer limited to intracellular structures but was predominantly localized at cell boundaries and protrusions (Fig. 1D). 
The localization pattern of sortilin and p75NTR in the diabetic retinas was highly suggestive of expression in retinal Müller cells (MC). Co-immunostaining for a marker of reactive MCs, GFAP, and sortilin revealed a high degree of colocalization in the diabetic retinas (Fig. 2A, Supplementary Fig. S2). Noticeably, the colocalization signal spanned across the retina, but was most pronounced in the inner retina (Figs. 2A, 2B, Supplementary Fig. S2). The GFAP signal is expectedly lower in the nondiabetic retina. Thus additional co-immunostaining of sortilin and the MC-marker glutamine synthetase (GS) was performed in sections from controls without DM. Limited colocalization was observed in the inner retina (Fig. 2C). Thus sortilin levels increase in MCs in parallel with their development of a reactive phenotype during the development of DR. 
Figure 2.
 
Colocalization of sortilin and a Müller cell marker in human diabetic retinas. Localization of GFAP and sortilin in retinas from patients with DR compared with control retinas. (A) Paraffin sections of retinas from patients with DR (D1 and D6) and a control (C5) co-labeled with anti-sortilin (red), anti-GFAP (green), and DAPI (blue). Colocalization is shown in the right column (yellow). (B) High magnification of the inner retina from patients D6 and C5. Scale bars: 20 µm. (C) Colocalization of sortilin and GS in control retina. Paraffin section from control C1 labeled with anti-sortilin (red), anti-GS (green), and DAPI. Colocalization is shown in the right column (yellow); example is marked with white arrowhead. Scale bars: 20 µm. INL, inner nuclear layer; ONL, outer nuclear layer.
Figure 2.
 
Colocalization of sortilin and a Müller cell marker in human diabetic retinas. Localization of GFAP and sortilin in retinas from patients with DR compared with control retinas. (A) Paraffin sections of retinas from patients with DR (D1 and D6) and a control (C5) co-labeled with anti-sortilin (red), anti-GFAP (green), and DAPI (blue). Colocalization is shown in the right column (yellow). (B) High magnification of the inner retina from patients D6 and C5. Scale bars: 20 µm. (C) Colocalization of sortilin and GS in control retina. Paraffin section from control C1 labeled with anti-sortilin (red), anti-GS (green), and DAPI. Colocalization is shown in the right column (yellow); example is marked with white arrowhead. Scale bars: 20 µm. INL, inner nuclear layer; ONL, outer nuclear layer.
SMA was used as a positive control stain in all diabetic (D1-D6) and control (C1-C5) human retinas (Supplementary Fig. S3). No significant difference in background intensity was observed between the two groups (Supplementary Fig. S3). The negative controls revealed an autofluorescence signal corresponding to the RPE (Supplementary Fig. S4A). IF staining for sortilin was performed in cultured cells (Supplementary Fig. S5). As expected, sortilin was identified across the examined cell types including the human MC-line MIO-M1. 
Sortilin and p75NTR are Upregulated in the Retinas of STZ-Induced Diabetic Mice
Sortilin and p75NTR protein and mRNA levels were determined at 20 weeks after diabetes induction with STZ. Total protein levels of sortilin were increased by 58% (12%, 103%; P = 0.02; n = 5-6), and the total protein levels of p75NTR were increased by 138% (87%, 188%; P = 0.0004; n = 5-6) in diabetic murine retinas compared with nondiabetic retinas (Figs. 3A–D, Supplementary Fig. S6). A similar pattern was found on the mRNA level, where sortilin and p75NTR mRNA expression were increased by 84% (26%, 142%; P = 0.012; n = 4) and 194% (78%, 309%; P = 0.009; n = 4), respectively (Figs. 3E, 3F). Increased levels of sortilin and p75NTR were also observed using IF in retinal sections at 20 weeks (Fig. 4A). 
Figure 3.
 
Sortilin and p75NTR mRNA and protein levels are elevated in the eyes of diabetic mice. (A, B) Western blots of retinas from STZ-induced diabetic mice 20 weeks after the onset of DM and nondiabetic control mice. Bands corresponding to sortilin and p75NTR compared with GAPDH are shown. (C, D) Corresponding bar plots with mean ± SD sortilin and p75NTR protein levels compared with GAPDH from retinas of diabetic mice and control mice. (E) Bar plot with mean ± SD sortilin mRNA levels relative to HPRT evaluated by RT-qPCR. (F) Bar plot with mean ± SD p75NTR mRNA levels relative to HPRT evaluated by RT-qPCR. DM, Diabetes mellitus; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HPRT, hypoxanthine-guanine phosphoribosyl transferase. Significance levels: * [0.01, 0.05]; ** [0.001, 0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 3.
 
Sortilin and p75NTR mRNA and protein levels are elevated in the eyes of diabetic mice. (A, B) Western blots of retinas from STZ-induced diabetic mice 20 weeks after the onset of DM and nondiabetic control mice. Bands corresponding to sortilin and p75NTR compared with GAPDH are shown. (C, D) Corresponding bar plots with mean ± SD sortilin and p75NTR protein levels compared with GAPDH from retinas of diabetic mice and control mice. (E) Bar plot with mean ± SD sortilin mRNA levels relative to HPRT evaluated by RT-qPCR. (F) Bar plot with mean ± SD p75NTR mRNA levels relative to HPRT evaluated by RT-qPCR. DM, Diabetes mellitus; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HPRT, hypoxanthine-guanine phosphoribosyl transferase. Significance levels: * [0.01, 0.05]; ** [0.001, 0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 4.
 
