Next Article in Journal
1,6-Hexanediol Is Inducing Homologous Recombination by Releasing BLM from Assemblysomes in Drosophila melanogaster
Next Article in Special Issue
Small Extracellular Vesicles and Oxidative Pathophysiological Mechanisms in Retinal Degenerative Diseases
Previous Article in Journal
Linking Gene Fusions to Bone Marrow Failure and Malignant Transformation in Dyskeratosis Congenita
Previous Article in Special Issue
The Healthy and Diseased Retina Seen through Neuron–Glia Interactions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Manifestation of Pathology in Animal Models of Diabetic Retinopathy Is Delayed from the Onset of Diabetes

by
Samuel Cubillos
1 and
Andrius Kazlauskas
2,*
1
University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
2
Lions Illinois Eye Research Institute, Chicago, IL 60612, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(3), 1610; https://doi.org/10.3390/ijms25031610
Submission received: 18 December 2023 / Revised: 18 January 2024 / Accepted: 22 January 2024 / Published: 28 January 2024
(This article belongs to the Special Issue Retinal Diseases and Cell Signaling)

Abstract

:
Diabetic retinopathy (DR) is the most common complication that develops in patients with diabetes mellitus (DM) and is the leading cause of blindness worldwide. Fortunately, sight-threatening forms of DR develop only after several decades of DM. This well-documented resilience to DR suggests that the retina is capable of protecting itself from DM-related damage and also that accumulation of such damage occurs only after deterioration of this resilience. Despite the enormous translational significance of this phenomenon, very little is known regarding the nature of resilience to DR. Rodent models of DR have been used extensively to study the nature of the DM-induced damage, i.e., cardinal features of DR. Many of these same animal models can be used to investigate resilience because DR is delayed from the onset of DM by several weeks or months. The purpose of this review is to provide a comprehensive overview of the literature describing the use of rodent models of DR in type-1 and type-2 diabetic animals, which most clearly document the delay between the onset of DM and the appearance of DR. These readily available experimental settings can be used to advance our current understanding of resilience to DR and thereby identify biomarkers and targets for novel, prevention-based approaches to manage patients at risk for developing DR.

1. Introduction

Diabetic retinopathy (DR) is a chronic sequelae of diabetes mellitus (DM) and a leading cause of blindness worldwide [1,2,3]. Early detection of DR symptoms is crucial for timely intervention and prevention of irreversible vision loss [2]. In humans, the diagnosis of DR is based exclusively on the presence of retinal vascular abnormalities such as microaneurysms, hemorrhages, exudates, leakage resulting in edema, and neovascularization [1,2,3,4]. Although neural and visual abnormalities also develop, they are not considered in the diagnosis [1,2].
In contrast, all types of retinal dysfunction (vascular, neural, and visual) are used to assess DR in animal models [4]. Common pathologic features of DR in rodents affect both the vascular and neural compartments of the retina. Vascular abnormalities include inflammation of the endothelium, increased retinal vascular permeability, and manifestation of acellular capillaries. Within the neural retina, diagnostic features of DR include the death of various neural cell types (thinning of the retina), reactive gliosis, and neural dysfunction. These outcomes are typically detected using immunofluorescence, immunohistochemical staining, electroretinogram (ERG), and optical coherence tomography (OCT). While rodent models develop the early stages of DR, they do not develop the advanced, proliferative stage of DR, which involves neovascularization of the retina. Retinal neovascularization can be modeled with the oxygen-induced retinopathy protocol [5]; however, animals are not DM in this experimental setting and, hence, constitute a major difference from the clinical scenario. Thus, DM induces the early stages of retinal damage in experimental animals, similar to what occurs in humans.
Rodent models are some of the most useful models for studying the pathology of DR. Not only does the pathogenesis closely mimic the retinopathy-free period and then initial phases of DR in a fraction of the time (Figure 1), but rodents are also extraordinarily versatile when it comes to gene modification. Rodent models have been and continue to be instrumental in documenting the duration of DM necessary for first detecting DM-related damage in both the vascular and neural compartments of the retina, and the progressive nature of such damage as the duration of DM is prolonged. Such descriptive studies are the bedrock for both mechanistic studies seeking to understand the nature of DM-induced damage as well as translational efforts seeking to identify agents that prevent and even reverse such damage. This large body of the literature, which is focused on DR, i.e., damage to the retina that becomes detectable after weeks or months of DM and progresses as the duration of DM is prolonged, only indirectly addresses resilience to DR.
Recent studies focused on resilience to DR have provided additional evidence for its existence and have begun to elucidate the underlying mechanism [6,7]. In both streptozotocin-induced type 1 diabetic (T1D) and db/db genetically modified type 2 diabetic (T2D) mice, durations of DM that were insufficient to cause detectable damage increased expression of NAD(P)H Quinone Dehydrogenase 1, Sod2, Gclc, and other antioxidant defense genes within the retina [7]. Furthermore, the retinal vessels of these DM mice became resistant to oxidative stress-induced death. As the duration of DM increased, resilience was lost—the expression of antioxidant defense genes declined; the vasculature became more (instead of less) vulnerable to death, and both neural and vascular hallmarks of DM appeared. Thus, the delay in developing DR corresponded to a period of resilience to DM-associated damage.
Figure 1. DR pathogenesis involves loss of resilience, which is a prerequisite for the subsequent accumulation of damage within the retina. In both humans and experimental animals, a retinopathy-free period precedes the appearance of DR. Recent discoveries in both T1D and T2D mice indicate that the retinopathy-free period is associated with enhanced tolerance of insults that cause damage to the retina, such as increased mitochondrial oxidative stress and cytokine production [7]. Loss of resilience sets the stage for progressive accumulation of retinal damage, which indicates the existence of DR.
Figure 1. DR pathogenesis involves loss of resilience, which is a prerequisite for the subsequent accumulation of damage within the retina. In both humans and experimental animals, a retinopathy-free period precedes the appearance of DR. Recent discoveries in both T1D and T2D mice indicate that the retinopathy-free period is associated with enhanced tolerance of insults that cause damage to the retina, such as increased mitochondrial oxidative stress and cytokine production [7]. Loss of resilience sets the stage for progressive accumulation of retinal damage, which indicates the existence of DR.
Ijms 25 01610 g001
Of the two commonly used rodent models of DR, mice are used more often than rats. While the eye of a rat is larger than a mouse’s eye, the lower cost and greater selection of genetically modified mice are some of the reasons why mice are more popular than rats in the field of DR research.
In this review, we provide a comprehensive overview of the current literature demonstrating that DR does not develop coincident with the onset of DM in murine and rat models of T1D and T2D. Rather, DR becomes detectable after weeks or months of DM, whereupon its severity increases as the duration of DM is prolonged. The progressive nature of DR suggests the existence of resilience, which protects from and/or repairs DM-induced damage, and that deterioration of such systems is an essential component of DR pathogenesis.

