IMR Press / FBL / Volume 28 / Issue 4 / DOI: 10.31083/j.fbl2804083
Open Access Original Research
Comparing and Contrasting the Effects of the SGLT Inhibitors Canagliflozin and Empagliflozin on the Progression of Retinopathy
Show Less
1 Dobney Hypertension Centre, School of Biomedical Sciences – Royal Perth Hospital Unit, University of Western Australia, Crawley, WA 6009, Australia
2 Department of Molecular Ophthalmology, University of Western Australia, Crawley, WA 6009, Australia
3 Dobney Hypertension Centre, Medical School – Royal Perth Hospital Unit, University of Western Australia, Crawley, WA 6009, Australia
4 Department of Cardiology and Department of Nephrology, Royal Perth Hospital, Perth, WA 6000, Australia
*Correspondence: vance.matthews@uwa.edu.au (Vance B. Matthews)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(4), 83; https://doi.org/10.31083/j.fbl2804083
Submitted: 15 February 2023 | Revised: 5 April 2023 | Accepted: 7 April 2023 | Published: 27 April 2023
(This article belongs to the Special Issue An Update on Sodium Glucose Co-Transporters)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Diabetic retinopathy (DR) is a leading cause of end-stage blindness globally and is arguably one of the most disabling complications of both Type 1 and Type 2 diabetes. Sodium Glucose Cotransporter-2 (SGLT2) inhibitors have now been successfully introduced to clinical medicine and exert multiple beneficial effects in diabetic patients. Given the broad therapeutic application of SGLT2 inhibitors, we hypothesised that SGLT2 inhibition may alleviate the progression of DR. Therefore, we aimed to compare the effectiveness of two clinically available SGLT2 inhibitors, Empagliflozin and Canagliflozin, on the progression of Retinopathy and DR using well-characterised mouse models, Kimba and Akimba, respectively. Methods: Empagliflozin, Canagliflozin (25 mg/kg/day) or vehicle was administered to 10-week-old mice via drinking water for 8-weeks. Urine glucose levels were measured to ascertain SGLT2 inhibition promoted glucose excretion. Weekly body weight and water intake measurements were obtained. After 8-weeks of treatment, body weight, daily water intake, fasting blood glucose levels were measured and eye tissue was harvested. The retinal vasculature was assessed using immunofluorescence. Results: Empagliflozin treated Akimba mice exhibited metabolic benefits suggested by healthy body weight gain and significantly reduced fasting blood glucose levels. Treatment with Empagliflozin reduced retinal vascular lesions in both Kimba and Akimba mice. Canagliflozin improved body weight gain, reduced blood glucose levels in Akimba mice, and reduced the development of retinal vascular lesions in Kimba mice. Conclusions: Our data demonstrates that Empagliflozin has future potential as a therapeutic for Retinopathy and DR and should now be considered for human trials.

Keywords
diabetic retinopathy
diabetes
Akimba
Kimba
mouse models
sodium glucose cotransporters
SGLT2 inhibitors
empagliflozin
canagliflozin
retinal vasculature
1. Introduction

Diabetic retinopathy (DR) is recognised as one of the most prevalent chronic microvascular complications of both Type 1 [1, 2] and Type 2 diabetes [3, 4] and is one of the leading causes of vision impairment globally [5, 6]. The classification of DR is primarily based on the changes that occur in the pre-existing retinal microvasculature and presence or absence of retinal neovascularisation [7, 8]. Diabetic retinopathy is divided into two principal stages. The early Non-Proliferative Diabetic Retinopathy (NPDR) stage is characterized by the presence of microaneurysms and the more advanced stage Proliferative Diabetic Retinopathy (PDR) is characterised by neovascularisation which may eventually lead to vision loss [7, 8].

It is shown that intensive therapy leading to near-normal blood glucose levels reduces the likelihood of developing retinopathy [9]. Beyond glycemic control, most treatment options such as anti-vascular endothelial growth factor (VEGF) injections, panretinal laser photocoagulation and vitrectomy largely target the late PDR stage where vision loss occurs [10]. Therefore, therapies targeting the potentially reversible early pathogenic stage of DR is urgently needed to prevent the onset or slow the progression of DR and eventual vision loss.

The kidneys play an important role in maintaining glucose homeostasis. Sodium glucose co-transporter 2 (SGLT2) is a high-capacity, low affinity glucose co-transport protein which helps to reabsorb about 90–95% of glucose in the S1 and S2 segments of the renal proximal tubular epithelial cells [11]. Interestingly, a new class of anti-diabetic drugs act by inhibition of SGLT2, thereby decreasing the reabsorption of glucose from the renal proximal tubules. Hence, specific SGLT2 inhibition facilitates the excretion of glucose via urine (glucosuria), resulting in reduced plasma glucose levels and improved glycaemic parameters [11]. The SGLT2 inhibitors have been shown to reduce HbA1c, fasting plasma glucose levels, blood pressure, and body weight [12, 13]. Additionally, it has been shown that SGLT2 inhibitors have protective effects in cardiovascular and kidney disease, which are two of the main complications of diabetes [14]. Nevertheless, systematic studies in both pre-clinical and clinical settings, to determine the effect of SGLT2 inhibition on retinal vascular disorders such as DR is limited [15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25]. With respect to the eye, retinal pericytes are identified as a novel source of SGLT2 expression [17, 26]. Furthermore, we have shown elevated levels of Sglt2 mRNA in the whole eye and SGLT2 protein expression in retinal ganglion cells [25]. Leley et al. [27] has also shown a statistically significant increase in Sglt2 mRNA levels in the retina of diabetic mice. In an attempt to elucidate the effect of SGLT2 inhibition on the development of DR, we have shown for the first time that the treatment with Empagliflozin (EMPA) profoundly reduced retinal vascular abnormalities such as capillary dropout, vessel tortuosity, microaneurysms, intraretinal microvascular abnormalities (IRMA) and neovascular tufts in young diabetic Akimba mice (4–5 weeks postnatal) [25]. However, thus far, the effect of SGLT2 inhibition on the progression of DR has not been examined. In addition, to the best of our knowledge there is no data comparing individual SGLT2 inhibitors in the context of DR, and therefore, it remains unclear whether a class effect of SGLT2 inhibitors on DR pathogenesis is present. It is reported that the possible difference in pharmacological effects and outcomes between different SGLT2 inhibitors is particularly due to the degree of SGLT2 selectivity [28, 29, 30]. Given the extensive use of SGLT2 inhibitors in clinical practice, particularly for the treatment of cardiovascular and kidney disease, it is important to compare the effect of different SGLT2 inhibitors on features of DR.

Here we report the first in vivo study comparing the two independent SGLT2 inhibitors, EMPA and Canagliflozin (CANA), on the progression of retinopathy and DR in the following well-established mouse models: (1) Kimba and (2) Akimba. Importantly, these models capture an array of hallmark features seen in human DR and therefore our findings are likely to have clinical relevance [31, 32, 33, 34, 35, 36].

2. Materials and Methods
2.1 Mice and Genotyping

Specific pathogen free 10-wk-old male Kimba and Akimba mice [32, 35, 36, 37] were bred and obtained from the Animal Resources Centre (ARC, Perth, Australia). These models are highly relevant for the study of retinal vascular disease [15, 18, 31, 32, 33]. In our study, we investigated 10-week-old animals with the aim of evaluating the effect of EMPA and CANA on the progression of retinopathy (Kimba model) and DR (Akimba model). Only male mice were used for these studies as disease progression in females is slower and inconsistent [32, 37]. Genotyping of Kimba and Akimba mice were performed as published previously [33, 37, 38].

2.2 Animal Experiments

The animal handling and experimental procedures were conducted at the animal holding facility of the Harry Perkins Institute for Medical Research in Perth, Western Australia, following the guidelines of the Institutional Animal Care and Use Committee. The study was approved by the Harry Perkins Institute for Medical Research Animal Ethics Committee (AE141/2019 approved: 12/02/19). All procedures were carried out in compliance with the Association for Research in Vision and Ophthalmology (ARVO) statement for the use of animals in ophthalmic and vision research.