Elevated levels of sortilin colocalize with p75NTR and a Müller cell marker in the retinas of diabetic mice. (A) Sortilin levels are increased and colocalize with p75NTR in the inner retina of diabetic mice. Paraffin sections from STZ-induced diabetic mice 20 weeks after the onset of diabetes and nondiabetic controls labeled with anti-sortilin (red) and anti-p75NTR (green). (B) Sortilin colocalizes with the Müller cell marker GS in diabetic retinas. Paraffin sections from mice after 20 weeks of diabetes and control mice labeled with anti-sortilin (red) and anti-GS (green). Colocalization is shown in the second rightmost column (yellow). Inserts to the right show magnification of the merged images. Examples of colocalization are indicated by white arrowheads. Scale bars: 20 µm. INL, inner nuclear layer; ONL, outer nuclear layer.
Figure 4.
 
Elevated levels of sortilin colocalize with p75NTR and a Müller cell marker in the retinas of diabetic mice. (A) Sortilin levels are increased and colocalize with p75NTR in the inner retina of diabetic mice. Paraffin sections from STZ-induced diabetic mice 20 weeks after the onset of diabetes and nondiabetic controls labeled with anti-sortilin (red) and anti-p75NTR (green). (B) Sortilin colocalizes with the Müller cell marker GS in diabetic retinas. Paraffin sections from mice after 20 weeks of diabetes and control mice labeled with anti-sortilin (red) and anti-GS (green). Colocalization is shown in the second rightmost column (yellow). Inserts to the right show magnification of the merged images. Examples of colocalization are indicated by white arrowheads. Scale bars: 20 µm. INL, inner nuclear layer; ONL, outer nuclear layer.
Sortilin is Expressed in MC in the Diabetic Murine Retina and Colocalizes With p75NTR
The localization of sortilin and p75NTR was evaluated by IF in retinal sections (Fig. 4A) from mice sacrificed at 20 weeks after diabetes induction. Sortilin and p75NTR were detectable in all murine retinas examined. Sortilin immunostaining was most prominent in the inner retina but spanned the entire retina (Fig. 4A). This pattern is comparable to the pattern observed in the human sections. Prominent sortilin and p75NTR colocalization was observed in the retina of diabetic mice, particularly in the inner retina (Fig. 4A, insert). The MC marker GS colocalized with sortilin. The colocalization was most evident in diabetic retinas with increased expression of sortilin (Fig. 4B, insert). The negative controls revealed an intense autofluorescence signal in photoreceptor outer segments and the RPE (Supplementary Fig. S4B). 
NGFR/Ngfr are Differentially Expressed Across Retinal Cell Populations in the Normal Human and Murine Retina
Analysis of scRNA-seq data from a transcriptome atlas of the adult human retina32 revealed almost uniform expression levels of sortilin (SORT1) across retinal cell populations. In contrast, p75NTR (NGFR) showed more varying expression with particular high expression levels in the MC population (Supplementary Fig. S7A). Similarly, analysis of scRNA-seq from the adult murine retina33 revealed expression levels of sortilin (Sort1) across retinal cell populations whereas p75NTR (Ngfr) showed high and relative uniform expression in MCs and to a lesser extent subpopulations of rods and bipolar cells (Supplementary Fig. S7B). 
Intravitreal Administration of Anti-Sortilin Antibodies Preserves Retinal Structure and Reduces RGC Death in Diabetic Mice
The effect of sortilin inhibition on diabetic retinal neurodegeneration was subsequently evaluated in STZ-induced diabetic mice (blood glucose and weight are provided in Supplementary Fig. S8C). Anti-sortilin antibodies or PBS were injected IVT 2.5 weeks (T2.5) after induction of DM (Fig. 5A). The ability of the antibody to neutralize sortilin was validated by a protein-blocking experiment in N2A cells; incubation of the anti-sortilin antibody with recombinant murine sortilin completely blocked sortilin immunostaining (Supplementary Fig. S9). No obvious group differences in sortilin levels using IF (Supplementary Fig. S10) were evident at eight weeks (T8). This suggests the primary action to result from ligand blocking rather than a general reduction of sortilin levels. 
As a common marker of glial reactivity, GFAP immunostaining was performed on retinal sections (n = 4) from eyes harvested at T8. An innermost retinal signal corresponding to retinal astrocytes was observed in all sections. However, an increased GFAP signal in the vertically oriented MC was evident in PBS-injected diabetic mice compared with both the anti-sortilin injected diabetic and the nondiabetic mice (Fig. 5B). 
Figure 5.
 
Intravitreal delivery of anti-sortilin antibody preserves retinal structure and retinal ganglion cells in diabetic mice. (A) Timeline of the experiment. DM was induced with five intraperitoneal injections of STZ. At 2.5 weeks after onset of DM (blood glucose > 15 mmol/L) mice were injected IVT with anti-sortilin polyclonal antibodies and PBS in opposite eyes. (B) Representative images of GFAP (red) and DAPI (blue) labelled paraffin-embedded retinal sections at eight weeks after onset of DM. Scale bar: 20 µm. (C) Representative OCT scans from nondiabetic control mice, IVT PBS-injected, and IVT anti-sortilin antibody-injected eyes at different time points. The white bar marks the combined thickness of the nerve fiber, retinal ganglion cell, and inner plexiform layer (NGI-thickness). (D) Box and dot plots of NGI-thickness; gray lines connect paired data points (i.e., eyes from the same animal). Significant comparisons between the groups are presented. (E) Representative images of DAPI stained retinal flat mounts. Scale bar: 50 µm. (F) Bar plots with mean ± SD of RGC density in retinal flat mounts from control mice, IVT PBS-injected and IVT anti-sortilin pAb-injected eyes. pAb, polyclonal antibody; INL, inner nuclear layer; ONL, outer nuclear layer. Significance levels: * [0.01, 0.05]; ** [0.001,0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 5.
 