2. The Literature Review

2.1. Murine Models of Type-1 Diabetic-Induced Retinopathy

T1D rodent models mimic the autoimmune destruction of pancreatic β-cells, leading to insulin deficiency. Mouse models such as the streptozotocin (STZ)-induced diabetic mouse and rat, non-obese diabetic (NOD) mouse, and alloxan-induced mouse models have been extensively employed in investigations of DR. STZ and alloxan are chemical compounds commonly used to induce T1D in mice through intravenous or intraperitoneal injection. Although the exact mechanism behind alloxan’s effects is not fully understood, both of these chemicals preferentially affect the pancreatic β-cells due to the cells’ elevated levels of the GLUT2 glucose transporter. STZ and alloxan enter β-cells through GLUT2 and are metabolized into toxic side-products that damage DNA, cause oxidative stress, and ultimately lead to the death of these insulin-producing cells. At a concentration of STZ that kills Glut2-expressing cells, it is non-cytotoxic toward cells that do not express its transporter [8]. Multiple injections of moderate doses of STZ cause gradual death of beta cells that results from the combined action of STZ-mediated cytotoxicity (alkylating DNA) and immune attack [9,10,11]. It is this destruction of insulin-producing cells by which STZ induces DM; insulin supplementation or a pancreatic islet transplant prevents DM in STZ-treated animals [12]. Similarly, the breakdown of the blood-retinal barrier does not occur in STZ-treated animals that are given insulin [12,13,14]. While STZ has effects unrelated to eliminating the insulin-producing cells, such side effects are more common in single, high-dose STZ protocol that also induces DM [15]. NOD mice, on the other hand, exhibit T1D-like phenotypes due to the interplay of several factors that result in genetic susceptibility, autoimmune dysregulation, inflammatory infiltration, and environmental triggers that lead to the destruction of pancreatic β-cells [4,16]. Unlike the way DR is diagnosed in humans (based exclusively on vascular parameters), the presence of DR in T1D is evaluated considering vascular, neural, and visual dysfunction. The earliest signs of DR in T1D mice have been reported as early as one week after DM onset, as can be seen in Table 1.
Throughout the literature on T1D mouse models, there has been consistent notice of a delay between the onset of DM and outcomes seen in DR, although of variable length. Once these outcomes have developed, they persist and intensify as DM and DR continue to progress, with the exception of leukostasis and inflammation [17]. Consequently, the longer the animal has DM, the greater the differences will be between the DM and non-DM groups. For this reason, investigators focused on DM-induced damage of the retina, prolonging the duration of DM until after resilience had deteriorated. The earliest signs of vascular dysfunction were seen following one week of DM and consisted of degenerated pericytes, acellular capillaries, and minor vessel proliferation [18,19,20,21,22,23,24,25,26,27,28,29]. Neural dysfunction, which was measured with electroretinogram (ERG), was absent after 1 week of DM and detectable two weeks after the onset of DM.
Table 1. Articles discussing Type-1 diabetic mouse models.
Table 1. Articles discussing Type-1 diabetic mouse models.
Authors, (Year)ModelCollected TissueMechanism of Analysis in Diabetic RetinopathyMonitoring of DR ProgressionEarliest Detection of Diabetic RetinopathyResult/Outcome
Weerasekera, L.Y., Balmer, L.A., Ram, R., et al. (2015) [18]STZ
Alloxan
RetinaFlat-mount whole retina
Microscopy
Microarray
Retinal trypsin digest
Yes1 week of diabetesMice retinas showed degenerated pericytes, acellular capillaries, minor vascular proliferation, gliosis of Müller cells, and loss of ganglion cells (GC).
Muriach, M., Bosch-Morell, F., Alexander, G., et al. (2006) [30]AlloxanRetinaOxidative stress markers and functional tests
ERG
Morris water maze test
None2 weeks of diabetesMice retinas showed that ocular malondialdehyde (MDA) concentration was higher than controls, whereas GSH concentration and GPx activity decreased in the diabetic retina. ERG B-wave amplitude decreased in diabetic animals compared to controls.
Lee, Y.J., Jung, S.H., Hwang, J., et al. (2017) [31]STZRetinaFluorescein angiography
Confocal microscopy
ELISA
None2 weeks of diabetesMice retinas showed higher levels of extravasation of FITC-dextran, which were observed in the retinas of diabetic mice. ROS levels were also elevated in diabetic compared to normal mouse retinas.
Leal, E.C., Manivannan, A., Hosoya, K., et al. (2007) [32]STZRetinaFlat-mount microscopy
Confocal microscopy
Evans Blue Western blot
Flow cytometry
Immunohistochemistry
Yes2 weeks of diabetesMice retinas showed blood-retinal barrier (BRB) breakdown, leukocyte number, ICAM-1 protein levels, eNOS levels, and Nitrotyrosine immunostaining, which increased compared to controls.
He, J., Wang, H., Liu, Y., et al. (2015) [33]STZ
Ins2Akita
RetinaWestern blot
Immunofluorescence
RT-PCR
None2 weeks of diabetesMice retinas showed significantly increased numbers of leukocytes adhering to retinal vasculature compared to controls.
Sergeys, J., Etienne, I., Van Hove, I., et al. (2019) [17]STZRetinaVisual acuity
Contrast sensitivity
ERG
SD-OCT
Leukostasis
Immunohistochemistry
FITC-BSA
Yes2 weeks of diabetesMice retinas showed a rapid increase in leukostasis retinal vasculature compared to controls.
Feit-Leichman, R.A., Kinouchi, R., Takeda, M., et al. (2005) [27]STZRetinaTUNEL assay
Western blot
Photomicrography
Trypsin-digest
Transmission electron microscopy
Yes2 weeks of diabetesMice retinas showed an increase in TUNEL-positive cells in the GCL compared to controls.
Lee, S., Harris, N.R. (2008) [34]NODRetinaRetinal arteriolar diameter analysis
RBC velocities
None3 weeks of diabetesHyperglycemic non-obese diabetic (NOD) mice retinas demonstrated retinal arterioles closely paired with venules that were roughly 16% more constricted, and flow was roughly 33% reduced compared to normoglycemic NOD controls.
Rangasamy, S., McGuire, P.G., Franco Nitta, C., et al. (2014) [20]STZRetinaCell Culture
RT-qPCR
ELISA
Confocal microscopy
Flow Cytometry
Electrical Cell-Substrate Impedance Sensing (ECIS)
Quantitative Assessment of BRB Permeability
None4 weeks of diabetesMice retinas showed numerous GFP+ round cells (monocytes/macrophages), without processes, present within the retinal tissue and outside the vasculature.
Sasaki, M., Ozawa, Y., Kurihara, T., et al. (2010) [35]STZRetinaERG
Immunoblotting
Histologic analysis
TUNEL and caspase-3 staining
None4 weeks of diabetesMice retinas demonstrated DHE staining in all retinal layers with decreased ERG amplitude of OPs reflecting inner retinal functional status. Synaptophysin protein and BDNF were decreased, while ERK activation was elevated compared to controls.
Kurihara, T., Ozawa, Y., Shinoda, K., et al. (2006) [36]STZRetinaImmunoblotting
RT-qPCR
ERG
None4 weeks of diabetesMice retinas showed increased retinal levels of angiotensin II and its receptor AT1R, enhanced ERK phosphorylation, prolonged implicit time of OP2 and OP3, and lower amplitude OP3 compared to controls.
Li, C.-R., Sun, S.-G. (2010) [19]NODRetinaELISA
Transmission electron microscopy
Yes4 weeks of diabetesFemale NOD mice retinas demonstrated a significantly increased membrane thickness, microvascular occlusion, and perivascular edema in retinal capillary basement, chromatin margination with nuclear deformation in the endothelial and pericyte cells, and an irregular aggregation of nuclear chromatin with vacuolar degeneration in the ganglion cells compared to controls.
Wang, Q., Navitskaya, S., Chakravarthy, H., et al. (2016) [22]STZRetinaRT-qPCR
TaqMan miRNA Assays
Confocal microscopy
Fluorescence microscopy
None4 weeks of diabetesMice retinas showed increased vascular permeability in the retina compared to controls.
Miloudi, K., Oubaha, M., Ménard, C., et al. (2019) [37]STZRetinaRT-qPCR
Western Blot
Yes4 weeks of diabetesMice retinas demonstrated increased levels of Jag1, DLL4, and Notch1 transcripts compared to controls.