All animals were acclimatised for one week. Urine glucose levels were obtained (Keta-Diabur-Test 5000, Roche Diagnostics, Leverkusen, Germany) prior to treatment to confirm the diabetic status of Akimba mice and the non-diabetic status of Kimba mice. The 10-week-old mice were housed individually under a 12-hour light/dark cycle and maintained at a temperature of 21 ± 2 °C. Animals were provided free access to a standard Chow diet (Specialty Feeds, Glen Forrest, Western Australia, Australia) and drinking water containing the SGLT2 inhibitor (CANA or EMPA) at a dose of 25 mg/kg (Ark Pharma Scientific Limited, Wuhan, China) or vehicle Dimethylsulfoxide (DMSO; Sigma-Aldrich, Sydney, Australia). The mice were treated for 8 weeks. Drinking water containing CANA, EMPA or vehicle was freshly prepared and replenished on a weekly basis.

2.3 Measurement of Metabolic Parameters

Body weights, fluid intake and urine glucose levels (Keta-Diabur-Test 5000, Roche Diagnostics, Leverkusen, Germany) were recorded weekly. At the end of experiment, mice were fasted for 5 hours with free access to treated drinking water. Fasting blood glucose levels were measured using Accu-Chek Performa blood glucose monitoring system (Roche Diagnostics, North Ryde, Australia) with a range of 0.6 to 33.3 mmol/L. Readings over 33.3 mmol/L were treated as 33.3 mmol/L during data analysis.

2.4 Specimen Collection

At the experimental endpoint, all animals were deeply anesthetised with isoflurane inhalation and were euthanised by cervical dislocation. For retinal vascular assessment, eyes were enucleated, cornea was penetrated with a 27G insulin needle and was fixed in ice-cold 10% buffered paraformaldehyde (PFA; Sigma-Aldrich, Sydney, Australia) for 2 hours at 4 °C.

2.5 Retinal Isolation, Whole-Mount Preparation and Immunofluorescence

Retinal whole-mounts were prepared and analysed as previously published by our team [15, 25]. In brief, fixed eyes were washed in ice cold 1×PBS and retinas were dissected and cleaned. Fixed retinas were permeabilized for 2 hours in PBS-BSA-Triton-X (1×PBS, 1% Bovine Serum Albumin (BSA), 1% Triton-X) at room temperature and washed in Wash Buffer (1×PBS, 0.5% Triton-X). Retinas were incubated overnight in the dark at 4 °C with Isolectin-B4 (1:100 in Wash Buffer; Griffonia Simplicifolia Lectin I (GSL I) isolectin B4, Biotinylated; Catalog#: B-1205-.5; Vector Laboratories, Burlingame, CA, USA). Retinas were washed in 1×PBS and incubated for five hours in the dark at room temperature with Cy3-Streptavidin (1:500 in 1×PBS; GE Healthcare, Amersham, UK). Retinas were washed in 1×PBS and mounted with VECTASHIELD HardSet Anti-fade Mounting Medium (Vector Laboratories, Burlingame, CA, USA).

2.6 Retinal Whole-Mount Imaging and Analysis

Whole-mounted retinas were imaged with an inverted fluorescent microscope (Nikon Eclipse Ti, Nikon, Tokyo, Japan) equipped with a digital camera (CoolSNAP HQ2, Photometrics, Tucson, AZ, USA) linked to a computer running the image analysis software ‘NIS-Elements Advanced Research’ (Nikon, Tokyo, Japan). Serially overlapping high-resolution images of the whole retina were captured using the 4× objective. Captured frames were merged to generate a montage of the full retina. Montages were semi-quantified for vascular morphological abnormalities with minor modifications to the parameters as published previously [15, 25]. These modifications were incorporated to capture the advanced retinal phenotype observed in both Kimba and Akimba mice due to disease progression. In addition, retinal vascular features such as microaneurysms, IRMA, exceptionally dense vascular budding, monocytes/macrophages and neovascular tufts (all structural abnormalities intensely showing lectin positivity) were counted using the NIS-Elements Advanced Research software (Nikon, Tokyo, Japan) within 2000 μm radius from the optic nerve head in low magnification photo montages. Z-stacks (2 μm steps) were compiled into a focused image representing all three vascular layers of the retina (superficial, intermediate and deep capillary layers).

2.7 Statistical Analysis

All morphometric data were analyzed using two-tailed Student’s t-test or one-way analysis of variance (ANOVA) and graphs were produced using GraphPad Prism 9 (GraphPad Software Inc., San Diego, CA, USA). Quantitative data is presented as mean ± SEM and p 0.05 was deemed statistically significant.

3. Results
3.1 SGLT2 Inhibition Promotes Glucosuria in Non-Diabetic Kimba Mice

Testing of urine glucose levels during the acclimatisation period confirmed the genotype of each strain. The non-diabetic Kimba mice did not show the presence of glucose in their urine while the diabetic Akimba mice showed the presence of glucose indicated by the brown colour (111 mmol/L) in the testing strips (data not shown).

The Kimba model lacks the hyperglycemic background. Therefore, Kimba mice act as an internal control for the determination of the effectiveness of the SGLT2 inhibitors EMPA and CANA used in our study. As anticipated, the consumption of EMPA (Fig. 1A) and CANA (Fig. 1B) in drinking water at a concentration of 25 mg/kg/day, showed marked glucose excretion in the urine of the non-diabetic Kimba mice as indicated by the brown colour (111 mmol/L). All vehicle treated mice showed a reading of 0 mmol/L of glucose in their urine indicated by the light blue colour (Fig. 1A,B).

Fig. 1.

SGLT2 inhibition promotes glucosuria in Non-Diabetic Kimba mice. Representative image of urine glucose levels in (A) EMPA (25 mg/kg) and (B) CANA (25 mg/kg) treated mice. Blue = 0 mmol/L glucose; Dark brown 111 mmol/L glucose; SGLT2, Sodium glucose cotransporter 2; EMPA, Empagliflozin; CANA, Canagliflozin.

3.2 Effects of EMPA and CANA on Blood Glucose Levels in Kimba and Akimba Mice

At the end of treatment (after 8 weeks), relative to vehicle treated mice, EMPA or CANA did not significantly alter fasting blood glucose levels in Kimba mice (Fig. 2A,C, respectively). However, fasting blood glucose levels in diabetic Akimba mice were significantly reduced after 8 weeks of EMPA (Fig. 2B) or CANA (Fig. 2D) administration. Interestingly, when compared to EMPA (Vehicle 32.67 ± 0.6 mmol/L vs EMPA 21.9 ± 1 mmol/L; p = 0.00044), CANA showed a greater reduction in fasting blood glucose levels (Vehicle 32.46 ± 0.5 mmol/L versus CANA 17.43 ± 1.3 mmol/L; p = 0.0000039).

Fig. 2.

Role of SGLT2 inhibition on fasting blood glucose in Kimba and Akimba mice treated with EMPA or CANA. Graphs show fasting blood glucose levels after 8 weeks of treatment with EMPA in (A) Kimba and (B) Akimba mice and treatment with CANA in (C) Kimba and (D) Akimba mice. ***p 0.001; ****p 0.0001; data represented as mean ± SEM of 3–6 mice per group. SGLT2, Sodium glucose cotransporter 2; EMPA, Empagliflozin; CANA, Canagliflozin.

3.3 Effects of EMPA and CANA on Body Weight

Firstly, we compared the starting (day 0) and final body weight (day 49; 7 weeks post-treatment) of Kimba and Akimba mice treated with vehicle. After 7 weeks non-diabetic Kimba mice showed a significant increase in body weight compared to the starting body weight (Fig. 3). However, diabetic Akimba mice showed lack of weight gain (Fig. 3), a hallmark of the failure to thrive phenotype in these animals.

Fig. 3.

Failure to thrive phenotype in diabetic Akimba mice. Graph depicts the raw body weight of non-diabetic Kimba and diabetic Akimba mice without treatments at the start of the experiment (day 0) and 7 weeks post (49 days). ****p 0.0001; data represented as mean ± SEM of 9–10 mice per group.