Intravitreal delivery of anti-sortilin antibody preserves retinal structure and retinal ganglion cells in diabetic mice. (A) Timeline of the experiment. DM was induced with five intraperitoneal injections of STZ. At 2.5 weeks after onset of DM (blood glucose > 15 mmol/L) mice were injected IVT with anti-sortilin polyclonal antibodies and PBS in opposite eyes. (B) Representative images of GFAP (red) and DAPI (blue) labelled paraffin-embedded retinal sections at eight weeks after onset of DM. Scale bar: 20 µm. (C) Representative OCT scans from nondiabetic control mice, IVT PBS-injected, and IVT anti-sortilin antibody-injected eyes at different time points. The white bar marks the combined thickness of the nerve fiber, retinal ganglion cell, and inner plexiform layer (NGI-thickness). (D) Box and dot plots of NGI-thickness; gray lines connect paired data points (i.e., eyes from the same animal). Significant comparisons between the groups are presented. (E) Representative images of DAPI stained retinal flat mounts. Scale bar: 50 µm. (F) Bar plots with mean ± SD of RGC density in retinal flat mounts from control mice, IVT PBS-injected and IVT anti-sortilin pAb-injected eyes. pAb, polyclonal antibody; INL, inner nuclear layer; ONL, outer nuclear layer. Significance levels: * [0.01, 0.05]; ** [0.001,0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Mice were non-invasively monitored with OCT until sacrifice at T8 (Fig. 5A). Four weeks after onset of diabetes, inner retinal thickness as quantified by the combined thickness of the NFL, GCL, and inner IPL (i.e., the NGI) (Figs. 5C, 5D) was significantly reduced by 5.0 µm (−7.7, −2.3; P = 0.00053; n = 8) in the eyes of diabetic mice injected IVT with PBS compared with the eyes injected with anti-sortilin antibodies. At six weeks, a reduction by 6.7 µm (−10.0, −3.4; P = 0.00055; n = 4-5) was observed. The protective effect of anti-sortilin antibodies on NGI thickness was maintained until sacrifice at T8 (i.e., for at least 5.5 weeks). At this point, the NGI of diabetic anti-sortilin antibody IVT-injected mice measured 66.6 ± 1.66 µm compared with 60.1 ± 2.61 µm in diabetic PBS IVT-injected eyes, corresponding to a reduction by 6.4 µm (−8.5, −4.4; P < 0.0001; n = 11−12). From four weeks, highly significant differences comparable to those reported in the literature19,38,39 were observed between the PBS-injected group and the nondiabetic group. On the other hand, the NGI thickness in anti-sortilin antibody-injected eyes was comparable to nondiabetic mice with no significant difference at any time point further demonstrating the degree of rescue. IVT injection of anti-sortilin antibodies was well tolerated, as demonstrated further by fundus imaging (Supplementary Fig. S8A). No difference in outer nuclear layer thickness was observed between the treated group and the control group (P = 0.524; n = 8) (Supplementary Fig. S8B). 
After sacrifice at T8 (i.e., 5.5 weeks after treatment), RGCs were counted on retinal flat mounts (Figs. 5E, 5F). A reduction in the number of RGCs of 23.9% (−33.8%, −14.0%; P = 0.0002; n = 4) was found in the PBS-injected diabetic eyes compared with nondiabetic control mice, whereas the 4.3% (−14.2%, 5.6%) reduction in the anti-sortilin antibody-injected eyes was insignificant compared with nondiabetic control mice (P = 0.47; n = 4). The increased loss of RGCs in PBS-injected diabetic eyes compared with anti-sortilin injected diabetic eyes amounted to 20.5% (−30.8%, −10.2%; P = 0.0009). TUNEL-stained retinal sections of eyes harvested at T8 contained very limited number of positive cells (Supplementary Fig. S11), suggesting that the apoptotic process occurred at an earlier stage. 
Discussion
The multifunctional receptor sortilin has been investigated comprehensively, especially in the nervous system. Sortilin is believed to play a pivotal role in major human diseases involving neurodegeneration, but its role in the human eye remains elusive. In this study, immunostaining for sortilin in six patients with DR and five controls revealed (1) higher levels of sortilin in the diabetic retina, (2) sortilin colocalization with GFAP, (3) sortilin levels covarying with GFAP and p75NTR, and (4) sortilin colocalization with p75NTR in diabetic but not in nondiabetic retinas. Similar findings were demonstrated in diabetic murine retinas. Interestingly, the delivery of anti-sortilin antibodies rescued STZ-induced diabetic mice from inner retinal neurodegeneration. The almost-complete preservation of inner retinal structure with concomitant reduction of RGC death after sortilin-inhibition is highly suggestive of an indispensable role of sortilin in the neurodegenerative pathology of DR. 
P75NTR and its co-receptor sortilin are abundantly expressed in the brain and retina during development, but the expression is limited in healthy, adult tissue.13 In the nondiabetic human and murine retina, we similarly found low levels of both receptors compared with the diabetic retina. The intracellular localization of sortilin without colocalization with p75NTR is consistent with findings in the postnatal mouse retina where sortilin was detected in the Golgi apparatus, and no double-labeling with p75NTR was seen at postnatal day 10.21 Moreover, cell studies have shown that the majority of sortilin (∼90%) is found in the Golgi apparatus and vesicles, leaving only a minor fraction at the surface of the cells.40 
Sortilin has been implicated in light-induced photoreceptor death in adult rodents41 and retinal neuron death following elevated intraocular pressure-induced ischemia in rats.42 In these studies, retinal sortilin and p75NTR levels were both increased. However, in a study with STZ-induced diabetic rats, increased p75NTR expression was not associated with significantly different sortilin levels after 4-5 weeks of diabetes.43 In contrast, we demonstrated significantly increased retinal sortilin and p75NTR levels in both human patients with DM and STZ-induced diabetic mice at 20 weeks of diabetes. This difference could be explained by retinal sortilin accumulation during the course of DM. 
The retinal localization pattern of neurotrophins and their receptors is not completely delineated.44 In the adult rodent retina, p75NTR is predominately expressed in MCs,12, 45-47 as well as in photoreceptor cells41 and vascular cells.48 Notably, the expression in RGCs is debated.44 Sortilin expression has been reported in RGC,41,42,46 astrocytes,41,46 MCs46 and photoreceptor cells.41 In our study, sortilin similarly localized intracellularly across cell types in control retinas, while p75NTR showed a much more restricted distribution. Interestingly, similar findings were observed at the mRNA level when analyzing retinal scRNA-seq data of human and murine origin. Both sortilin and p75NTR localized to GFAP/GS positive cells corresponding to MCs in both the human and murine retina, and the diabetes-induced accumulation of sortilin and p75NTR was most pronounced in MC projections enveloping RGCs. Interestingly, the levels covaried with GFAP which is increased in activated MCs developing a gliotic phenotype in response to retinal insults.49,50 