Yoon, C.H., Choi, Y.E., Cha, Y.R., et al. (2016) [21]STZRetinaImmunofluorescence
Confocal microscopy
TUNEL assay
Flow cytometry
RT-qPCR
In-vitro angiogenesis
None4 weeks of diabetesMice retinas showed decreased capillary area/retinal area ratio, increased number of acellular capillaries, and decreased capillary diameter compared to controls.
Chung, Y.R., Choi, J.A., Koh, J.Y., et al. (2017) [38]STZRetinaFluorescein angiography
FITC staining
Immunohistochemistry
Western blot
None4 weeks of diabetesMice retinas showed increased expression levels of GRP78, pPERK, peIF2α, MCP-1, cytokines, and TNFα in retinal tissues compared to controls.
Barber, A.J., Antonetti, D.A., Kern, T.S., et al. (2005) [39]Ins2AkitaRetinaGFAP staining
Immunoreactivity
Yes4 weeks of diabetesHyperglycemic Ins2Akita mice retinas showed increased apoptosis identified by immunoreactivity for active caspase-3 compared to controls.
Kumar, S., Zhuo, L. (2010) [40]STZRetinaConfocal scanning laser ophthalmoscopic imaging
Immunohistochemistry
RT-qPCR
Yes5 weeks of diabetesMice retinas showed increased GFAP-GFP transgene expression, average TFGI, and average number of astrocytic cell bodies compared to controls.
Sohn, E.H., Van Dijk, H.W., Jiao, C., et al. (2016) [41]STZ
Db/db
RetinaSD-OCT
Immunohistochemistry
Trypsin-digest
DAPI nuclear counterstaining
Yes6 weeks of diabetesMice retinas showed significant thinning of the nerve fiber layer (NFL) and GCL on OCT, and GC loss but no reduction in density compared to controls.
Nagai, N., Izumi-Nagai, K., Oike, Y., et al. (2007) [42]STZRetinaRT-qPCR
Western blot
RT-qPCR
ELISA
None7 weeks of diabetesMice retinas showed increased mRNA expression of angiotensinogen, AT1-R, AT2-R, upregulated protein levels of angiotensin II, AT1-R, AT2-R, ICAM-1, VEGF, enhanced nuclear translocation of NF-κB p65, and MCP-1 compared to controls.
Zheng, L., Du, Y., Miller, C., et al. (2007) [43]STZRetinaFluorometric assay kit
Western blot
ELISA
FITC
Flat-mount and fluorescence microscopy
Trypsin digest
Immunohistochemical stains
ERG
Yes8 weeks of diabetesMice retinas showed an increase in eNOS production compared to controls.
Vincent, J.A., Mohr, S. (2007) [44]STZRetinaMinocycline studies
Tissue cultures
Retinal and cell lysates
Caspase activity assay
Interleukin-1B ELISA
Histological assessment of retinal vascular pathology
None8 weeks of diabetesMice retinas showed significantly higher levels of caspase-1 activity and IL-1β levels compared to controls.
Mohr, S., Xi, X., Tang, J., et al. (2002) [45]STZRetinaWestern blot
TUNEL assay
Yes8 weeks of diabetesMice retinas demonstrated increased activity of several caspases -1, -2, -4, -5, -6, -8, and -9 compared to controls.
Kady, N.M., Liu, X., Lydic, T.A., et al. (2018) [23]STZRetinaRT-qPCR
Immunoblotting
siRNA
Western blot
Immunocytochemistry
Immunogold Electron Microscopy
Mass Spectrometry
None8 weeks of diabetesMice retinas showed significant increase in vascular permeability and downregulation of endothelial ELOVL4 expression compared to controls.
Saadane, A., Lessieur, E.M., Du, Y., et al. (2020) [46]STZRetinaSD-OCT
ERG
Western blot
Lucigenin assay
Yes8 weeks of diabetesMice retinas showed increased vascular permeability and reduced ELOVL4 compared to controls.
McVicar, C.M., Ward, M., Colhoun, L.M., et al. (2015) [47]STZRetinaRT-qPCR
Confocal microscopy
Fluorescent confocal microscopy
Retinal flat mounts
Epifluorescent microscopy
Yes8 weeks of diabetesMice retinas showed accumulation of methylglyoxal (MG)-derived adducts and MG levels compared to controls.
Ouyang, H., Mei, X., Zhang, T., et al. (2018) [24]STZRetinaEvans blue
Retinal histology
Western blot
ELISA
Immunofluorescence
Immunohistochemical analysis
None8 weeks of diabetesMice retinas showed increased leakage of Evans blue dye, decreased percentage of claudin-1 and -9, significant thinning of INL and ONL, increased percentage of iba1/actin ratio, increased nuclear expression of NFκBp65, increased mRNA expression of inflammatory cytokines, elevated levels and mRNA expression of VEGF compared to controls.
Liu, H., Tang, J., Du, Y., et al. (2015) [48]STZRetinaFluorescence microscopy
Elastase digestion and PAS staining
Photomicrography
Immunoblotting
Chromatography
Virtual optokinetics
None8 weeks of diabetesMice retinas showed increased superoxide generation and expression of inflammatory proteins compared to controls.
Du, Y., Cramer, M., Lee, C.A., et al. (2015) [49]STZRetinaSpatial frequency threshold
Contrast sensitivity
Yes8 weeks of diabetesMice retinas showed decreased spatial frequency and increased superoxide production compared to controls.
Gaucher, D., Chiappore, J.A., Pâques, M., et al. (2007) [4]AlloxanRetinaFluorescein Angiography
ERG
TUNEL assay
Immunocytochemistry
Scanner laser ophthalmoscope (SLO)
Yes8 weeks of diabetesMice retinas showed increased superoxide, iNOS, and b-wave amplitude seen on ERG compared to controls.
Li, G., Veenstra, A.A., Talahalli, R.R., et al. (2012) [50]STZRetinaFITC
Immunohistochemistry
RT-qPCR
Coculture
flow cytometry
mREC luminescence
Trypsin digest
None8–10 weeks of DMMice retinas showed increased superoxide production, mRNA for iNOS, COX-2, TNF-α, ICAM-1, leukostasis, and dead mRECs compared to controls.
Gubitosi-Klug, R.A., Talahalli, R., Du, Y., et al. (2008) [51]STZRetinaTrypsin digest
FITC
Fluorescence microscopy
Western blot
Immunohistochemistry
Yes12 weeks of diabetesMice retinas showed increased leukostasis and superoxide production compared to controls.
Gastinger, M.J., Singh, R.S., Barber, A.J. (2006) [52]Ins2AkitaRetinaTUNEL analysis
Immunohistochemistry
None12 weeks of diabetesThe Ins2Akita mice retinas showed 72% more TUNEL-positive nuclei, suggesting increased apoptosis, than controls.
Gastinger, M.J., Kunselman, A.R., Conboy, E.E., et al. (2008) [53]Ins2AkitaRetinaMeasuring density and morphology of retinal ganglion cells.
Immunohistochemistry
Confocal imaging
Morphometric analysis via Sholl analysis
None12 weeks of diabetesMice retinas showed loss of GC from the peripheral retina, morphologic changes in dendrites from surviving large ON-type cells, increased vascular permeability, microglial cell reactivity, leukostasis, caspase-3 activity, and reduced insulin receptor kinase activity compared to controls.
Shaw, S.G., Boden, J.P., Biecker, E., et al. (2006) [54]NODRetinaConfocal microscopy
RT-PCR
None16 weeks of diabetesNOD mice retinas showed regional microvascular degeneration and/or lack of perfusion in the retina with approximately 50% reduced vascular filling compared to wild-type controls.
Liu, Q., Zhang, F., Zhang, X., et al. (2018) [55]STZRetinaImmunofluorescence staining
Fluorescence microscopy
Concanavalin A lectin perfusion
Evans blue
Western blot
RT-qPCR
None32 weeks of diabetesMice retinas showed mild to moderate reactive gliosis, upregulation of Nrf2 and NLRP3 inflammasome, enhanced ROS formation and CellROX fluorescence, increased leukostasis and vascular leakage, and increased expression of NLRP3, Caspase-1 p20, IL-1β p17, and ICAM-1 compared to controls.
Joussen, A.M., Poulaki, V., Le, M.L., et al. (2004) [28]STZRetinaEvans blue
Trypsin digest
Electron microscopy
None44 weeks of diabetesMice retinas showed increased number of adherent leukocytes, endothelial cell injury, and BRB breakdown compared to controls.
Once outcomes such as visual acuity decreases, reduced contrast sensitivity, ERG changes, NFL-GCL thinning, RGC and amacrine cell neurodegeneration, and gliosis have developed, they persist and intensify with the duration of DM, with the exception of leukostasis and inflammation [17]. Although a variety of markers have been used throughout the literature to study the role of inflammation in DR, some of the more common markers included an elevated number of leukocytes, leukocyte adhesion, microglial infiltration, production of cytokines, eNOS, and several caspases [20,32,33,38,43,44,45]. Every murine model of DR shows the progression of the disease, i.e., the extent and nature of the damage of the retina increases, but not the transition from NPDR to PDR.