As shown in Fig. 4, EMPA treatment showed a reduction in percentage body weight gain in non-diabetic Kimba (Fig. 4A) mice throughout the treatment period from week 1 to 7. Interestingly, the percentage weight reduction was significant from week 2–7. In contrast, throughout the experiment period, the weight gain was increased in diabetic Akimba mice after treatment with EMPA (Fig. 4B). The CANA treatment also resulted in a reduction in percentage body weight gain in Kimba mice compared to vehicle treated mice (Fig. 4C). The percentage weight reduction was significant only at week 7. Similar to EMPA, treatment with CANA progressively increased body weight in diabetic Akimba mice (Fig. 4D).

Fig. 4.

Effect of SGLT2 inhibition on body weight in Kimba and Akimba mice treated with EMPA or CANA. Graphs show the normalized body weight percentage during 7 weeks of treatment with EMPA in (A) Kimba and (B) Akimba mice and treatment with CANA in (C) Kimba and (D) Akimba mice. *p 0.05; data represented as mean ± SEM of 4–6 mice per group. SGLT2, Sodium glucose cotransporter 2; EMPA, Empagliflozin; CANA, Canagliflozin.

3.4 Effects of EMPA and CANA on Water Intake

At the age of 10 weeks, non-diabetic Kimba mice had a water intake of less than 10 mL per day, which was considerably lower than that of the diabetic Akimba mice. In addition, diabetic Akimba mice showed a significant increase in water intake from day 3 to day 31 of the experiment (Fig. 5) suggestive of excessive thirst or polydipsia associated with the progression of diabetes.

Fig. 5.

Increased water intake in diabetic Akimba mice. Graph shows water intake in non-diabetic Kimba and diabetic Akimba mice after 3 days of treatment (day 3) and 4 weeks (31 days) post treatment. *p 0.01; ****p 0.0001; data represented as mean ± SEM of 9–10 mice per group.

In Kimba mice treated with EMPA or CANA (Fig. 6A,C), water intake from day 0 to day 31 post-treatment was slightly increased when compared to vehicle treated mice. There was no significant effect of EMPA on water intake in Akimba mice (Fig. 6B). Interestingly, CANA promoted reductions in water intake in treated mice at days 24- and 31-days post-treatment (Fig. 6D).

Fig. 6.

Effect of SGLT2 inhibition on water volume consumed by Kimba and Akimba mice treated with EMPA or CANA. Graphs depict the volume of water consumed at day 3, 10, 17, 24 and 31 by (A) Kimba and (B) Akimba mice treated with EMPA and (C) Kimba and (D) Akimba mice treated with CANA. Data represented as mean ± SEM of 4–8 mice per group. EMPA, Empagliflozin; CANA, Canagliflozin.

3.5 Effects of EMPA and CANA on the Retinal Vasculature

The objective of our study was to investigate whether inhibition of SGLT2 can delay the advancement of retinopathy and DR. To achieve this, the severity of retinal vascular lesions was evaluated after 8 weeks of treatment in Kimba and Akimba mice administered with either vehicle, EMPA or CANA. The number of vascular lesions in Kimba mice treated with EMPA was noticeably reduced when compared to vehicle treated mice (Fig. 7A–C). Interestingly, when compared to vehicle treatment, CANA significantly reduced retinal vascular lesions in Kimba mice (Fig. 7D–F). Similar to Kimba mice treated with EMPA, Akimba mice also showed a noteworthy reduction in retinal vascular lesions (Fig. 8A–C). However, there was no appreciable difference between the number of lesions in Akimba mice receiving vehicle or CANA (Fig. 8D–F).

Fig. 7.

The effect of SGLT2 inhibition on retinal microvascular lesions in non-diabetic Kimba mice. Representative areas of isolectin-B4 and Cy-3 stained vasculature from Kimba mice treated with (A and D) vehicle or (B) EMPA or (E) CANA and (C and F) quantification of retinal vascular lesions. *p 0.05; data represented as mean ± SEM of 5–6 mice per group. White arrows represent retinal microvascular lesions. Scale bar 100 μm; EMPA, Empagliflozin; CANA, Canagliflozin.

Fig. 8.

The effect of SGLT2 inhibition on retinal microvascular lesions in diabetic Akimba mice. Representative areas of isolectin-B4 and Cy-3 stained vasculature from Akimba mice treated with (A and D) vehicle or (B) EMPA or (E) CANA and (C and F) quantification of retinal vascular lesions. Data represented as mean ± SEM of 5–6 mice per group. White arrows represent retinal microvascular lesions. Scale bar 100 μm; EMPA, Empagliflozin; CANA, Canagliflozin.

An in-depth analysis of the retinal vasculature was carried out with the semi-quantitative grading of the retinal vascular features in EMPA or CANA treated Kimba and Akimba mice. These results are summarised in Tables 1,2 (Kimba and Akimba, respectively). When compared to vehicle treated mice, EMPA treated Kimba mice showed a marked reduction in capillary non-profusion, loss of vessel integrity and vascular leakage (Table 1 Panel A; symbol $) as well as vessel tortuosity (Table 1; Panel A; symbols &, $ and ^, respectively). The vascular leakage in EMPA treated Kimba mice was often confined to the areas with large IRMA. In CANA treated Kimba mice, capillary non-profusion, and vessel tortuosity (Table 1 Panel B; symbols & and ^, respectively) was not improved with SGLT2 inhibitor treatment. However, vascular leakage was markedly reduced in CANA treated Kimba mice when compared to vehicles (Table 1 Panel B; symbol $). In vehicle treated Kimba mice, although the number of microaneurysms/IRMA/vessel tufts appeared reduced these structures were often large and convoluted (Table 1 Panel A; symbol #) spanning into the mid and deep capillary beds of the retina. In EMPA treated mice, this phenomenon was noted only in 25% of retinas, whereas 75% of the retinas appeared to have minimal microaneurysms/IRMA/vessel tufts. The number of infiltrating macrophages and monocytes were also decreased with EMPA (Table 1 Panel A; symbol @) and CANA (Table 1 Panel B; symbol @) treatment in Kimba mice.

Table 1.Semiquantitative grading of the retinal vascular lesions in Kimba mice treated with vehicle or EMPA and vehicle or CANA.
Characteristic Panel A Panel B
Kimba + Vehicle Kimba + EMPA Kimba + Vehicle Kimba + CANA
1 2 3 4 5 1 2 3 4 5 1 2 3 4 5 1 2 3 4 5 6
Capillary non-profusion & ++++ + ++++ ++++ +++ ++++ + + ++ ++ ++ +++ ++++ ++ ++++ +++ +++ +++ ++ ++ ++
Loss of vessel integrity and leakage $ +++ - +aa +aa +a +a - - - - - - +a - +++ - - - - - -
Vessel tortuosity ^ +++ ++ ++ ++ ++ ++ -/+ -/+ -/+ + ++ ++ +++ ++ +++ ++ +++ ++ + ++ ++
Microaneurysms/IRMA/vessel tufts # +*/** + +* +*/** ++** +* -/+*** + + + ++ + +* ++* +* ++ ++** + + + +
Monocytes/macrophages @ ++++ - ++++ ++++ ++ ++++ - - - - ++ ++ +++ ++ +++ + ++ + - + +
&: -, Absent; +, in 1 quadrant; ++, in 2 quadrants, +++, in 3 quadrants, ++++, in 4 quadrants.
$: -, Absent; +, mild; ++, moderate; +++, severe.
^: -, Absent; + mild; ++ moderate; +++, severe.
#: -, Absent; +, less than 300 microaneurysms/IRMA/vessel tufts; ++, more than 300 microaneurysms/IRMA/ vessel tufts.
@: -, Absent; +, in 1 quadrant; ++, in 2 quadrants, +++, in 3 quadrants, ++++, in 4 quadrants.
*: Expansive retinopathy/capillary dropout resulted in the retina showing a lower incident of vascular lesions such as microaneurysms/IRMA/vessel tufts.
**: Large convoluted IRMA.
***: Showed less than 50 microaneurysms/IRMA/vessel tufts and the overall retinal vasculature was clearly organised.
a: Leakage confined to areas of IRMA.
aa: Minimal vascular capillaries remaining, these capillaries could be occluded leading to less vascular leakage.
EMPA, Empagliflozin; CANA, Canagliflozin; IRMA; Intraretinal microvascular abnormalities.
Table 2.Semiquantitative grading of the retinal vascular lesions in Akimba mice treated with vehicle or EMPA and vehicle or CANA.
Characteristic Panel A Panel B
Akimba + Vehicle Akimba + EMPA Akimba + Vehicle Akimba + CANA
1 2 3 4 1 2 3 4 1 2 3 4 5 1 2 3 4 5 6
Capillary non-profusion & ++++ +++ ++ ++++ + ++++ + + ++ ++ ++ +++ + ++ ++++ ++++ ++++ ++ ++++
Loss of vessel integrity and leakage $ ++ - - +++ - +++ - - + - - - - - -aa +++ +a - -aa
Vessel tortuosity ^ +++ +++ ++ +++ - / + +++ -/+ -/+ +++ ++ +++ ++ + ++ +++ +++ +++ ++ +++
Microaneurysms/IRMA/vessel tufts # +* ++ ++ +* -/+*** +* -/+*** -/+*** ++ + ++ ++** + + +* +* +* + +*
Monocytes/macrophages @ ++++ - - ++++ - ++ - - +++ - - ++ - - ++ ++ ++ - ++
&: -, Absent; +, in 1 quadrant; ++, in 2 quadrants, +++, in 3 quadrants, ++++, in 4 quadrants.
$: -, Absent; +, mild; ++, moderate; +++, severe.
^: -, Absent; + mild; ++ moderate; +++, severe.
#: -, Absent; +, less than 300 microaneurysms/IRMA/vessel tufts; ++, more than 300 microaneurysms/IRMA/ vessel tufts.
@: -, Absent; +, in 1 quadrant; ++, in 2 quadrants, +++, in 3 quadrants, ++++, in 4 quadrants.
*: Expansive retinopathy/capillary dropout resulted in the retina showing a lower incident of vascular lesions such as microaneurysms/IRMA/vessel tufts.
**: Large convoluted IRMA.
***: Showed less than 50 microaneurysms/IRMA/vessel tufts and the overall retinal vasculature was clearly organised.
a: Leakage confined to areas of IRMA.
aa: Minimal vascular capillaries remaining, these capillaries could be occluded leading to less vascular leakage.
EMPA, Empagliflozin; CANA, Canagliflozin; IRMA; Intraretinal microvascular abnormalities.