In retinal insult models, p75NTR is overexpressed in MCs and not the cells that undergo apoptosis. Accordingly, RGC death through paracrine mechanisms has been demonstrated after proNGF stimulation of retinal glial cells in the ischemic retina,46 models of ocular proNGF overexpression,12 optic nerve lesions and glaucoma,20 and in animal models of STZ-induced diabetes.39 As an example, IVT injections of proNGF induced robust MC expression of TNFα in the murine eye, and the consequent RGC death was abolished in p75NTR and sortilin knockout mice.12 The observed colocalization of sortilin and p75NTR on MC projections suggests that the effects of sortilin are partly indirect by regulating signaling through p75NTR on MCs. This is further substantiated by the observation of reduced glial reactivity evaluated by GFAP IF after sortilin inhibition. The loss of pericytes and endothelial cells in DR may involve similar paracrine mechanisms or direct actions of proneurotrophins on the p75NTR/sortilin complex.48 MCs are essential for maintaining retinal homeostasis,49 and the dysfunction of MCs that are intercalated between the vasculature and neurons probably forms an important link between neuronal dysfunction and microvascular changes in the progression of DR.51 Indeed, the vascular occlusions in DR have been suggested to occur because of the invasion of vessels by gliotic MCs based on serial histological sections in our cohort of patients with DM.25,26 Although proneurotrophin signaling through the p75NTR/sortilin complex is the most obvious cause of the observed pathology, other sortilin-dependent mechanism might be involved. The cellular sorting of neurotrophic and neurotoxic factors and their receptors may be affected. As examples, sortilin is known to regulate the release of cytokines52 and exosomes53 in certain contexts. 
We observed a remarkable neuroprotective effect after IVT injection of anti-sortilin antibodies in diabetic mice. Targeting proNGF and p75NTR has similarly enabled the rescue of RGCs in rodent models of DM,19,39,54 optic nerve lesions,20 and ocular hypertension.20 Our findings suggest sortilin as a target for retinal neuroprotection and support the evidence for disturbed neurotrophin signaling in retinal disease, in particular as an early event in DR. Sortilin inhibition would protect retinal cells from proneurotrophin signaling through the p75NTR/sortilin complex while enabling unbound proneurotrophins to be converted into mature forms for trophic support. Importantly, this will not be the case when blocking p75NTR, which is crucial for trophic support by interacting with specific tyrosine receptor kinase-receptors.55 The rodent model used only partially recapitulates the clinical entity of DR and within a limited time frame. Thus the further elucidation of sortilin action in the diabetic retina requires an integrated view of the interactions between vascular and nonvascular cells, as well as the neuroglial interactions. Nonetheless, our findings strongly suggest that sortilin and its interaction with p75NTR is a target in DR and potentially other neurodegenerative pathologies of the retina. 
Acknowledgments
The authors thank Tina Hindkjær for expert technical assistance and Anne Kruse Hollensen for kindly providing the sortilin expression plasmid. Single cell RNA-seq analysis was performed by the Bioinformatics Core Facility at the Department of Biomedicine, Aarhus University, and the authors thank the staff at the core facility for their excellent support. 
Supported by the VELUX Foundation (ALA., Grant No. 17739), Gene Therapy Initiative Aarhus (GTI-Aarhus) funded by the Lundbeck Foundation (T.J.C., Grant No. R126-2012–12456), the Lundbeck Foundation (A.N., Grant Nos. R248-2017-431 and R315-2018-3066), Independent Research Fund Denmark (T.J.C., Grant No. 2034-00036B), Fight for Sight Denmark (T.S.J.), the Synoptik Foundation (T.S.J.), and Insusense Therapeutics. 
Disclosure: T.S. Jakobsen, None; J.A. Østergaard, None; M. Kjolby, Biogen (R); E.L. Birch, None; T. Bek, None; A. Nykjaer, None; T.J. Corydon, None; A.L. Askou, None 
References
Fong DS, Aiello L, Gardner TW, et al. Retinopathy in diabetes. Diabetes Care. 2004; 27: s84–s87. [CrossRef] [PubMed]
Antonetti DA, Silva PS, Stitt AW. Current understanding of the molecular and cellular pathology of diabetic retinopathy. Nat Rev Endocrinol. 2021; 17: 195–206. [CrossRef] [PubMed]
Cogan DG. Retinal vascular patterns. Arch Ophthalmol. 1961; 66: 366. [CrossRef] [PubMed]
Mizutani M, Kern TS, Lorenzi M. Accelerated death of retinal microvascular cells in human and experimental diabetic retinopathy. J Clin Invest. 1996; 97: 2883–2890. [CrossRef] [PubMed]
Bresnick GH. Diabetic retinopathy viewed as a neurosensory disorder. Arch Ophthalmol. 1986; 104: 989–990. [CrossRef] [PubMed]
Jonsson KB, Frydkjaer-Olsen U, Grauslund J. Vascular changes and neurodegeneration in the early stages of diabetic retinopathy: Which comes first? Ophthalmic Res. 2016; 56: 1–9. [CrossRef] [PubMed]
Eggers ED, Carreon TA. The effects of early diabetes on inner retinal neurons. Vis Neurosci. 2020; 37: E006. [CrossRef] [PubMed]
Sohn EH, Van Dijk HW, Jiao C, et al. Retinal neurodegeneration may precede microvascular changes characteristic of diabetic retinopathy in diabetes mellitus. Proc Natl Acad Sci. 2016; 113: E2655–E2664. [PubMed]
Nykjaer A, Willnow TE. Sortilin: A receptor to regulate neuronal viability and function. Trends Neurosci. 2012; 35: 261–270. [CrossRef] [PubMed]
Nykjaer A, Lee R, Teng KK, et al. Sortilin is essential for proNGF-induced neuronal cell death. Nature. 2004; 427: 843–848. [CrossRef] [PubMed]
Chao MV. Neurotrophins and their receptors: A convergence point for many signalling pathways. Nat Rev Neurosci. 2003; 4: 299–309. [CrossRef] [PubMed]
Lebrun-Julien F, Bertrand MJ, De Backer O, et al. ProNGF induces TNFα-dependent death of retinal ganglion cells through a p75NTR non-cell-autonomous signaling pathway. Proc Natl Acad Sci. 2010; 107: 3817–3822. [CrossRef] [PubMed]
Jansen P, Giehl K, Nyengaard JR, et al. Roles for the pro-neurotrophin receptor sortilin in neuronal development, aging and brain injury. Nat Neurosci. 2007; 10: 1449–1457. [CrossRef] [PubMed]
Carlo A-S, Nykjaer A, Willnow TE. Sorting receptor sortilin—a culprit in cardiovascular and neurological diseases. J Mol Med. 2014; 92: 905–911. [CrossRef] [PubMed]
Goettsch C, Kjolby M, Aikawa E. Sortilin and its multiple roles in cardiovascular and metabolic diseases. Arterioscler Thromb Vasc Biol. 2018; 38: 19–25. [CrossRef] [PubMed]
Rhost S, Hughes É, Harrison H, et al. Sortilin inhibition limits secretion-induced progranulin-dependent breast cancer progression and cancer stem cell expansion. Breast Cancer Res. 2018; 20: 137. [CrossRef] [PubMed]
Ali TK, Al-Gayyar MMH, Matragoon S, et al. Diabetes-induced peroxynitrite impairs the balance of pro-nerve growth factor and nerve growth factor, and causes neurovascular injury. Diabetologia. 2011; 54: 657–668. [CrossRef] [PubMed]
Ali TK, Matragoon S, Pillai BA, Liou GI, El-Remessy AB. Peroxynitrite mediates retinal neurodegeneration by inhibiting nerve growth factor survival signaling in experimental and human diabetes. Diabetes. 2008; 57: 889–898. [CrossRef] [PubMed]