2.2. Murine Models of Type-2 Diabetic-Induced Retinopathy

T2D rodent models mimic insulin resistance and impaired glucose metabolism. The T2D models rely on various genetic modifications that lead to DM and thus show a much wider variation between strains. The leptin receptor-deficient (db/db) mouse and high-sugar diet-induced models are some of the commonly used models in this realm [4,16]. Because the leptin receptor in db/db mice is non-functional, the leptin signaling pathway is disrupted and leads to dysregulation of appetite and satiety [4,16]. Such mice become obese, resistant to insulin, hyperinsulinemic, and develop pancreatic β-cell dysfunction that causes T2D symptoms. In a similar fashion, high-sugar diet-induced models of T2D are fed specific diets that lead to obesity and glucose dysregulation, causing overt diabetic phenotypes [4]. Db/db mice develop DM between 4–12 weeks of life, but commonly at 8 weeks. In contrast to the terminology found in the literature on T1D STZ-induced mouse models, the literature studying the db/db model uses “weeks of age” instead of time from DM onset. This is because the onset of DM is more difficult to pinpoint in this T2D mouse model as compared to the STZ-induced T1D models. In these scenarios, the approximate duration of DM is 8 weeks shorter than the age of the mice. The section earliest detection of diabetic retinopathy, of Table 2 depicts this shift in terminology.
Damage to the retina is time-sensitive, with prolonged DM resulting in worsening manifestations of DR without any worsening of DM, suggesting the progressive nature of DR. In T2D mice, the earliest indication of retinal dysfunction occurred at 10 weeks of age (after approximately 2 weeks of DM) and included signs of apoptotic changes in the INL, ONL, and GCL retina layers, along with increased vascular permeability and leakage [41,56,57,58].
Table 2. Articles of Note discussing Type-2 diabetic mouse models (db/db).
Table 2. Articles of Note discussing Type-2 diabetic mouse models (db/db).
Authors, (Year)ModelCollected TissueMechanism of Analysis in Diabetic RetinopathyMonitoring of DR ProgressionEarliest Detection of Diabetic RetinopathyResult/Outcome
Samuels, I.S., Bell, B.A., Pereira, A., et al. (2014) [59]Db/dbRetinaERG
OCT
Yes8 weeks of age (DM onset at 4 weeks of age)Mice retinas showed reduced b-wave amplitudes compared to controls.
Bogdanov, P., Corraliza, L., Villena, J.A., et al. (2014) [60]Db/dbRetinaERG
Transendothelial electrical resistance
Mitophagy
TBH challenge assay
Seahorse analysis
Western blot
PCR analysis
RNA sequencing
LDH
Fluorescence-activating cell sorting
Glucose consumption rate
Tissue culture
Yes8 weeks of ageMice retinas showed higher levels of glutamate, glial activation, TUNEL-positive cells, activated caspase-3, thickening in ONL and INL, and decreased number of cells in GCL compared to controls.
Hernández, C., Bogdanov, P., Corraliza, L., et al. (2016) [56]Db/dbRetinaERG
Immunohistochemistry
TUNEL assay
Albumin leakage monitoring
Western blot
None10 weeks of ageMice retinas showed higher percentage of apoptotic cells in whole retina and Outer nuclear layer (ONL), Inner nuclear layer (INL), and Ganglion Cell (GC) layer, overexpression of VEGF and IL-1β, increased blood-retinal barrier (BRB) breakdown, glial activation, levels of TUNEL-positive cells, and lower amplitudes in a- and b-waves compared to controls.
Ly, A., Scheerer, M.F., Zukunft, S., et al. (2014) [61]Db/dbRetinaImmunofluorescence
RT-qPCR
None10 weeks of ageMice retinas showed decreased levels of proteins related to synaptic transmission and cell signaling, increased VGLUT1, and decreased Slc17a7/VGLUT1 compared to controls.
Sohn, E.H., Van Dijk, H.W., Jiao, C., et al. (2016) [41]Db/db
STZ
RetinaSD-OCTYes10 weeks of ageMice retinas showed thinning in NFL and GCL compared to controls.
Tadayoni, R., Paques, M., Gaudric, A., et al. (2003) [62]Db/dbRetinaModified epifluorescence microscopy
FITC-labeled RBC
Rhodamine-labeled WBC
Yes10 weeks of ageMice retinas showed higher RBC velocity in retinal capillaries compared to controls.
Byrne, E.M., Llorián-Salvador, M., Lyons, T.J., et al. (2021) [57]Db/dbRetinaAlbumin and Isolectin B4 staining
pJAK1 staining
None12 weeks of ageMice retinas showed BRB leakage and elevated pJAK1 levels compared to controls.
Jung, E., Kim, J., Kim, C.S., et al. (2015) [63]Db/db
Oxygen-induced retinopathy
RetinaImmunohistochemistry
TUNEL and FITC-dextran leakage
None12 weeks of ageMice retinas showed increased E/P ratio, TUNEL-positive pericytes, and endothelial cells compared to controls.
Tang, L., Zhang, Y., Jiang, Y., et al. (2011) [64]Db/dbRetinaWestern blot
Fluorometric assay
Light microscopy
None14 weeks of ageMice retinas showed thinner whole central retina, INL, photoreceptor layer, decreased GC, elevated protein expression of ER stress biomarkers, BiP, PERK, ATF6, and active caspase-12 compared to controls.
Yang, Q., Xu, Y., Xie, P., et al. (2015) [65]Db/dbRetinaPattern electroretinogram (PERG)
Fundus fluorescein angiography (FFA)
OCT
Immunohistochemistry
Yes20 weeks of ageMice retinas showed reduced P1 amplitudes and increased TUNEL-positive cells localized in GCL and INL compared to controls.
Liu, M., Pan, Q., Chen, Y., et al. (2015) [66]Db/dbRetinaPAS staining
Immunofluorescent staining
Light microscopy
None20 weeks of ageMice retinas showed reduced whole retinal thickness, increased acellular capillaries, increased CAS-3 expression in photoreceptor layer, and increased MMP-2 and -9 mRNA expression compared to controls.
Clements, R.S. Jr., Robison, W.G. Jr., Cohen, M.P. (1998) [67]Db/dbRetinaHistologic analysis
Morphometric analysis
None22 weeks of ageMice retinas showed microvessel basement membrane thickening and 20% greater fractional basement membrane area compared to controls.
Sampedro, J., Bogdanov, P., Ramos, H., et al. (2019) [58]Db/dbRetinaERG in vivo
TUNEL
Evans blue
Immunofluorescence
Western Blot
RT-PCR
None24 weeks of ageMice retinas showed lower amplitude a- and b-waves, aberrant GFAP extent, increased TUNEL-positive cells in INL and ONL, decreased total cells in GCL, INL, and ONL, upregulation of several proinflammatory cytokines (mRNA and protein), including IL-1β and IL-6, elevated TNFα, upregulation of NLRP3 inflammasome pathway, overexpression of IL-18, albumin leakage, extravasation of Evans Blue, and elevated protein levels of VEGF compared to controls.
Barile, G.R., Pachydaki, S.I., Tari, S.R., et al. (2005) [68]Db/dbRetinaERG
Immunohistochemistry, RT-qPCR, autofluorescence, ELISA
None24 weeks of ageMice retinas showed prolonged oscillatory potentials and b-waves, accelerated development of acellular capillaries and pericyte ghosts, and increased AGE accumulation in retinal extracellular matrix compared to controls.
Hinder, L.M., Sas, K.M., O’Brien, P.D., et al. (2019) [69]Db/dbRetinaELISA
Virtual-reality optokinetic tracking
None24 weeks of ageMice retinas showed features of early-stage DR, increased retinal apoptosis, and compromised visual performance compared to controls.
Hammer, S.S., Beli, E., Kady, N., et al. (2017) [70]Db/dbRetinaCell culture
RT-qPCR
Oxysterol measurement
BMDM
siRNA transfection
Western blot
ELISA
None40 weeks of ageMice retinas showed increased acellular capillaries, expression of TNFα, decreased LXR-SIRT signaling, levels of LXRα, Sirtuin 1, LXR, and CAC migration compared to controls.
Beli, E., Yan, Y., Moldovan, L., et al. (2018) [71]Db/dbRetinaTrypsin digest
Immunofluorescence staining
qRT-PCR
Yes44 weeks of ageMice retinas showed increased numbers of acellular capillaries, IBA-1 positive cells, and CD45+ cell infiltration compared to controls.
Kern, T.S., Engerman, R.L. (1996) [72]Db/dbRetinaTrypsin digestYes60 weeks of ageMice retinas showed increased acellular capillaries compared to controls.
Cheung, A.K., Fung, M.K., Lo, A.C., et al. (2005) [73]Db/dbRetinaImmunohistochemistry
Retinal sections
Digital photographing
None60 weeks of ageMice retinas showed increased AR-immunoreactivity, expression of AR levels, formation of nitrotyrosine, GFAP Muller cell processes in Inner limiting membrane (ILM), Inner plexiform layer (IPL), outer plexiform layer (OPL), PCNA-positive cells, IgG extravasation from capillaries, caspase-3 activity, and >25% decreased pericyte density compared to controls.
Once outcomes such as visual acuity decreases, reduced contrast sensitivity, ERG changes, NFL-GCL thinning, RGC and amacrine cell neurodegeneration, and gliosis have developed, they persist and intensify with the duration of DM, with the exception of leukostasis and inflammation [17]. Although a variety of markers have been used throughout the literature to study the role of inflammation in DR, some of the more common markers included the elevated number of leukocytes, leukocyte adhesion, microglial infiltration, production of cytokines, eNOS, and several caspases [20,32,33,38,43,44,45]. Every murine model of DR shows the progression of the disease, i.e., the extent and nature of the damage of the retina increases, but not the transition from NPDR to PDR.

2.3. Rat Model of Type-1 Diabetic-Induced Retinopathy

The rat model has also been in use for some time by researchers and has been shown to reproduce a similar pathology to human DM. Most commonly, STZ injections have been used to induce a non-autoimmune form of T1D. The rat model has also shown a delay in the detection of diabetic retinopathy after induction/onset of diabetes; however, it may be less than that seen in T1D mice. In contrast to the STZ-induced mice, the STZ-induced DM rats demonstrated leakage of retinal blood vessels to very small molecules such as fluorescein after as early as 2 days of DM, as can be seen in Table 3 [12,14,74]. A shorter delay in the T1D rats may suggest that they are less appropriate models for studying resilience to DR when compared to T1D mouse models. However, this conclusion is weak because different outcomes were performed in different labs to assess vascular damage.

2.4. Potential Pathways of Resilience to DR-Associated Damage

Resilience is likely to prevent those events that cause damage to the retina. These would include oxidative stress and cytokines [39,47,48,49,86,87,88,89,90,91,92,93,94]. Also, engaging endogenous systems, such as mitophagy, which prevent mitochondrial oxidative stress, are likely to be at least part of the mechanism of resilience [7].

3. Discussion

DR is a complex and progressive disease characterized by a spectrum of pathologic changes, which include retinal vascular abnormalities, inflammatory processes, and neurodegeneration. The delay between the onset of DM and the manifestation of retinal damage/dysfunction indicates the existence of resistance to DR, which must be overcome before outcomes of the disease appear.
There are multiple explanations for resilience, i.e., the absence of damage in the face of insults that are capable of causing such damage. It is possible that resilience relates to the level of sensitivity of current approaches to detect damage. Damage may commence with the onset of DM, but its extent is below the level of detection. This scenario predicts that lowering the detection limit will reduce or eliminate the period of resilience. However, technological advances that have been made to date have not demonstrated that DR manifests coincident with the onset of DM.
An alternative explanation of resilience is that cells adapt to hyperglycemia (HG) and, thereby, become resistant to its deleterious effects. Our recent publications provided compelling support for this possibility. In DM mice, resistance is transient, and its loss sets the stage for accumulation of damage, i.e., manifestation of DR. More specifically, the retinal vasculature of mice that had experienced a short duration of DM acquired resistance to oxidative stress/ischemia- or cytokine-induced death [7]. As the duration of DM was prolonged, protection waned and was replaced by increased vulnerability; i.e., there was more insult-driven death in retinal vessels from DM mice as compared with age-matched non-DM mice [7]. Finally, the appearance of vulnerability coincided with the manifestation of DR—vascular and neural dysfunctions [7].
The underlying mechanism of resilience involves increased mitophagy (clearance of dysfunctional mitochondria), which boosts mitochondrial functionality and prevents mitochondrial oxidative stress in the face of HG [6]. It resides in endothelial cells but not in pericytes of human retinal capillaries [6]. Whether resilience exists in other retinal cell types has not been investigated to date. The antioxidant component of resilience has been noted by other groups reporting that expression of RBP3 (retinol-binding protein 3), an antioxidant [89,90,91], is elevated in patients who are resistant to diabetic retinopathy [92,93]. Taken together, it appears that retinopathy is held in check during the resilience phase because the endothelium has engaged in mitophagy-based defense against the deleterious effects of HG (Figure 2). Loss of resilience is a prerequisite for accumulation of damage in the retina, which is recognized as DR.
The appreciation of resilience, or the innate ability of the retina to remain healthy in the face of DM, raises essential questions regarding the potential role of genetic susceptibility and genetic protection in the development of DR. Moreover, it opens the door to a world of novel biomarkers and potential therapeutic targets of DR that must be further explored to manage people at risk of developing DR and resisting the deleterious effects of DM. Such alternatives are necessary because the current prophylactic options are not effective for all people. This is especially crucial when considering that 26% of the diabetic US population has some form of DR [94].