Overall, Akimba mice showed more pronounced vascular disorganization, microaneurysms, IRMA and neovascular tufts when compared to Kimba mice as previously published [32, 35, 37, 39]. With the treatment of EMPA, 75% of the DR Akimba mice showed a marked improvement in all retinal vascular parameters when compared to vehicles (Table 2 Panel A). However, 25% EMPA treated Akimba mice showed extensive damage to the retina which was similar to vehicle treated mice (Supplementary Fig. 1). The treatment with CANA did not show an overall improvement in the retinal vascular features when compared to vehicle treated Akimba mice (Table 2 Panel B). While the number of microaneurysms/IRMA/vessel tufts appeared to be reduced, there were expansive regions without retinal capillaries in Akimba mice treated with CANA (Table 2 Panel B; symbol # and &, respectively).

4. Discussion

SGLT2 inhibitors are now an established therapy for the treatment of diabetes due to their anti-diabetic properties and their potential benefits for cardiovascular and renal complications [40, 41, 42, 43, 44]. Many pre-clinical studies on both Type 1 or Type 2 diabetic animals have shown that glycemic control with SGLT2 inhibitors decreased the progression of major microvascular complications associated with diabetes [21, 45, 46, 47]. In this novel study, we examined the therapeutic effect of two SGLT2 inhibitors, EMPA and CANA, on retinopathy and DR, utilizing the well-characterised Kimba and Akimba mice, respectively. We chose Kimba and Akimba mice for the current study, as these models are regarded as two of the most suitable models of retinopathy and DR [15, 25, 32, 35, 37]. The key findings of this study are (i) both SGLT2 inhibitors significantly lowered fasting blood glucose levels and improved weight gain in diabetic Akimba mice; (ii) EMPA exerts protective effects for both retinopathy and DR as shown by the reduction in retinal vascular abnormalities and (iii) CANA showed protective effects against retinopathy in Kimba mice only.

Similar to human trials, EMPA and CANA reduced fasting blood glucose levels in our type 1 diabetic Akimba model, in agreement with other rodent studies [45, 48, 49, 50, 51]. Interestingly, CANA showed a more profound reduction in fasting blood glucose levels compared to EMPA (Vehicle 32.46 ± 0.5 mmol/L versus CANA 17.43 ± 1.3 mmol/L; p = 0.0000039 and Vehicle 32.67 ± 0.6 mmol/L vs EMPA 21.9 ± 1 mmol/L; p = 0.00044). Gliflozins present various potencies of inhibition of SGLT1 and SGLT2 [52]. It is reported that SGLT1 inhibition by CANA contributes to 10.6% of daily urinary glucose excretion, whereas SGLT1 inhibition by EMPA is 2.41% [53]. This shows that in comparison to EMPA, CANA demonstrates a greater inhibitory effect on SGLT1 [54].

Glucose variability or fluctuation of glucose levels are a contributing factor to diabetes-related macrovascular complications [55]. It is shown that high glucose variability can lead to endothelial dysfunction [56, 57], which in turn may contribute to the development and progression of DR. SGLT2 inhibitors have been shown to reduce glucose variability in diabetic patients [58] thereby reducing endothelial dysfunction. Therefore, it is plausible to hypothesise that SGLT2 inhibitors may in fact improve DR by efficiently managing glucose variability and thereby reducing endothelial dysfunction. Therefore, future studies should investigate these aspects in relation to DR and SGLT2 inhibitors.

Typically, impaired or stalled growth is a characteristic feature of uncontrolled hyperglycaemia [59]. Type 1 diabetic Akimba mice typically show poor weight gain which is a hallmark of the failure to thrive phenotype in these animals. We have previously shown that EMPA prevented the failure to thrive phenotype in these animals when treatment started at 4 weeks of age [25]. Similarly, in our current study, the SGLT2 inhibitors EMPA and CANA both induced improved weight gain in Akimba mice. However, an opposite effect on body weight was observed in the non-diabetic Kimba mice. Similar findings have been reported in a study by Vallon et al. [45] in diabetic Akita mice and its non-diabetic counterparts.

Consistent with previous reports in SGLT2-deficient non-diabetic mice [60, 61], the increased loss of glucose calories into the urine (glucosuria) of SGLT2 inhibitor-treated (both EMPA and CANA) non-diabetic Kimba mice could be responsible for the increased fluid intake. Furthermore, Masuda et al. [62], reported that in non-diabetic rats with free access to food and water, SGLT2 inhibition increased glucosuria, urine volume and Na+ excretion, which was associated with increased fluid intake. Similarly, in non-diabetic Kimba mice, the mechanism of action of EMPA and CANA potentially caused hyperosmolar urine (due to the presence of Na+ and glucose) leading to polyuria (excessive urination), due to osmotic diuresis. This was likely responsible for polydipsia (excessive thirst resulting in increased fluid intake) noted in these animals. It can be suggested that the increase in fluid intake in response to SGLT2 inhibition may be compensating the primary diuretic, glucosuric, and natriuretic effect of EMPA and CANA to maintain fluid and electrolyte balance in Kimba mice [62].

A very limited number of studies have investigated the effect of SGLT2 inhibition on the retinal vasculature. A substantial decrease in the number of acellular capillaries were observed with the treatment of Dapagliflozin db/db mouse retinas [63]. In early stages of DR, microaneurysm, exudation, and retinal hypoperfusion was reduced in db/db mice with the treatment of EMPA [19]. Eid et al. [21] showed that EMPA reduced retinal apoptosis and thereby showed a trend toward a salutary effect on DR in type I diabetic animals. Similarly, we have recently demonstrated that the features of DR in young Akimba mice (4 weeks of age) were decreased with the treatment of EMPA [25]. When compared to younger Akimba mice, older mice used in this study showed a severe disease phenotype [25, 32, 39]. In this study, we have shown that in older Akimba mice, EMPA attenuated diabetes-associated increases in retinal microvascular abnormalities such as microaneurysms, IRMA, capillary dropouts and vessel tortuosity. Taken together, our investigations in the specific Type 1 Diabetes Akimba mouse model suggests that the SGLT2 inhibitor EMPA could be a promising therapeutic for the treatment of DR development and progression.