Galan A, Barcelona PF, Nedev H, Sarunic MV, Jian Y, Saragovi HU. Subconjunctival delivery of p75NTR antagonists reduces the inflammatory, vascular, and neurodegenerative pathologies of diabetic retinopathy. Invest Ophthalmol Vis Sci. 2017; 58: 2852–2862. [CrossRef] [PubMed]
Bai Y, Dergham P, Nedev H, et al. Chronic and acute models of retinal neurodegeneration TrkA activity are neuroprotective whereas p75NTR activity is neurotoxic through a paracrine mechanism. J Biol Chem. 2010; 285: 39392–39400. [CrossRef] [PubMed]
Nakamura K, Namekata K, Harada C, Harada T. Intracellular sortilin expression pattern regulates proNGF-induced naturally occurring cell death during development. Cell Death Differ. 2007; 14: 1552–1554. [CrossRef] [PubMed]
Harada C, Harada T, Nakamura K, Sakai Y, Tanaka K, Parada LF. Effect of p75NTR on the regulation of naturally occurring cell death and retinal ganglion cell number in the mouse eye. Dev Biol. 2006; 290: 57–65. [CrossRef] [PubMed]
Bek T. Transretinal histopathological changes in capillary-free areas of diabetic retinopathy. Acta Ophthalmol (Copenh). 1994; 72: 409–415. [CrossRef] [PubMed]
Bek T, Jensen PK. Three-dimensional structure of human retinal vessels studied by vascular casting. Acta Ophthalmol (Copenh). 1993; 71: 506–513. [CrossRef] [PubMed]
Bek T. Glial cell involvement in vascular occlusion of diabetic retinopathy. Acta Ophthalmol Scand. 1997; 75: 239–243. [CrossRef] [PubMed]
Bek T. Immunohistochemical characterization of retinal glial cell changes in areas of vascular occlusion secondary to diabetic retinopathy. Acta Ophthalmol Scand. 1997; 75: 388–392. [CrossRef] [PubMed]
Schneider CA, Rasband WS, Eliceiri KW. NIH Image to ImageJ: 25 years of image analysis. Nat Methods. 2012; 9: 671–675. [CrossRef] [PubMed]
Tesch GH, Allen TJ. Rodent models of streptozotocin-induced diabetic nephropathy (Methods in Renal Research). Nephrology. 2007; 12: 261–266. [CrossRef] [PubMed]
Askou AL, Alsing S, Benckendorff JNE, et al. Suppression of Choroidal Neovascularization by AAV-Based Dual-Acting Antiangiogenic Gene Therapy. Mol Ther Nucleic Acids. 2019; 16: 38–50. [CrossRef] [PubMed]
Askou AL, Benckendorff JNE, Holmgaard A, et al. Suppression of choroidal neovascularization in mice by subretinal delivery of multigenic lentiviral vectors encoding anti-angiogenic microRNAs. Hum Gene Ther Methods. 2017; 28: 222–233. [CrossRef] [PubMed]
Limb GA, Salt TE, Munro PM, Moss SE, Khaw PT. In vitro characterization of a spontaneously immortalized human Müller cell line (MIO-M1). Invest Ophthalmol Vis Sci. 2002; 43: 864–869. [PubMed]
Lukowski SW, Lo CY, Sharov AA, et al. A single-cell transcriptome atlas of the adult human retina. Embo J. 2019; 38: e100811. [CrossRef] [PubMed]
Fadl BR, Brodie SA, Malasky M, et al. An optimized protocol for retina single-cell RNA sequencing. Mol Vis. 2020; 26: 705–717. [PubMed]
Gardeux V, David FPA, Shajkofci A, Schwalie PC, Deplancke B. ASAP: A web-based platform for the analysis and interactive visualization of single-cell RNA-seq data. Bioinformatics. 2017; 33: 3123–3125. [CrossRef] [PubMed]
Hao Y, Hao S, Andersen-Nissen E, et al. Integrated analysis of multimodal single-cell data. Cell. 2021; 184: 3573–3587.e3529. [CrossRef] [PubMed]
McInnes L, Healy J, Melville J. UMAP: Uniform manifold approximation and projection for dimension reduction. arXiv preprint arXiv:1802.03426. 2018 Feb 9.
Waltman L, Van Eck NJ. A smart local moving algorithm for large-scale modularity-based community detection. Eur Physical J B. 2013; 86: 1–14. [CrossRef]
Sergeys J, Etienne I, Van Hove I, et al. Longitudinal in vivo characterization of the streptozotocin-induced diabetic mouse model: Focus on early inner retinal responses. Invest Ophthalmol Vis Sci. 2019; 60: 807. [CrossRef] [PubMed]
Barcelona PF, Sitaras N, Galan A, et al. p75NTR and its ligand ProNGF activate paracrine mechanisms etiological to the vascular, inflammatory, and neurodegenerative pathologies of diabetic retinopathy. J Neurosci. 2016; 36: 8826–8841. [CrossRef] [PubMed]
Nielsen MS, Madsen P, Christensen EI, et al. The sortilin cytoplasmic tail conveys Golgi-endosome transport and binds the VHS domain of the GGA2 sorting protein. Embo J. 2001; 20: 2180–2190. [CrossRef] [PubMed]
Santos AM, López-Sánchez N, Martín-Oliva D, De La Villa P, Cuadros MA, Frade JM. Sortilin participates in light-dependent photoreceptor degeneration in vivo. PLoS ONE. 2012; 7: e36243. [CrossRef] [PubMed]
Wei Y, Wang N, Lu Q, Zhang N, Zheng D, Li J. Enhanced protein expressions of sortilin and p75NTR in retina of rat following elevated intraocular pressure-induced retinal ischemia. Neurosci Lett. 2007; 429: 169–174. [CrossRef] [PubMed]
Mysona BA, Al-Gayyar MMH, Matragoon S, et al. Modulation of p75NTR prevents diabetes- and proNGF-induced retinal inflammation and blood–retina barrier breakdown in mice and rats. Diabetologia. 2013; 56: 2329–2339. [CrossRef] [PubMed]
Garcia TB, Hollborn M, Bringmann A. Expression and signaling of NGF in the healthy and injured retina. Cytokine Growth Factor Revi. 2017; 34: 43–57. [CrossRef]
Hu B, Yip HK, So KF. Localization of p75 neurotrophin receptor in the retina of the adult SD rat: An immunocytochemical study at light and electron microscopic levels. Glia. 1998; 24: 187–197. [CrossRef] [PubMed]
Xu F, Wei Y, Lu Q, et al. Immunohistochemical localization of sortilin and p75(NTR) in normal and ischemic rat retina. Neurosci Lett. 2009; 454: 81–85. [CrossRef] [PubMed]
Harada T, Harada C, Nakayama N, et al. Modification of glial–neuronal cell interactions prevents photoreceptor apoptosis during light-induced retinal degeneration. Neuron. 2000; 26: 533–541. [CrossRef] [PubMed]
Shanab AY, Mysona BA, Matragoon S, El-Remessy AB. Silencing p75(NTR) prevents proNGF-induced endothelial cell death and development of acellular capillaries in rat retina. Mol Ther Methods Clin Dev. 2015; 2: 15013. [CrossRef] [PubMed]
Bringmann A, Pannicke T, Grosche J, et al. Müller cells in the healthy and diseased retina. Prog Retin Eye Res. 2006; 25: 397–424. [CrossRef] [PubMed]
Carpi-Santos R, de Melo Reis RA, Gomes FCA, Calaza KC. Contribution of Müller cells in the diabetic retinopathy development: Focus on oxidative stress and inflammation. Antioxidants (Basel). 2022; 11: 617. [CrossRef] [PubMed]
Coughlin BA, Feenstra DJ, Mohr S. Müller cells and diabetic retinopathy. Vision Res. 2017; 139: 93–100. [CrossRef] [PubMed]
Mortensen MB, Kjolby M, Gunnersen S, et al. Targeting sortilin in immune cells reduces proinflammatory cytokines and atherosclerosis. J Clin Invest. 2014; 124: 5317–5322. [CrossRef] [PubMed]
Wilson CM, Naves T, Vincent F, et al. Sortilin mediates the release and transfer of exosomes in concert with two tyrosine kinase receptors. J Cell Sci. 2014; 127: 3983–3997. [PubMed]
Barcelona PF, Galan A, Nedev H, Jian Y, Sarunic MV, Saragovi HU. The route of administration influences the therapeutic index of an anti-proNGF neutralizing mAb for experimental treatment of Diabetic Retinopathy. Plos One. 2018; 13: e0199079. [CrossRef] [PubMed]
Nykjaer A, Willnow TE, Petersen CM. P75NTR—live or let die. Curr Opin Neurobiol. 2005; 15: 49–57. [CrossRef] [PubMed]
Figure 1.
 