Author Contributions

Conceptualization, A.K. and S.C.; methodology, S.C.; investigation, S.C.; resources, S.C. and A.K.; data curation, A.K.; writing—original draft preparation, S.C.; writing—review and editing, S.C. and A.K.; visualization, S.C.; supervision, A.K.; project administration, A.K.; funding acquisition, A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by grants from the Illinois Society to Prevent Blindness, Retina Research Foundation; National Institute of Health (EY031350, EY001792) and an unrestricted grant from the Research to Prevent Blindness Foundation.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank Yanliang Li for her contributions to the conceptualization and scope of this review article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, W.; Lo, A.C.Y. Diabetic Retinopathy: Pathophysiology and Treatments. Int. J. Mol. Sci. 2018, 19, 1816. [Google Scholar] [CrossRef] [PubMed]
  2. Duh, E.J.; Sun, J.K.; Stitt, A.W. Diabetic retinopathy: Current understanding, mechanisms, and treatment strategies. JCI Insight 2017, 2, e93751. [Google Scholar] [CrossRef]
  3. Olivares, A.M.; Althoff, K.; Chen, G.F.; Wu, S.; Morrisson, M.A.; DeAngelis, M.M.; Haider, N. Animal Models of Diabetic Retinopathy. Curr. Diabetes Rep. 2017, 17, 93. [Google Scholar] [CrossRef]
  4. Gaucher, D.; Chiappore, J.-A.; Pâques, M.; Simonutti, M.; Boitard, C.; Sahel, J.A.; Massin, P.; Picaud, S. Microglial changes occur without neural cell death in diabetic retinopathy. Vis. Res. 2007, 47, 612–623. [Google Scholar] [CrossRef]
  5. E Smith, L.; Wesolowski, E.; McLellan, A.; Kostyk, S.K.; D’Amato, R.; Sullivan, R.; D’Amore, P.A. Oxygen-induced retinopathy in the mouse. Investig. Ophthalmol. Vis. Sci. 1994, 35, 101–111. [Google Scholar]
  6. Serikbaeva, A.; Li, Y.; Ganesh, B.; Zelkha, R.; Kazlauskas, A. Hyperglycemia Promotes Mitophagy and Thereby Mitigates Hyperglycemia-Induced Damage. Am. J. Pathol. 2022, 192, 1779–1794. [Google Scholar] [CrossRef]
  7. Li, Y.; Baccouche, B.; Del-Risco, N.; Park, J.; Song, A.; McAnany, J.J.; Kazlauskas, A. The Slow Progression of Diabetic Retinopathy Is Associated with Transient Protection of Retinal Vessels from Death. Int. J. Mol. Sci. 2023, 24, 10869. [Google Scholar] [CrossRef]
  8. Schnedl, W.J.; Ferber, S.; Johnson, J.H.; Newgard, C.B. STZ Transport and Cytotoxicity: Specific Enhancement in GLUT2-Expressing Cells. Diabetes 1994, 43, 1326–1333. [Google Scholar] [CrossRef]
  9. Kolb, H. Mouse models of insulin dependent diabetes: Low-dose streptozocin-induced diabetes and nonobese diabetic (NOD) mice. Diabetes/Metabolism Res. Rev. 1987, 3, 751–778. [Google Scholar] [CrossRef]
  10. Like, A.A.; Rossini, A.A. Streptozotocin-Induced Pancreatic Insulitis: New Model of Diabetes Mellitus. Science 1976, 193, 415–417. [Google Scholar] [CrossRef] [PubMed]
  11. Weide, L.G.; E Lacy, P. Low-Dose Streptozocin-Induced Autoimmune Diabetes in Islet Transplantation Model. Diabetes 1991, 40, 1157–1162. [Google Scholar] [CrossRef]
  12. Krupin, T.; Waltman, S.R.; Szewczyk, P.; Koloms, B.; Farber, M.; Silverstein, B.; Becker, B. Fluorometric Studies on the Blood-Retinal Barrier in Experimental Animals. Arch. Ophthalmol. 1982, 100, 631–634. [Google Scholar] [CrossRef]
  13. Jones, C.W.; Cunha-Vaz, J.; Zweig, K.O.; Stein, M. Kinetic Vitreous Fluorophotometry in Experimental Diabetes. Arch. Ophthalmol. 1979, 97, 1941–1943. [Google Scholar] [CrossRef]
  14. Jones, C.W.; Cunha-Vaz, J.G.; Rusin, M.M. Vitreous fluorophotometry in the alloxan- and streptozocin-treated rat. Arch. Ophthalmol. 1982, 100, 1141–1145. [Google Scholar] [CrossRef]
  15. Furman, B.L. Streptozotocin-Induced Diabetic Models in Mice and Rats. Curr. Protoc. 2021, 1, e78. [Google Scholar] [CrossRef] [PubMed]
  16. Robinson, R.; Barathi, V.A.; Chaurasia, S.S.; Wong, T.Y.; Kern, T.S. Update on animal models of diabetic retinopathy: From molecular approaches to mice and higher mammals. Dis. Model. Mech. 2012, 5, 444–456. [Google Scholar] [CrossRef] [PubMed]
  17. Sergeys, J.; Etienne, I.; Van Hove, I.; Lefevere, E.; Stalmans, I.; Feyen, J.H.M.; Moons, L.; Van Bergen, T. Longitudinal In Vivo Characterization of the Streptozotocin-Induced Diabetic Mouse Model: Focus on Early Inner Retinal Responses. Investig. Opthalmology Vis. Sci. 2019, 60, 807–822. [Google Scholar] [CrossRef] [PubMed]
  18. Weerasekera, L.Y.; Balmer, L.A.; Ram, R.; Morahan, G. Characterization of Retinal Vascular and Neural Damage in a Novel Model of Diabetic Retinopathy. Investig. Opthalmol. Vis. Sci. 2015, 56, 3721–3730. [Google Scholar] [CrossRef] [PubMed]
  19. Li, C.-R.; Sun, S.-G. VEGF expression and cell apoptosis in NOD mouse retina. Int. J. Ophthalmol. 2010, 3, 224–227. [Google Scholar] [CrossRef] [PubMed]
  20. Rangasamy, S.; McGuire, P.G.; Nitta, C.F.; Monickaraj, F.; Oruganti, S.R.; Das, A. Chemokine Mediated Monocyte Trafficking into the Retina: Role of Inflammation in Alteration of the Blood-Retinal Barrier in Diabetic Retinopathy. PLoS ONE 2014, 9, e108508. [Google Scholar] [CrossRef] [PubMed]
  21. Yoon, C.-H.; Choi, Y.-E.; Cha, Y.R.; Koh, S.-J.; Choi, J.-I.; Kim, T.-W.; Woo, S.J.; Park, Y.-B.; Chae, I.-H.; Kim, H.-S. Diabetes-Induced Jagged1 Overexpression in Endothelial Cells Causes Retinal Capillary Regression in a Murine Model of Diabetes Mellitus. Circulation 2016, 134, 233–247. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, Q.; Navitskaya, S.; Chakravarthy, H.; Huang, C.; Kady, N.; Lydic, T.A.; Chen, Y.E.; Yin, K.-J.; Powell, F.L.; Martin, P.M.; et al. Dual Anti-Inflammatory and Anti-Angiogenic Action of miR-15a in Diabetic Retinopathy. EBioMedicine 2016, 11, 138–150. [Google Scholar] [CrossRef]
  23. Kady, N.M.; Liu, X.; Lydic, T.A.; Syed, M.H.; Navitskaya, S.; Wang, Q.; Hammer, S.S.; O’reilly, S.; Huang, C.; Seregin, S.S.; et al. ELOVL4-Mediated Production of Very Long-Chain Ceramides Stabilizes Tight Junctions and Prevents Diabetes-Induced Retinal Vascular Permeability. Diabetes 2018, 67, 769–781. [Google Scholar] [CrossRef]
  24. Ouyang, H.; Mei, X.; Zhang, T.; Lu, B.; Ji, L. Ursodeoxycholic acid ameliorates diabetic retinopathy via reducing retinal inflammation and reversing the breakdown of blood-retinal barrier. Eur. J. Pharmacol. 2018, 840, 20–27. [Google Scholar] [CrossRef]
  25. Han, Z.; Guo, J.; Conley, S.M.; Naash, M.I. Retinal Angiogenesis in the Ins2AkitaMouse Model of Diabetic Retinopathy. Investig. Opthalmol. Vis. Sci. 2013, 54, 574–584. [Google Scholar] [CrossRef]
  26. Su, L.; Ji, J.; Bian, J.; Fu, Y.; Ge, Y.; Yuan, Z. Tacrolimus (FK506) prevents early retinal neovascularization in streptozotocin-induced diabetic mice. Int. Immunopharmacol. 2012, 14, 606–612. [Google Scholar] [CrossRef] [PubMed]
  27. Feit-Leichman, R.A.; Kinouchi, R.; Takeda, M.; Fan, Z.; Mohr, S.; Kern, T.S.; Chen, D.F. Vascular Damage in a Mouse Model of Diabetic Retinopathy: Relation to Neuronal and Glial Changes. Investig. Opthalmol. Vis. Sci. 2005, 46, 4281–4287. [Google Scholar] [CrossRef] [PubMed]
  28. Joussen, A.M.; Poulaki, V.; Le, M.L.; Koizumi, K.; Esser, C.; Janicki, H.; Schraermeyer, U.; Kociok, N.; Fauser, S.; Kirchhof, B.; et al. A central role for inflammation in the pathogenesis of diabetic retinopathy. FASEB J. 2004, 18, 1450–1452. [Google Scholar] [CrossRef]