Loss of neural cell populations and thinning and changes in retinal layers are well-documented in both Kimba and Akimba mice [33, 34, 35, 37, 39]. We have recently shown that, in Akimba mice, the SGLT2 inhibitor Dapagliflozin preserved the well-structured neural layers, reduced Muller cell gliosis and minimised abnormal blood vessel transgression to the outer retinal neural layers (outer plexiform and nuclear layer) [15]. Similarly, Ipragliflozin has been shown to improve retinal neural layer modifications in spontaneously diabetic Torii fatty rats [64] and reduced modifications to the nerve fiber layer and ganglion cell layer and prevented retinal neural apoptosis in db/db mice [19]. Therefore, future studies should investigate the effect of both EMPA and CANA on the retinal neural structure of Kimba and Akimba mice to gain a better understanding of the role of SGLT2 inhibition in the neurodegeneration of the retina.

The frequency and severity of DR among patients with diabetes mellitus is heterogeneous [65]. Although not entirely explained, known risk factors such as duration of diabetes and glycaemic control, explains some of the observed heterogeneity in DR [66, 67]. For instance, some diabetic patients develop DR and progress through the disease, despite a short duration of diabetes and/or excellent glycaemic control. Interestingly, other patients do not develop DR with long durations of diabetes and/or long-term poorly managed hyperglycaemia [65]. As summarised in Tables 1,2, this heterogenous disease phenotype was evident in the Kimba mice displaying retinopathy and Akimba mice demonstrating DR.

Differential response to the same treatment by different patients with different characteristics of DR has been shown [68]. Similarly, although EMPA decreased the overall progression of retinal vascular lesions in Akimba mice, a small percentage of animals did not show an improvement in retinal vascular abnormalities due to the deferential response to EMPA treatment by Akimba mice with DR. Furthermore, it is evident that upon the complete disruption of the retinal vasculature (Supplementary Fig. 1), treatment may not achieve improvements in the disease pathology [69]. There is cumulative evidence demonstrating the effectiveness of various treatment options earlier in the pathogenesis of DR [70, 71]. Therefore, choosing the appropriate treatment window with an earlier initiation of treatment may potentially result in improved outcomes in DR [19, 25, 71].

Monitoring vascular and non-vascular alterations of retinopathy and DR using in vivo ophthalmic modalities are often used in translational research [72]. However, one limitation of our study is that we were unable to perform longitudinal in vivo imaging and functional investigations to evaluate the efficacy of SGLT2 inhibitor treatment on the retinal vasculature, neural structure and function. To date, a very limited number of studies have utilised in vivo techniques to study the effects of SGLT2 inhibitors on the retina. For instance, Takakura and others showed that the treatment of Iplegliflozin in spontaneously diabetic Torii obese rats improved the oscillating potential when retinal electroretinogram was conducted [64]. It is also shown that, in db/db mice, Dapagliflozin treatment improved the Electroretinogram (ERG) b-wave amplitude [63] and Empagliflozin alleviated retinal edema around the disc observed by optical coherence tomography (OCT) [19]. Therefore, future studies should utilize in vivo techniques such as OCT, fundus fluorescein angiography (FFA), electrophysiological testing (ERG) and fundus photography (FP) to study the structure (vascular and neural) and function of the retina [72]. These studies will reveal clinically relevant findings and provide important insights into the effectiveness of SGLT2 inhibitor treatment.

Prolonged hyperglycaemia is an established, independent, and prominent risk factor for the development and progression of DR and good glycaemic control is strongly related to the progression and improvement of DR [66, 73]. In part, the beneficial retinal vascular outcomes in the treated diabetic Akimba mice are likely due to glycaemic control conferred by SGLT2 inhibitors. However, the fact that (i) Akimba-CANA mice did not demonstrate an improvement in the retinal vasculature and (ii) the improvement in the retinal vascular phenotype in the non-diabetic Kimba mice (with EMPA or CANA) indicates that there are perhaps other mechanisms influenced by the treatment of this drug class. Hence, inflammation, oxidative stress, blood flow autoregulatory mechanisms, increased sorbitol in retinal cells as a consequence of osmotic effects, advanced glycosylation end product formation and the role of growth-factor/alternative angiogenic factor production should be investigated in future studies to determine the mechanisms of action of SGLT2 inhibitors [15, 16, 18, 20, 74, 75, 76, 77].

The relationship between dyslipidemia and DR is controversial and not yet fully understood [78, 79, 80]. Some studies have shown diabetic patients display high levels of triglycerides and low-density lipoprotein (LDL) cholesterol, along with low high-density lipoprotein (HDL) cholesterol and these patients may have an increased risk of development and progression of DR [81, 82, 83, 84]. However, the NO BLIND study showed that high HDL values are associated to DR [85] and as revealed by the United Kingdom Prospective Diabetes Study (UKPDS), higher levels of HDL cholesterol levels correlated with more advanced retinopathy [86]. A recent study reported that the parental strain of the Akimba, the Akita mouse, showed significantly increased LDL cholesterol while HDL cholesterol levels were significantly higher [87]. Therefore, it is likely that the Akimba mouse model with DR may possess a similar lipid profile. However, future studies should firstly investigate the lipid profiles of Akimba mice and secondly, conduct dose dependant HDL therapy studies [88] to determine the effect of HDL in the development and progression of DR in the Akimba model. As recently reviewed by Yaribeygi et al. [89], evidence suggest that SGLT2 inhibitors improved dyslipidaemia by decreasing LDL-levels, triglycerides and increasing HDL-levels, thus providing cardiovascular benefits in diabetic patients. Given improving dyslipidaemia may improve retinal endothelial function, retinal blood flow, inflammation, retinal vascular leakage, and thus delay the development and progression of DR [90], further research is needed to determine the long-term effects of SGLT2 inhibitors on HDL levels and DR.

Tahara and colleagues used diabetic and non-diabetic rodents to demonstrate that each SGLT2 inhibitor has distinct pharmacokinetics, pharmacodynamics, and pharmacologic effects [91, 92, 93]. Although all SGLT2 inhibitors block the function of the SGLT2 protein in the kidneys, there are differences in their onset of action, half-life, renal threshold, selectivity, absorption, distribution, and metabolism. Additionally, their relative affinity for the SGLT2 protein versus the SGLT1 protein is significant in understanding the distinct effects of these medications [94]. While both SGLT2 inhibitors in this study effectively increased urinary glucose excretion and decreased hyperglycemia, the actual differences reflected in the pharmacokinetics, pharmacodynamics, and pharmacologic profiles of each of these SGLT2 inhibitor might be responsible for the distinct SGLT2 inhibitor mediated variations in metabolic parameters, retinopathy and diabetic retinopathy observed in our specific mouse models.

5. Conclusions

Our findings suggest beneficial effects of SGLT2 inhibition on DR, which now warrants clinical investigations to determine the overall effect of SGLT2 inhibition on the full spectrum of DR. Such clinical studies will be able to address the differences in outcomes between individual SGLT2 inhibitors and also determine if these outcomes are due to differences in SGLT2 selectivity.

Availability of Data and Materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Author Contributions

Conceptualization, LYH, VBM, MPS and EPR; formal analysis, LYH and VBM; funding acquisition, LYH, VBM and MPS; investigation, LYH, JRM, VBM and MPS; writing-original draft, LYH, JRM and VBM; writing-review and editing, LYH, JRM, MPS, EPR and VBM. All authors have read and agreed to the published version of the manuscript.

Ethics Approval and Consent to Participate

Animal ethics was approved by both the Royal Perth Hospital Animal Ethics Committee (R537/17-20; approved: 15/08/17) and Harry Perkins Institute for Medical Research Animal Ethics Committee (AE141/2019; approved: 12/02/19).

Acknowledgment

We thank Dr. Aaron Magno (University of Western Australia, Western Australia) for technical assistance. Authors thank Animal Resources Centre (Murdoch, Western Australia) and the Bioresources, Harry Perkins Institute of Medical Research (Nedlands, Western Australia) for providing animal care services.

Funding

This study was funded by the Royal Perth Hospital-Research Foundation WA (VM2018), Diabetes Australia Research Program (Y20G-MATV), Diabetes Research WA (DRWA-LHerat-2020) and Raine Medical Research Foundation, WA (Raine-LHerat-2020).