Increased levels and colocalization of sortilin and p75NTR in the human diabetic retina. Sortilin and p75NTR levels in retinas from patients with DR compared with control retinas determined by immunofluorescence and confocal microscopy analysis. (A) Paraffin sections from patients with DR D1, D4, and D6, and controls C1, C2, and C5 labeled with anti-sortilin (red) and anti-p75NTR (green). Colocalization is shown in the right column (yellow). Nuclei are stained with DAPI (blue). Scale bars: 20 µm. (B) Bar plots with mean ± SD of total fluorescence intensity of sortilin (red) and p75NTR (green) in retinas from patients with DR and controls. (C) Mean fluorescence intensities of sortilin (red) and p75NTR (green) in different retinal layers in eyes from patients with DR (D) compared with controls (C). Mean ± SD is shown. (D) Detail of the inner retina from patients D6 and C5. White arrows (D6) show sortilin at cell boundaries and protrusions, and gray arrows (C5) show sortilin in intracellular vesicles. Scale bars: 20 µm. INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; PR, inner and outer segments of the photoreceptors; D, patients with DM; C, controls; AU, arbitrary units. Significance levels: * [0.01, 0.05]; ** [0.001, 0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 1.
 
Increased levels and colocalization of sortilin and p75NTR in the human diabetic retina. Sortilin and p75NTR levels in retinas from patients with DR compared with control retinas determined by immunofluorescence and confocal microscopy analysis. (A) Paraffin sections from patients with DR D1, D4, and D6, and controls C1, C2, and C5 labeled with anti-sortilin (red) and anti-p75NTR (green). Colocalization is shown in the right column (yellow). Nuclei are stained with DAPI (blue). Scale bars: 20 µm. (B) Bar plots with mean ± SD of total fluorescence intensity of sortilin (red) and p75NTR (green) in retinas from patients with DR and controls. (C) Mean fluorescence intensities of sortilin (red) and p75NTR (green) in different retinal layers in eyes from patients with DR (D) compared with controls (C). Mean ± SD is shown. (D) Detail of the inner retina from patients D6 and C5. White arrows (D6) show sortilin at cell boundaries and protrusions, and gray arrows (C5) show sortilin in intracellular vesicles. Scale bars: 20 µm. INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; PR, inner and outer segments of the photoreceptors; D, patients with DM; C, controls; AU, arbitrary units. Significance levels: * [0.01, 0.05]; ** [0.001, 0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 2.
 