  29. A Cuthbertson, R.; E Mandel, T. The effect of murine fetal islet transplants on renal and retinal capillary basement membrane thickness. Transplant. Proc. 1987, 19, 2919–2921. [Google Scholar]
  30. Muriach, M.; Bosch-Morell, F.; Alexander, G.; Blomhoff, R.; Barcia, J.; Arnal, E.; Almansa, I.; Romero, F.J.; Miranda, M. Lutein effect on retina and hippocampus of diabetic mice. Free. Radic. Biol. Med. 2006, 41, 979–984. [Google Scholar] [CrossRef]
  31. Lee, Y.-J.; Jung, S.-H.; Hwang, J.; Jeon, S.; Han, E.-T.; Park, W.S.; Hong, S.-H.; Kim, Y.-M.; Ha, K.-S. Cysteamine prevents vascular leakage through inhibiting transglutaminase in diabetic retina. J. Endocrinol. 2017, 235, 39–48. [Google Scholar] [CrossRef] [PubMed]
  32. Leal, E.C.; Manivannan, A.; Hosoya, K.-I.; Terasaki, T.; Cunha-Vaz, J.; Ambro´sio, A.F.; Forrester, J.V. Inducible Nitric Oxide Synthase Isoform Is a Key Mediator of Leukostasis and Blood-Retinal Barrier Breakdown in Diabetic Retinopathy. Investig. Opthalmol. Vis. Sci. 2007, 48, 5257–5265. [Google Scholar] [CrossRef] [PubMed]
  33. He, J.; Wang, H.; Liu, Y.; Li, W.; Kim, D.; Huang, H. Blockade of Vascular Endothelial Growth Factor Receptor 1 Prevents Inflammation and Vascular Leakage in Diabetic Retinopathy. J. Ophthalmol. 2015, 2015, 1–11. [Google Scholar] [CrossRef]
  34. Lee, S.; Harris, N.R. Losartan and Ozagrel Reverse Retinal Arteriolar Constriction in Non-Obese Diabetic Mice. Microcirculation 2008, 15, 379–387. [Google Scholar] [CrossRef] [PubMed]
  35. Sasaki, M.; Ozawa, Y.; Kurihara, T.; Kubota, S.; Yuki, K.; Noda, K.; Kobayashi, S.; Ishida, S.; Tsubota, K. Neurodegenerative influence of oxidative stress in the retina of a murine model of diabetes. Diabetologia 2010, 53, 971–979. [Google Scholar] [CrossRef] [PubMed]
  36. Kurihara, T.; Ozawa, Y.; Shinoda, K.; Nagai, N.; Inoue, M.; Oike, Y.; Tsubota, K.; Ishida, S.; Okano, H. Neuroprotective Effects of Angiotensin II Type 1 Receptor (AT1R) Blocker, Telmisartan, via Modulating AT1R and AT2R Signaling in Retinal Inflammation. Investig. Opthalmol. Vis. Sci. 2006, 47, 5545–5552. [Google Scholar] [CrossRef] [PubMed]
  37. Miloudi, K.; Oubaha, M.; Ménard, C.; Dejda, A.; Guber, V.; Cagnone, G.; Wilson, A.M.; Tétreault, N.; Mawambo, G.; Binet, F.; et al. NOTCH1 signaling induces pathological vascular permeability in diabetic retinopathy. Proc. Natl. Acad. Sci. USA 2019, 116, 4538–4547. [Google Scholar] [CrossRef]
  38. Chung, Y.-R.; Choi, J.A.; Koh, J.-Y.; Yoon, Y.H. Ursodeoxycholic Acid Attenuates Endoplasmic Reticulum Stress-Related Retinal Pericyte Loss in Streptozotocin-Induced Diabetic Mice. J. Diabetes Res. 2017, 2017, 1–10. [Google Scholar] [CrossRef]
  39. Barber, A.J.; Antonetti, D.A.; Kern, T.S.; Reiter, C.E.N.; Soans, R.S.; Krady, J.K.; Levison, S.W.; Gardner, T.W.; Bronson, S.K. The Ins2AkitaMouse as a Model of Early Retinal Complications in Diabetes. Investig. Opthalmol. Vis. Sci. 2005, 46, 2210–2218. [Google Scholar] [CrossRef]
  40. Kumar, S.; Zhuo, L. Longitudinal in vivo imaging of retinal gliosis in a diabetic mouse model. Exp. Eye Res. 2010, 91, 530–536. [Google Scholar] [CrossRef]
  41. Sohn, E.H.; van Dijk, H.W.; Jiao, C.; Kok, P.H.B.; Jeong, W.; Demirkaya, N.; Garmager, A.; Wit, F.; Kucukevcilioglu, M.; van Velthoven, M.E.J.; et al. Retinal neurodegeneration may precede microvascular changes characteristic of diabetic retinopathy in diabetes mellitus. Proc. Natl. Acad. Sci. USA 2016, 113, E2655–E2664. [Google Scholar] [CrossRef] [PubMed]
  42. Nagai, N.; Izumi-Nagai, K.; Oike, Y.; Koto, T.; Satofuka, S.; Ozawa, Y.; Yamashiro, K.; Inoue, M.; Tsubota, K.; Umezawa, K.; et al. Suppression of Diabetes-Induced Retinal Inflammation by Blocking the Angiotensin II Type 1 Receptor or Its Downstream Nuclear Factor-κB Pathway. Investig. Opthalmol. Vis. Sci. 2007, 48, 4342–4350. [Google Scholar] [CrossRef] [PubMed]
  43. Zheng, L.; Du, Y.; Miller, C.; Gubitosi-Klug, R.A.; Kern, T.S.; Ball, S.; Berkowitz, B.A. Critical role of inducible nitric oxide synthase in degeneration of retinal capillaries in mice with streptozotocin-induced diabetes. Diabetologia 2007, 50, 1987–1996. [Google Scholar] [CrossRef] [PubMed]
  44. Vincent, J.A.; Mohr, S. Inhibition of Caspase-1/Interleukin-1β Signaling Prevents Degeneration of Retinal Capillaries in Diabetes and Galactosemia. Diabetes 2007, 56, 224–230. [Google Scholar] [CrossRef] [PubMed]
  45. Mohr, S.; Xi, X.; Tang, J.; Kern, T.S. Caspase Activation in Retinas of Diabetic and Galactosemic Mice and Diabetic Patients. Diabetes 2002, 51, 1172–1179. [Google Scholar] [CrossRef]
  46. Saadane, A.; Lessieur, E.M.; Du, Y.; Liu, H.; Kern, T.S. Successful induction of diabetes in mice demonstrates no gender difference in development of early diabetic retinopathy. PLoS ONE 2020, 15, e0238727. [Google Scholar] [CrossRef]
  47. McVicar, C.M.; Ward, M.; Colhoun, L.M.; Guduric-Fuchs, J.; Bierhaus, A.; Fleming, T.; Schlotterer, A.; Kolibabka, M.; Hammes, H.-P.; Chen, M.; et al. Role of the receptor for advanced glycation endproducts (RAGE) in retinal vasodegenerative pathology during diabetes in mice. Diabetologia 2015, 58, 1129–1137. [Google Scholar] [CrossRef]
  48. Liu, H.; Tang, J.; Du, Y.; Lee, C.A.; Golczak, M.; Muthusamy, A.; Antonetti, D.A.; Veenstra, A.A.; Amengual, J.; von Lintig, J.; et al. Retinylamine Benefits Early Diabetic Retinopathy in Mice. J. Biol. Chem. 2015, 290, 21568–21579. [Google Scholar] [CrossRef] [PubMed]
  49. Du, Y.; Cramer, M.; Lee, C.A.; Tang, J.; Muthusamy, A.; Antonetti, D.A.; Jin, H.; Palczewski, K.; Kern, T.S. Adrenergic and serotonin receptors affect retinal superoxide generation in diabetic mice: Relationship to capillary degeneration and permeability. FASEB J. 2015, 29, 2194–2204. [Google Scholar] [CrossRef] [PubMed]
  50. Li, G.; Veenstra, A.A.; Talahalli, R.R.; Wang, X.; Gubitosi-Klug, R.A.; Sheibani, N.; Kern, T.S. Marrow-Derived Cells Regulate the Development of Early Diabetic Retinopathy and Tactile Allodynia in Mice. Diabetes 2012, 61, 3294–3303. [Google Scholar] [CrossRef]
  51. Gubitosi-Klug, R.A.; Talahalli, R.; Du, Y.; Nadler, J.L.; Kern, T.S. 5-Lipoxygenase, but Not 12/15-Lipoxygenase, Contributes to Degeneration of Retinal Capillaries in a Mouse Model of Diabetic Retinopathy. Diabetes 2008, 57, 1387–1393. [Google Scholar] [CrossRef]
  52. Gastinger, M.J.; Singh, R.S.J.; Barber, A.J. Loss of Cholinergic and Dopaminergic Amacrine Cells in Streptozotocin-Diabetic Rat and Ins2Akita-Diabetic Mouse Retinas. Investig. Opthalmol. Vis. Sci. 2006, 47, 3143–3150. [Google Scholar] [CrossRef]
  53. Gastinger, M.J.; Kunselman, A.R.; Conboy, E.E.; Bronson, S.K.; Barber, A.J. Dendrite Remodeling and Other Abnormalities in the Retinal Ganglion Cells of Ins2AkitaDiabetic Mice. Investig. Opthalmol. Vis. Sci. 2008, 49, 2635–2642. [Google Scholar] [CrossRef]
  54. Shaw, S.G.; Boden, J.P.; Biecker, E.; Reichen, J.; Rothen, B. Endothelin antagonism prevents diabetic retinopathy in NOD mice: A potential role of the angiogenic factor adrenomedullin. Exp. Biol. Med. 2006, 231, 1101–1105. [Google Scholar]