Conflict of Interest

MS has received research support from Boehringer Ingelheim. The other authors declare no conflicts of interests.

References
[1]
Grauslund J, Green A, Sjølie AK. Prevalence and 25 year incidence of proliferative retinopathy among Danish type 1 diabetic patients. Diabetologia. 2009; 52: 1829–1835.
[2]
Romero-Aroca P, Baget-Bernaldiz M, Fernandez-Ballart J, Plana-Gil N, Soler-Lluis N, Mendez-Marin I, et al. Ten-year incidence of diabetic retinopathy and macular edema. Risk factors in a sample of people with type 1 diabetes. Diabetes Research and Clinical Practice. 2011; 94: 126–132.
[3]
Jones CD, Greenwood RH, Misra A, Bachmann MO. Incidence and progression of diabetic retinopathy during 17 years of a population-based screening program in England. Diabetes Care. 2012; 35: 592–596.
[4]
Thomas RL, Dunstan F, Luzio SD, Roy Chowdury S, Hale SL, North RV, et al. Incidence of diabetic retinopathy in people with type 2 diabetes mellitus attending the Diabetic Retinopathy Screening Service for Wales: retrospective analysis. BMJ (Clinical Research Ed.). 2012; 344: e874.
[5]
Atlas D. IDF Diabetes Atlas. 10th edn. International Diabetes Federation: Brussels, Belgium. 2021.
[6]
Williams R, Airey M, Baxter H, Forrester J, Kennedy-Martin T, Girach A. Epidemiology of diabetic retinopathy and macular oedema: a systematic review. Eye (London, England). 2004; 18: 963–983.
[7]
Shukla UV, Tripathy K. Diabetic Retinopathy. StatPearls Publishing: Treasure Island (FL). 2023.
[8]
Cheung N, Mitchell P, Wong TY. Diabetic retinopathy. Lancet (London, England). 2010; 376: 124–136.
[9]
. The relationship of glycemic exposure (HbA1c) to the risk of development and progression of retinopathy in the diabetes control and complications trial. Diabetes. 1995; 44: 968–983.
[10]
Wang W, Lo ACY. Diabetic Retinopathy: Pathophysiology and Treatments. International Journal of Molecular Sciences. 2018; 19: 1816.
[11]
Chao EC, Henry RR. SGLT2 inhibition–a novel strategy for diabetes treatment. Nature Reviews. Drug Discovery. 2010; 9: 551–559.
[12]
Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. The New England Journal of Medicine. 2015; 373: 2117–2128.
[13]
Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, et al. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. The New England Journal of Medicine. 2016; 375: 323–334.
[14]
Vallon V, Verma S. Effects of SGLT2 Inhibitors on Kidney and Cardiovascular Function. Annual Review of Physiology. 2021; 83: 503–528.
[15]
Herat LY, Matthews JR, Ong WE, Rakoczy EP, Schlaich MP, Matthews VB. Determining the Role of SGLT2 Inhibition with Dapagliflozin in the Development of Diabetic Retinopathy. Frontiers in Bioscience (Landmark Edition). 2022; 27: 321.
[16]
Herat LY, Matthews VB, Rakoczy PE, Carnagarin R, Schlaich M. Focusing on Sodium Glucose Cotransporter-2 and the Sympathetic Nervous System: Potential Impact in Diabetic Retinopathy. International Journal of Endocrinology. 2018; 2018: 9254126.
[17]
Wakisaka M, Nagao T. Sodium glucose cotransporter 2 in mesangial cells and retinal pericytes and its implications for diabetic nephropathy and retinopathy. Glycobiology. 2017; 27: 691–695.
[18]
Herat LY, Ward NC, Magno AL, Rakoczy EP, Kiuchi MG, Schlaich MP, et al. Sodium glucose co-transporter 2 inhibition reduces succinate levels in diabetic mice. World Journal of Gastroenterology. 2020; 26: 3225–3235.
[19]
Gong Q, Zhang R, Wei F, Fang J, Zhang J, Sun J, et al. SGLT2 inhibitor-empagliflozin treatment ameliorates diabetic retinopathy manifestations and exerts protective effects associated with augmenting branched chain amino acids catabolism and transportation in db/db mice. Biomedicine & Pharmacotherapy. 2022; 152: 113222.
[20]
Hu Y, Xu Q, Li H, Meng Z, Hao M, Ma X, et al. Dapagliflozin Reduces Apoptosis of Diabetic Retina and Human Retinal Microvascular Endothelial Cells Through ERK1/2/cPLA2/AA/ROS Pathway Independent of Hypoglycemic. Frontiers in Pharmacology. 2022; 13: 827896.
[21]
Eid SA, O’Brien PD, Hinder LM, Hayes JM, Mendelson FE, Zhang H, et al. Differential Effects of Empagliflozin on Microvascular Complications in Murine Models of Type 1 and Type 2 Diabetes. Biology. 2020; 9: 347.
[22]
Sabaner MC, Duman R, Dogan M, Akdogan M, Vurmaz A, Bozkurt E, et al. Do SGLT2 inhibitors prevent preclinical diabetic retinopathy? A Prospective Pilot Optical Coherence Tomography Angiography Study. Journal Francais D’ophtalmologie. 2021; 44: 1159–1167.
[23]
El Mouhayyar C, Riachy R, Khalil AB, Eid A, Azar S. SGLT2 Inhibitors, GLP-1 Agonists, and DPP-4 Inhibitors in Diabetes and Microvascular Complications: A Review. International Journal of Endocrinology. 2020; 2020: 1762164.
[24]
Matthews JR, Schlaich MP, Rakoczy EP, Matthews VB, Herat LY. The Effect of SGLT2 Inhibition on Diabetic Kidney Disease in a Model of Diabetic Retinopathy. Biomedicines. 2022; 10: 522.
[25]
Matthews J, Herat L, Rooney J, Rakoczy E, Schlaich M, Matthews VB. Determining the role of SGLT2 inhibition with Empagliflozin in the development of diabetic retinopathy. Bioscience Reports. 2022; 42: BSR20212209.
[26]
Wakisaka M, Yoshinari M, Nakamura S, Asano T, Sonoki K, Shi AH, et al. Suppression of sodium-dependent glucose uptake by captopril improves high-glucose-induced morphological and functional changes of cultured bovine retinal pericytes. Microvascular Research. 1999; 58: 215–223.
[27]
Leley SP, Luo Q, Bhatwadekar AD. 588-P: diabetes leads to sodium-glucose cotransporter 2 (SGLT2) increase in the retina. Diabetes. 2019; 68.
[28]
Schmidt DW, Argyropoulos C, Singh N. Are the Protective Effects of SGLT2 Inhibitors a “Class-Effect” or Are There Differences between Agents? Kidney360. 2021; 2: 881–885.
[29]
Memon RA, Akbariromani H, Vohra RR, Kundi H, Saleem RF, Ghaffari MA, et al. Comparison of Cardiovascular Outcomes Between Dapagliflozin and Empagliflozin in Patients With Type 2 Diabetes: A Meta-Analysis. Cureus. 2022; 14: e27277.
[30]
Wei XB, Wei W, Ding LL, Liu SY. Comparison of the effects of 10 GLP-1 RA and SGLT2 inhibitor interventions on cardiovascular, mortality, and kidney outcomes in type 2 diabetes: A network meta-analysis of large randomized trials. Primary Care Diabetes. 2021; 15: 208–211.
[31]
Binz N, Rakoczy EP, Ali Rahman IS, Vagaja NN, Lai CM. Biomarkers for Diabetic Retinopathy - Could Endothelin 2 Be Part of the Answer? PLoS ONE. 2016; 11: e0160442.
[32]
Rakoczy EP, Ali Rahman IS, Binz N, Li CR, Vagaja NN, de Pinho M, et al. Characterization of a mouse model of hyperglycemia and retinal neovascularization. The American Journal of Pathology. 2010; 177: 2659–2670.
[33]
Wisniewska-Kruk J, Klaassen I, Vogels IMC, Magno AL, Lai CM, Van Noorden CJF, et al. Molecular analysis of blood-retinal barrier loss in the Akimba mouse, a model of advanced diabetic retinopathy. Experimental Eye Research. 2014; 122: 123–131.