Colocalization of sortilin and a Müller cell marker in human diabetic retinas. Localization of GFAP and sortilin in retinas from patients with DR compared with control retinas. (A) Paraffin sections of retinas from patients with DR (D1 and D6) and a control (C5) co-labeled with anti-sortilin (red), anti-GFAP (green), and DAPI (blue). Colocalization is shown in the right column (yellow). (B) High magnification of the inner retina from patients D6 and C5. Scale bars: 20 µm. (C) Colocalization of sortilin and GS in control retina. Paraffin section from control C1 labeled with anti-sortilin (red), anti-GS (green), and DAPI. Colocalization is shown in the right column (yellow); example is marked with white arrowhead. Scale bars: 20 µm. INL, inner nuclear layer; ONL, outer nuclear layer.
Figure 2.
 
Colocalization of sortilin and a Müller cell marker in human diabetic retinas. Localization of GFAP and sortilin in retinas from patients with DR compared with control retinas. (A) Paraffin sections of retinas from patients with DR (D1 and D6) and a control (C5) co-labeled with anti-sortilin (red), anti-GFAP (green), and DAPI (blue). Colocalization is shown in the right column (yellow). (B) High magnification of the inner retina from patients D6 and C5. Scale bars: 20 µm. (C) Colocalization of sortilin and GS in control retina. Paraffin section from control C1 labeled with anti-sortilin (red), anti-GS (green), and DAPI. Colocalization is shown in the right column (yellow); example is marked with white arrowhead. Scale bars: 20 µm. INL, inner nuclear layer; ONL, outer nuclear layer.
Figure 3.
 
Sortilin and p75NTR mRNA and protein levels are elevated in the eyes of diabetic mice. (A, B) Western blots of retinas from STZ-induced diabetic mice 20 weeks after the onset of DM and nondiabetic control mice. Bands corresponding to sortilin and p75NTR compared with GAPDH are shown. (C, D) Corresponding bar plots with mean ± SD sortilin and p75NTR protein levels compared with GAPDH from retinas of diabetic mice and control mice. (E) Bar plot with mean ± SD sortilin mRNA levels relative to HPRT evaluated by RT-qPCR. (F) Bar plot with mean ± SD p75NTR mRNA levels relative to HPRT evaluated by RT-qPCR. DM, Diabetes mellitus; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HPRT, hypoxanthine-guanine phosphoribosyl transferase. Significance levels: * [0.01, 0.05]; ** [0.001, 0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 3.
 