  55. Liu, Q.; Zhang, F.; Zhang, X.; Cheng, R.; Ma, J.-X.; Yi, J.; Li, J. Fenofibrate ameliorates diabetic retinopathy by modulating Nrf2 signaling and NLRP3 inflammasome activation. Mol. Cell. Biochem. 2017, 445, 105–115. [Google Scholar] [CrossRef]
  56. Hernández, C.; Bogdanov, P.; Corraliza, L.; García-Ramírez, M.; Solà-Adell, C.; Arranz, J.A.; Arroba, A.I.; Valverde, A.M.; Simó, R. Topical Administration of GLP-1 Receptor Agonists Prevents Retinal Neurodegeneration in Experimental Diabetes. Diabetes 2015, 65, 172–187. [Google Scholar] [CrossRef] [PubMed]
  57. Byrne, E.M.; Llorián-Salvador, M.; Lyons, T.J.; Chen, M.; Xu, H. Tofacitinib Ameliorates Retinal Vascular Leakage in a Murine Model of Diabetic Retinopathy with Type 2 Diabetes. Int. J. Mol. Sci. 2021, 22, 11876. [Google Scholar] [CrossRef] [PubMed]
  58. Sampedro, J.; Bogdanov, P.; Ramos, H.; Solà-Adell, C.; Turch, M.; Valeri, M.; Simó-Servat, O.; Lagunas, C.; Simó, R.; Hernández, C. New Insights into the Mechanisms of Action of Topical Administration of GLP-1 in an Experimental Model of Diabetic Retinopathy. J. Clin. Med. 2019, 8, 339. [Google Scholar] [CrossRef] [PubMed]
  59. Samuels, I.S.; Bell, B.A.; Pereira, A.J.; Saxon, J.E.; Peachey, N.S. Early retinal pigment epithelium dysfunction is concomitant with hyperglycemia in mouse models of type 1 and type 2 diabetes. J. Neurophysiol. 2015, 113, 1085–1099. [Google Scholar] [CrossRef] [PubMed]
  60. Bogdanov, P.; Corraliza, L.; Villena, J.A.; Carvalho, A.R.; Garcia-Arumí, J.; Ramos, D.; Ruberte, J.; Simó, R.; Hernández, C. The db/db Mouse: A Useful Model for the Study of Diabetic Retinal Neurodegeneration. PLoS ONE 2014, 9, e97302. [Google Scholar] [CrossRef] [PubMed]
  61. Ly, A.; Scheerer, M.F.; Zukunft, S.; Muschet, C.; Merl, J.; Adamski, J.; de Angelis, M.H.; Neschen, S.; Hauck, S.M.; Ueffing, M. Retinal proteome alterations in a mouse model of type 2 diabetes. Diabetologia 2013, 57, 192–203. [Google Scholar] [CrossRef] [PubMed]
  62. Tadayoni, R.; Paques, M.; Gaudric, A.; Vicaut, E. Erythrocyte and leukocyte dynamics in the retinal capillaries of diabetic mice. Exp. Eye Res. 2003, 77, 497–504. [Google Scholar] [CrossRef] [PubMed]
  63. Jung, E.; Kim, J.; Kim, C.-S.; Kim, S.-H.; Cho, M.-H. Gemigliptin, a dipeptidyl peptidase-4 inhibitor, inhibits retinal pericyte injury in db/db mice and retinal neovascularization in mice with ischemic retinopathy. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2015, 1852, 2618–2629. [Google Scholar] [CrossRef] [PubMed]
  64. Tang, L.; Zhang, Y.; Jiang, Y.; Willard, L.; Ortiz, E.; Wark, L.; Medeiros, D.; Lin, D. Dietary wolfberry ameliorates retinal structure abnormalities in db/db mice at the early stage of diabetes. Exp. Biol. Med. 2011, 236, 1051–1063. [Google Scholar] [CrossRef] [PubMed]
  65. Yang, Q.; Xu, Y.; Xie, P.; Cheng, H.; Song, Q.; Su, T.; Yuan, S.; Liu, Q. Retinal Neurodegeneration in db/db Mice at the Early Period of Diabetes. J. Ophthalmol. 2015, 2015, 1–9. [Google Scholar] [CrossRef] [PubMed]
  66. Liu, M.; Pan, Q.; Chen, Y.; Yang, X.; Zhao, B.; Jia, L.; Zhu, Y.; Zhang, B.; Gao, X.; Li, X.; et al. Administration of Danhong Injection to diabetic db/db mice inhibits the development of diabetic retinopathy and nephropathy. Sci. Rep. 2015, 5, 11219. [Google Scholar] [CrossRef] [PubMed]
  67. Clements, R.S.; Robison, W.; Cohen, M.P. Anti-Glycated Albumin Therapy Ameliorates Early Retinal Microvascular Pathology in db/db Mice. J. Diabetes Its Complicat. 1998, 12, 28–33. [Google Scholar] [CrossRef] [PubMed]
  68. Barile, G.R.; Pachydaki, S.I.; Tari, S.R.; Lee, S.E.; Donmoyer, C.M.; Ma, W.; Rong, L.L.; Buciarelli, L.G.; Wendt, T.; Ho¨rig, H.; et al. The RAGE Axis in Early Diabetic Retinopathy. Investig. Opthalmol. Vis. Sci. 2005, 46, 2916–2924. [Google Scholar] [CrossRef]
  69. Hinder, L.M.; Sas, K.M.; O’brien, P.D.; Backus, C.; Kayampilly, P.; Hayes, J.M.; Lin, C.-M.; Zhang, H.; Shanmugam, S.; Rumora, A.E.; et al. Mitochondrial uncoupling has no effect on microvascular complications in type 2 diabetes. Sci. Rep. 2019, 9, 1–11. [Google Scholar] [CrossRef]
  70. Hammer, S.S.; Beli, E.; Kady, N.; Wang, Q.; Wood, K.; Lydic, T.A.; Malek, G.; Saban, D.R.; Wang, X.X.; Hazra, S.; et al. The Mechanism of Diabetic Retinopathy Pathogenesis Unifying Key Lipid Regulators, Sirtuin 1 and Liver X Receptor. EBioMedicine 2017, 22, 181–190. [Google Scholar] [CrossRef]
  71. Beli, E.; Yan, Y.; Moldovan, L.; Vieira, C.P.; Gao, R.; Duan, Y.; Prasad, R.; Bhatwadekar, A.; White, F.A.; Townsend, S.D.; et al. Restructuring of the Gut Microbiome by Intermittent Fasting Prevents Retinopathy and Prolongs Survival in db/db Mice. Diabetes 2018, 67, 1867–1879. [Google Scholar] [CrossRef]
  72. Kern, T.S.; Engerman, R.L. A Mouse Model of Diabetic Retinopathy. Arch. Ophthalmol. 1996, 114, 986–990. [Google Scholar] [CrossRef]
  73. Cheung, A.K.; Fung, M.K.; Lo, A.C.; Lam, T.T.; So, K.F.; Chung, S.S.; Chung, S.K. Aldose Reductase Deficiency Prevents Diabetes-Induced Blood-Retinal Barrier Breakdown, Apoptosis, and Glial Reactivation in the Retina of db/db Mice. Diabetes 2005, 54, 3119–3125. [Google Scholar] [CrossRef]
  74. Carmo, A.D.; Ramos, P.; Reis, A.; Proença, R.; Cunha-Vaz, J. Breakdown of the Inner and Outer Blood Retinal Barrier in Streptozotocin-Induced Diabetes. Exp. Eye Res. 1998, 67, 569–575. [Google Scholar] [CrossRef] [PubMed]
  75. Miyamoto, K.; Khosrof, S.; Bursell, S.-E.; Rohan, R.; Murata, T.; Clermont, A.C.; Aiello, L.P.; Ogura, Y.; Adamis, A.P. Prevention of leukostasis and vascular leakage in streptozotocin-induced diabetic retinopathy via intercellular adhesion molecule-1 inhibition. Proc. Natl. Acad. Sci. USA 1999, 96, 10836–10841. [Google Scholar] [CrossRef] [PubMed]
  76. Park, J.-W.; Park, S.-J.; Kim, K.-Y.; Chung, J.-W.; Chun, M.-H.; Oh, S.-J. Apoptotic death of photoreceptors in the streptozotocin-induced diabetic rat retina. Diabetologia 2003, 46, 1260–1268. [Google Scholar] [CrossRef]
  77. Ishida, S.; Usui, T.; Yamashiro, K.; Kaji, Y.; Ahmed, E.; Carrasquillo, K.G.; Amano, S.; Hida, T.; Oguchi, Y.; Adamis, A.P. VEGF164Is Proinflammatory in the Diabetic Retina. Investig. Opthalmol. Vis. Sci. 2003, 44, 2155–2162. [Google Scholar] [CrossRef] [PubMed]
  78. Rungger-Brändle, E.; A Dosso, A.; Leuenberger, P.M. Glial reactivity, an early feature of diabetic retinopathy. Investig. Ophthalmol. Vis. Sci. 2000, 41, 1971–1980. [Google Scholar]
  79. Barber, A.J.; Lieth, E.; A Khin, S.; A Antonetti, D.; Buchanan, A.G.; Gardner, T.W. Neural apoptosis in the retina during experimental and human diabetes. Early onset and effect of insulin. J. Clin. Investig. 1998, 102, 783–791. [Google Scholar] [CrossRef]
  80. Aizu, Y.; Oyanagi, K.; Hu, J.; Nakagawa, H. Degeneration of retinal neuronal processes and pigment epithelium in the early stage of the streptozotocin-diabetic rats. Neuropathology 2002, 22, 161–170. [Google Scholar] [CrossRef]
  81. Barber, A.J.; A Antonetti, D.; Gardner, T.W. Altered expression of retinal occludin and glial fibrillary acidic protein in experimental diabetes. The Penn State Retina Research Group. Investig. Ophthalmol. Vis. Sci. 2000, 41, 3561–3568. [Google Scholar]