[34]
Binz N, Ali Rahman IS, Chinnery HR, McKeone R, Simpson KM, Speed TP, et al. Effect of vascular endothelial growth factor upregulation on retinal gene expression in the Kimba mouse. Clinical & Experimental Ophthalmology. 2013; 41: 251–262.
[35]
Ali Rahman IS, Li CR, Lai CM, Rakoczy EP. In vivo monitoring of VEGF-induced retinal damage in the Kimba mouse model of retinal neovascularization. Current Eye Research. 2011; 36: 654–662.
[36]
Tee LBG, Penrose MA, O’Shea JE, Lai CM, Rakoczy EP, Dunlop SA. VEGF-induced choroidal damage in a murine model of retinal neovascularisation. The British Journal of Ophthalmology. 2008; 92: 832–838.
[37]
Lai CM, Dunlop SA, May LA, Gorbatov M, Brankov M, Shen WY, et al. Generation of transgenic mice with mild and severe retinal neovascularisation. The British Journal of Ophthalmology. 2005; 89: 911–916.
[38]
Vagaja NN, Chinnery HR, Binz N, Kezic JM, Rakoczy EP, McMenamin PG. Changes in murine hyalocytes are valuable early indicators of ocular disease. Investigative Ophthalmology & Visual Science. 2012; 53: 1445–1451.
[39]
Rahman ISA. Characterisation of the Akimba Mouse: A Model of Retinal Neovascularisation on a Hyperglycaemic Background and the in Vivo Study of Endothelin-2 in the Development of Neovascularisation. University of Western Australia. 2011.
[40]
Herat LY, Matthews VB, Magno AL, Kiuchi MG, Carnagarin R, Schlaich MP. An evaluation of empagliflozin and it’s applicability to hypertension as a therapeutic option. Expert Opinion on Pharmacotherapy. 2020; 21: 1157–1166.
[41]
Cowie MR, Fisher M. SGLT2 inhibitors: mechanisms of cardiovascular benefit beyond glycaemic control. Nature Reviews. Cardiology. 2020; 17: 761–772.
[42]
Verma S, McMurray JJV. SGLT2 inhibitors and mechanisms of cardiovascular benefit: a state-of-the-art review. Diabetologia. 2018; 61: 2108–2117.
[43]
Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet (London, England). 2019; 393: 31–39.
[44]
Neuen BL, Young T, Heerspink HJL, Neal B, Perkovic V, Billot L, et al. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: a systematic review and meta-analysis. The Lancet. Diabetes & Endocrinology. 2019; 7: 845–854.
[45]
Vallon V, Gerasimova M, Rose MA, Masuda T, Satriano J, Mayoux E, et al. SGLT2 inhibitor empagliflozin reduces renal growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice. American Journal of Physiology. Renal Physiology. 2014; 306: F194–204.
[46]
Gallo LA, Ward MS, Fotheringham AK, Zhuang A, Borg DJ, Flemming NB, et al. Once daily administration of the SGLT2 inhibitor, empagliflozin, attenuates markers of renal fibrosis without improving albuminuria in diabetic db/db mice. Scientific Reports. 2016; 6: 26428.
[47]
Lin B, Koibuchi N, Hasegawa Y, Sueta D, Toyama K, Uekawa K, et al. Glycemic control with empagliflozin, a novel selective SGLT2 inhibitor, ameliorates cardiovascular injury and cognitive dysfunction in obese and type 2 diabetic mice. Cardiovascular Diabetology. 2014; 13: 148.
[48]
Ashrafi Jigheh Z, Ghorbani Haghjo A, Argani H, Roshangar L, Rashtchizadeh N, Sanajou D, et al. Empagliflozin alleviates renal inflammation and oxidative stress in streptozotocin-induced diabetic rats partly by repressing HMGB1-TLR4 receptor axis. Iranian Journal of Basic Medical Sciences. 2019; 22: 384–390.
[49]
Al-Sharea A, Murphy AJ, Huggins LA, Hu Y, Goldberg IJ, Nagareddy PR. SGLT2 inhibition reduces atherosclerosis by enhancing lipoprotein clearance in Ldlr-/- type 1 diabetic mice. Atherosclerosis. 2018; 271: 166–176.
[50]
Miyata KN, Zhao S, Wu CH, Lo CS, Ghosh A, Chenier I, et al. Comparison of the effects of insulin and SGLT2 inhibitor on the Renal Renin-Angiotensin system in type 1 diabetes mice. Diabetes Research and Clinical Practice. 2020; 162: 108107.
[51]
Thrailkill KM, Nyman JS, Bunn RC, Uppuganti S, Thompson KL, Lumpkin CK, Jr, et al. The impact of SGLT2 inhibitors, compared with insulin, on diabetic bone disease in a mouse model of type 1 diabetes. Bone. 2017; 94: 141–151.
[52]
Faillie JL. Pharmacological aspects of the safety of gliflozins. Pharmacological Research. 2017; 118: 71–81.
[53]
Sokolov V, Yakovleva T, Chu L, Tang W, Greasley PJ, Johansson S, et al. Differentiating the Sodium-Glucose Cotransporter 1 Inhibition Capacity of Canagliflozin vs. Dapagliflozin and Empagliflozin Using Quantitative Systems Pharmacology Modeling. CPT: Pharmacometrics & Systems Pharmacology. 2020; 9: 222–229.
[54]
Takebayashi K, Inukai T. Effect of Sodium Glucose Cotransporter 2 Inhibitors With Low SGLT2/SGLT1 Selectivity on Circulating Glucagon-Like Peptide 1 Levels in Type 2 Diabetes Mellitus. Journal of Clinical Medicine Research. 2017; 9: 745–753.
[55]
Hsu CR, Chen YT, Sheu WHH. Glycemic variability and diabetes retinopathy: a missing link. Journal of Diabetes and its Complications. 2015; 29: 302–306.
[56]
Ceriello A, Esposito K, Piconi L, Ihnat M, Thorpe J, Testa R, et al. Glucose “peak” and glucose “spike”: Impact on endothelial function and oxidative stress. Diabetes Research and Clinical Practice. 2008; 82: 262–267.
[57]
Saisho Y. Glycemic variability and oxidative stress: a link between diabetes and cardiovascular disease? International Journal of Molecular Sciences. 2014; 15: 18381–18406.
[58]
Henry RR, Strange P, Zhou R, Pettus J, Shi L, Zhuplatov SB, et al. Effects of Dapagliflozin on 24-Hour Glycemic Control in Patients with Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Technology & Therapeutics. 2018; 20: 715–724.
[59]
Giannini C, Mohn A, Chiarelli F. Growth abnormalities in children with type 1 diabetes, juvenile chronic arthritis, and asthma. International Journal of Endocrinology. 2014; 2014: 265954.
[60]
Vallon V, Rose M, Gerasimova M, Satriano J, Platt KA, Koepsell H, et al. Knockout of Na-glucose transporter SGLT2 attenuates hyperglycemia and glomerular hyperfiltration but not kidney growth or injury in diabetes mellitus. American Journal of Physiology. Renal Physiology. 2013; 304: F156–67.
[61]
Vallon V, Platt KA, Cunard R, Schroth J, Whaley J, Thomson SC, et al. SGLT2 mediates glucose reabsorption in the early proximal tubule. Journal of the American Society of Nephrology: JASN. 2011; 22: 104–112.
[62]
Masuda T, Watanabe Y, Fukuda K, Watanabe M, Onishi A, Ohara K, et al. Unmasking a sustained negative effect of SGLT2 inhibition on body fluid volume in the rat. American Journal of Physiology. Renal Physiology. 2018; 315: F653–F664.
[63]
Luo Q, Leley SP, Bello E, Dhami H, Mathew D, Bhatwadekar AD. Dapagliflozin protects neural and vascular dysfunction of the retina in diabetes. BMJ Open Diabetes Research & Care. 2022; 10: e002801.
[64]
Takakura S, Toyoshi T, Hayashizaki Y, Takasu T. Effect of ipragliflozin, an SGLT2 inhibitor, on progression of diabetic microvascular complications in spontaneously diabetic Torii fatty rats. Life Sciences. 2016; 147: 125–131.