Sortilin and p75NTR mRNA and protein levels are elevated in the eyes of diabetic mice. (A, B) Western blots of retinas from STZ-induced diabetic mice 20 weeks after the onset of DM and nondiabetic control mice. Bands corresponding to sortilin and p75NTR compared with GAPDH are shown. (C, D) Corresponding bar plots with mean ± SD sortilin and p75NTR protein levels compared with GAPDH from retinas of diabetic mice and control mice. (E) Bar plot with mean ± SD sortilin mRNA levels relative to HPRT evaluated by RT-qPCR. (F) Bar plot with mean ± SD p75NTR mRNA levels relative to HPRT evaluated by RT-qPCR. DM, Diabetes mellitus; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HPRT, hypoxanthine-guanine phosphoribosyl transferase. Significance levels: * [0.01, 0.05]; ** [0.001, 0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 4.
 
Elevated levels of sortilin colocalize with p75NTR and a Müller cell marker in the retinas of diabetic mice. (A) Sortilin levels are increased and colocalize with p75NTR in the inner retina of diabetic mice. Paraffin sections from STZ-induced diabetic mice 20 weeks after the onset of diabetes and nondiabetic controls labeled with anti-sortilin (red) and anti-p75NTR (green). (B) Sortilin colocalizes with the Müller cell marker GS in diabetic retinas. Paraffin sections from mice after 20 weeks of diabetes and control mice labeled with anti-sortilin (red) and anti-GS (green). Colocalization is shown in the second rightmost column (yellow). Inserts to the right show magnification of the merged images. Examples of colocalization are indicated by white arrowheads. Scale bars: 20 µm. INL, inner nuclear layer; ONL, outer nuclear layer.
Figure 4.
 
Elevated levels of sortilin colocalize with p75NTR and a Müller cell marker in the retinas of diabetic mice. (A) Sortilin levels are increased and colocalize with p75NTR in the inner retina of diabetic mice. Paraffin sections from STZ-induced diabetic mice 20 weeks after the onset of diabetes and nondiabetic controls labeled with anti-sortilin (red) and anti-p75NTR (green). (B) Sortilin colocalizes with the Müller cell marker GS in diabetic retinas. Paraffin sections from mice after 20 weeks of diabetes and control mice labeled with anti-sortilin (red) and anti-GS (green). Colocalization is shown in the second rightmost column (yellow). Inserts to the right show magnification of the merged images. Examples of colocalization are indicated by white arrowheads. Scale bars: 20 µm. INL, inner nuclear layer; ONL, outer nuclear layer.
Figure 5.
 
Intravitreal delivery of anti-sortilin antibody preserves retinal structure and retinal ganglion cells in diabetic mice. (A) Timeline of the experiment. DM was induced with five intraperitoneal injections of STZ. At 2.5 weeks after onset of DM (blood glucose > 15 mmol/L) mice were injected IVT with anti-sortilin polyclonal antibodies and PBS in opposite eyes. (B) Representative images of GFAP (red) and DAPI (blue) labelled paraffin-embedded retinal sections at eight weeks after onset of DM. Scale bar: 20 µm. (C) Representative OCT scans from nondiabetic control mice, IVT PBS-injected, and IVT anti-sortilin antibody-injected eyes at different time points. The white bar marks the combined thickness of the nerve fiber, retinal ganglion cell, and inner plexiform layer (NGI-thickness). (D) Box and dot plots of NGI-thickness; gray lines connect paired data points (i.e., eyes from the same animal). Significant comparisons between the groups are presented. (E) Representative images of DAPI stained retinal flat mounts. Scale bar: 50 µm. (F) Bar plots with mean ± SD of RGC density in retinal flat mounts from control mice, IVT PBS-injected and IVT anti-sortilin pAb-injected eyes. pAb, polyclonal antibody; INL, inner nuclear layer; ONL, outer nuclear layer. Significance levels: * [0.01, 0.05]; ** [0.001,0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Figure 5.
 
Intravitreal delivery of anti-sortilin antibody preserves retinal structure and retinal ganglion cells in diabetic mice. (A) Timeline of the experiment. DM was induced with five intraperitoneal injections of STZ. At 2.5 weeks after onset of DM (blood glucose > 15 mmol/L) mice were injected IVT with anti-sortilin polyclonal antibodies and PBS in opposite eyes. (B) Representative images of GFAP (red) and DAPI (blue) labelled paraffin-embedded retinal sections at eight weeks after onset of DM. Scale bar: 20 µm. (C) Representative OCT scans from nondiabetic control mice, IVT PBS-injected, and IVT anti-sortilin antibody-injected eyes at different time points. The white bar marks the combined thickness of the nerve fiber, retinal ganglion cell, and inner plexiform layer (NGI-thickness). (D) Box and dot plots of NGI-thickness; gray lines connect paired data points (i.e., eyes from the same animal). Significant comparisons between the groups are presented. (E) Representative images of DAPI stained retinal flat mounts. Scale bar: 50 µm. (F) Bar plots with mean ± SD of RGC density in retinal flat mounts from control mice, IVT PBS-injected and IVT anti-sortilin pAb-injected eyes. pAb, polyclonal antibody; INL, inner nuclear layer; ONL, outer nuclear layer. Significance levels: * [0.01, 0.05]; ** [0.001,0.01]; *** [0.0001, 0.001]; **** [0, 0.0001].
Table.
 
Relevant Data of the Eyes Included in This Study Obtained at the Time of Examination
Table.
 
Relevant Data of the Eyes Included in This Study Obtained at the Time of Examination
×
×

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.

×