[65]
Cho H, Sobrin L. Genetics of diabetic retinopathy. Current Diabetes Reports. 2014; 14: 515.
[66]
Klein R, Klein BE, Moss SE, Cruickshanks KJ. The Wisconsin Epidemiologic Study of Diabetic Retinopathy: XVII. The 14-year incidence and progression of diabetic retinopathy and associated risk factors in type 1 diabetes. Ophthalmology. 1998; 105: 1801–1815.
[67]
Klein R, Klein BE, Moss SE, Cruickshanks KJ. Relationship of hyperglycemia to the long-term incidence and progression of diabetic retinopathy. Archives of Internal Medicine. 1994; 154: 2169–2178.
[68]
Halim S, Nugawela M, Chakravarthy U, Peto T, Madhusudhan S, Lenfestey P, et al. Topographical Response of Retinal Neovascularization to Aflibercept or Panretinal Photocoagulation in Proliferative Diabetic Retinopathy: Post Hoc Analysis of the CLARITY Randomized Clinical Trial. JAMA Ophthalmology. 2021; 139: 501–507.
[69]
Stitt AW, Curtis TM, Chen M, Medina RJ, McKay GJ, Jenkins A, et al. The progress in understanding and treatment of diabetic retinopathy. Progress in Retinal and Eye Research. 2016; 51: 156–186.
[70]
Effect of intensive diabetes treatment on the development and progression of long-term complications in adolescents with insulin-dependent diabetes mellitus: Diabetes Control and Complications Trial. Diabetes Control and Complications Trial Research Group. The Journal of Pediatrics. 1994; 125: 177–188.
[71]
Ferris FL, 3rd, Davis MD, Aiello LM. Treatment of diabetic retinopathy. The New England Journal of Medicine. 1999; 341: 667–678.
[72]
Sher I, Moverman D, Ketter-Katz H, Moisseiev E, Rotenstreich Y. In vivo retinal imaging in translational regenerative research. Annals of Translational Medicine. 2020; 8: 1096.
[73]
Klein R, Knudtson MD, Lee KE, Gangnon R, Klein BEK. The Wisconsin Epidemiologic Study of Diabetic Retinopathy: XXII the twenty-five-year progression of retinopathy in persons with type 1 diabetes. Ophthalmology. 2008; 115: 1859–1868.
[74]
Lahoti S, Nashawi M, Sheikh O, Massop D, Mir M, Chilton R. Sodium-glucose co-transporter 2 inhibitors and diabetic retinopathy: insights into preservation of sight and looking beyond. Cardiovascular Endocrinology & Metabolism. 2020; 10: 3–13.
[75]
Herat LY, Magno AL, Rudnicka C, Hricova J, Carnagarin R, Ward NC, et al. SGLT2 Inhibitor-Induced Sympathoinhibition: A Novel Mechanism for Cardiorenal Protection. JACC. Basic to Translational Science. 2020; 5: 169–179.
[76]
Sha W, Wen S, Chen L, Xu B, Lei T, Zhou L. The Role of SGLT2 Inhibitor on the Treatment of Diabetic Retinopathy. Journal of Diabetes Research. 2020; 2020: 8867875.
[77]
Salvatore T, Galiero R, Caturano A, Rinaldi L, Di Martino A, Albanese G, et al. An Overview of the Cardiorenal Protective Mechanisms of SGLT2 Inhibitors. International Journal of Molecular Sciences. 2022; 23: 3651.
[78]
Zhou Y, Wang C, Shi K, Yin X. Relationship between dyslipidemia and diabetic retinopathy: A systematic review and meta-analysis. Medicine. 2018; 97: e12283.
[79]
Bryl A, Mrugacz M, Falkowski M, Zorena K. The Effect of Hyperlipidemia on the Course of Diabetic Retinopathy-Literature Review. Journal of Clinical Medicine. 2022; 11: 2761.
[80]
Chou Y, Ma J, Su X, Zhong Y. Emerging insights into the relationship between hyperlipidemia and the risk of diabetic retinopathy. Lipids in Health and Disease. 2020; 19: 241.
[81]
Ezhilvendhan K, Sathiyamoorthy A, Prakash BJ, Bhava BS, Shenoy A. Association of Dyslipidemia with Diabetic Retinopathy in Type 2 Diabetes Mellitus Patients: A Hospital-Based Study. Journal of Pharmacy & Bioallied Sciences. 2021; 13: S1062–S1067.
[82]
Rathsman B, Haas J, Persson M, Ludvigsson J, Svensson AM, Lind M, et al. LDL cholesterol level as a risk factor for retinopathy and nephropathy in children and adults with type 1 diabetes mellitus: A nationwide cohort study. Journal of Internal Medicine. 2021; 289: 873–886.
[83]
Lyons TJ, Jenkins AJ, Zheng D, Lackland DT, McGee D, Garvey WT, et al. Diabetic retinopathy and serum lipoprotein subclasses in the DCCT/EDIC cohort. Investigative Ophthalmology & Visual Science. 2004; 45: 910–918.
[84]
Tomić M, Vrabec R, Bulum T, Ljubić S. HDL cholesterol is a protective predictor in the development and progression of retinopathy in type 1 diabetes: A 15-year follow-up study. Diabetes Research and Clinical Practice. 2022; 186: 109814.
[85]
Sasso FC, Pafundi PC, Gelso A, Bono V, Costagliola C, Marfella R, et al. High HDL cholesterol: A risk factor for diabetic retinopathy? Findings from NO BLIND study. Diabetes Research and Clinical Practice. 2019; 150: 236–244.
[86]
Kohner EM, Aldington SJ, Stratton IM, Manley SE, Holman RR, Matthews DR, et al. United Kingdom Prospective Diabetes Study, 30: diabetic retinopathy at diagnosis of non-insulin-dependent diabetes mellitus and associated risk factors. Archives of Ophthalmology (Chicago, Ill.: 1960). 1998; 116: 297–303.
[87]
Prasad R, Adu-Agyeiwaah Y, Floyd JL, Asare-Bediako B, Li Calzi S, Chakraborty D, et al. Sustained ACE2 Expression by Probiotic Improves Integrity of Intestinal Lymphatics and Retinopathy in Type 1 Diabetic Model. Journal of Clinical Medicine. 2023; 12: 1771.
[88]
Degoma EM, Rader DJ. Novel HDL-directed pharmacotherapeutic strategies. Nature Reviews. Cardiology. 2011; 8: 266–277.
[89]
Yaribeygi H, Maleki M, Reiner Ž, Jamialahmadi T, Sahebkar A. Mechanistic View on the Effects of SGLT2 Inhibitors on Lipid Metabolism in Diabetic Milieu. Journal of Clinical Medicine. 2022; 11: 6544.
[90]
Fu Z, Chen CT, Cagnone G, Heckel E, Sun Y, Cakir B, et al. Dyslipidemia in retinal metabolic disorders. EMBO Molecular Medicine. 2019; 11: e10473.
[91]
Tahara A, Takasu T, Yokono M, Imamura M, Kurosaki E. Characterization and comparison of sodium-glucose cotransporter 2 inhibitors in pharmacokinetics, pharmacodynamics, and pharmacologic effects. Journal of Pharmacological Sciences. 2016; 130: 159–169.
[92]
Tahara A, Takasu T, Yokono M, Imamura M, Kurosaki E. Characterization and comparison of sodium-glucose cotransporter 2 inhibitors: Part 2. Antidiabetic effects in type 2 diabetic mice. Journal of Pharmacological Sciences. 2016; 131: 198–208.
[93]
Tahara A, Takasu T, Yokono M, Imamura M, Kurosaki E. Characterization and comparison of SGLT2 inhibitors: Part 3. Effects on diabetic complications in type 2 diabetic mice. European Journal of Pharmacology. 2017; 809: 163–171.
[94]
Grempler R, Thomas L, Eckhardt M, Himmelsbach F, Sauer A, Sharp DE, et al. Empagliflozin, a novel selective sodium glucose cotransporter-2 (SGLT-2) inhibitor: characterisation and comparison with other SGLT-2 inhibitors. Diabetes, Obesity & Metabolism. 2012; 14: 83–90.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top