  82. Fico, E.; Rosso, P.; Triaca, V.; Segatto, M.; Lambiase, A.; Tirassa, P. NGF Prevents Loss of TrkA/VEGFR2 Cells, and VEGF Isoform Dysregulation in the Retina of Adult Diabetic Rats. Cells 2022, 11, 3246. [Google Scholar] [CrossRef]
  83. Gong, C.-Y.; Lu, B.; Hu, Q.-W.; Ji, L.-L. Streptozotocin induced diabetic retinopathy in rat and the expression of vascular endothelial growth factor and its receptor. Int. J. Ophthalmol. 2013, 6, 573–577. [Google Scholar] [CrossRef] [PubMed]
  84. Lieth, E.; Barber, A.J.; Xu, B.; Dice, C.; Ratz, M.J.; Tanase, D.; Strother, J.M. Glial reactivity and impaired glutamate metabolism in short-term experimental diabetic retinopathy. Penn State Retina Research Group. Diabetes 1998, 47, 815–820. [Google Scholar] [CrossRef]
  85. Agardh, E.; Bruun, A.; Agardh, C.-D. Retinal glial cell immunoreactivity and neuronal cell changes in rats with STZ-induced diabetes. Curr. Eye Res. 2001, 23, 276–284. [Google Scholar] [CrossRef]
  86. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef] [PubMed]
  87. Ferrington, D.A.; Fisher, C.R.; Kowluru, R.A. Mitochondrial Defects Drive Degenerative Retinal Diseases. Trends Mol. Med. 2019, 26, 105–118. [Google Scholar] [CrossRef]
  88. Kowluru, R.A.; Kowluru, A.; Mishra, M.; Kumar, B. Oxidative stress and epigenetic modifications in the pathogenesis of diabetic retinopathy. Prog. Retin. Eye Res. 2015, 48, 40–61. [Google Scholar] [CrossRef] [PubMed]
  89. Gonzalez-Fernandez, F.; Sung, D.; Haswell, K.M.; Tsin, A.; Ghosh, D. Thiol-dependent antioxidant activity of interphotoreceptor retinoid-binding protein. Exp. Eye Res. 2014, 120, 167–174. [Google Scholar] [CrossRef]
  90. Chen, J.; Shao, Y.; Sasore, T.; Moiseyev, G.; Zhou, K.; Ma, X.; Du, Y.; Ma, J.-X. Interphotoreceptor Retinol-Binding Protein Ameliorates Diabetes-Induced Retinal Dysfunction and Neurodegeneration Through Rhodopsin. Diabetes 2020, 70, 788–799. [Google Scholar] [CrossRef]
  91. Lee, M.; Li, S.; Sato, K.; Jin, M. Interphotoreceptor Retinoid-Binding Protein Mitigates Cellular Oxidative Stress and Mitochondrial Dysfunction Induced by All-trans-Retinal. Investig. Opthalmol. Vis. Sci. 2016, 57, 1553–1562. [Google Scholar] [CrossRef] [PubMed]
  92. Fickweiler, W.; Park, H.; Park, K.; Mitzner, M.G.; Chokshi, T.; Boumenna, T.; Gautier, J.; Zaitsu, Y.; Wu, I.-H.; Cavallerano, J.; et al. Elevated Retinol Binding Protein 3 Concentrations Are Associated With Decreased Vitreous Inflammatory Cytokines, VEGF, and Progression of Diabetic Retinopathy. Diabetes Care 2022, 45, 2159–2162. [Google Scholar] [CrossRef] [PubMed]
  93. Yokomizo, H.; Maeda, Y.; Park, K.; Clermont, A.C.; Hernandez, S.L.; Fickweiler, W.; Li, Q.; Wang, C.-H.; Paniagua, S.M.; Simao, F.; et al. Retinol binding protein 3 is increased in the retina of patients with diabetes resistant to diabetic retinopathy. Sci. Transl. Med. 2019, 11, eaau6627. [Google Scholar] [CrossRef] [PubMed]
  94. Lundeen, E.A.; Burke-Conte, Z.; Rein, D.B.; Wittenborn, J.S.; Saaddine, J.; Lee, A.Y.; Flaxman, A.D. Prevalence of Diabetic Retinopathy in the US in 2021. JAMA Ophthalmol. 2023, 141, 747–754. [Google Scholar] [CrossRef]
Figure 2. Mitophagy is an essential component of resilience. The onset of DM, i.e., enduring elevation of blood sugar, triggers mitochondrial adaptation, which includes enhanced clearance of dysfunctional mitochondria. Deterioration of this innate defense mechanism results in self-perpetuating progress damage of the retina, which manifests as DR.
Figure 2. Mitophagy is an essential component of resilience. The onset of DM, i.e., enduring elevation of blood sugar, triggers mitochondrial adaptation, which includes enhanced clearance of dysfunctional mitochondria. Deterioration of this innate defense mechanism results in self-perpetuating progress damage of the retina, which manifests as DR.
Ijms 25 01610 g002
Table 3. Articles of note discussing Type-1 diabetic rat models.
Table 3. Articles of note discussing Type-1 diabetic rat models.
Authors, (Year)ModelCollected TissueMechanism of Analysis in Diabetic RetinopathyMonitoring of DR ProgressionEarliest Detection of Diabetic RetinopathyResult/Outcome
Jones, C.W., Cunha-Vaz, J.G., Rusin, M.M. (1982) [14]STZ
Alloxan
RetinaVitreous fluorophotometryNone2 days of diabetesStreptozocin- or alloxan-induced diabetic rat retina showed increases in vitreal fluorescein levels compared to control rats.
Miyamoto, K., Khosrof, S., Bursell, S.E., et al. (1999) [75]STZRetinaAOLF and Fluorescein Angiography
Isotope Dilution Technique
Ribonuclease protection assay
Yes1 week of diabetesRat retina showed increased albumin permeation compared to controls.
Park, S.H., Park, J.W., Park, S.J., et al. (2003) [76]STZRetinaImmunohistochemistry
Histologic analysis
Electron microscopy
TUNEL
Yes1 week of diabetesRat retina showed myelinated and multivesicular features of mitochondria, increased thickness of inner retina, and the vitreal radial processes of Müller cells stained moderately and became prominent compared to controls.
Ishida, S., Usui, T., Yamashiro, K., et al. (2003) [77]STZRetinaFITC
FITC-dextran
ELISA
Immunohistochemistry
Yes2 weeks of diabetesRat retina showed increased leukostasis and blood-retinal barrier (BRB) breakdown compared to controls.
Rungger-Brändle, E., Dosso, A.A., Leuenberger, P.M. (2000) [78]STZRetinaImmunofluorescence
Confocal microscopy
Western blot
Evans Blue staining
Yes2 weeks of diabetesRat retina showed leakage of BRB compared to controls.
Barber, A.J., Lieth, E., Khin, S.A., et al. (1998) [79]STZRetinaTUNEL
Nissl staining
Microscopy
Yes4 weeks of diabetesRat retina showed increased TUNEL-positive cells, suggesting retinal cell apoptosis compared to controls.
Aizu, Y., Oyanagi, K., Hu, J., et al. (2002) [80]STZRetinaERG
Electron microscopy
Optical microscopy
Yes4 weeks of diabetesRat retina showed reduced thickness of inner plexiform layer (IPL, and photoreceptor segment layer (PSL), reduced a- and b-waves, oscillatory potentials, and deepened hollows in basal infoldings of retinal pigment epithelium (RPE) compared to controls.
Barber, A.J., Antonetti, D.A., Gardner, T.W. (2000) [81]STZRetinaImmunofluorescence
Histochemistry
Yes8 weeks of diabetesRat retina showed astrocytes with reduced GFAP intensity, multiple processes, enlarged cell bodies, and hypertrophied compared to controls. They also demonstrated regions of heavily punctate immunoreactivity and absence of distinct junctional distribution in major arterioles compared to controls.
Fico, E., Rosso, P., Triaca, V., et al. (2022) [82]STZRetinaMicroscopy
Immunofluorescence histology
Western blot
Confocal imaging
None8 weeks of diabetesRat retina showed fragmentation of retinal vessels, high expression Rock1 level, proNGF and its glycosylated form, decreased pVEGFR2 expression, and Akt levels compared to controls.
Gong, C.Y., Lu, B., Hu, Q.W., et al. (2013) [83]STZRetinaImmunofluorescence
RT-PCR
Yes12 weeks of diabetesRat retina showed morphological changes in INL, ONL, increased number of new vessels, mRNA expression of VEGF, VEGFR1, and VEGFR2 compared to controls.
Lieth, E., Barber, A.J., Xu, B., et al. (1998) [84]STZRetinaELISA
Immunohistochemistry
Immunoblotting
glutamate-to-glutamate conversion
Yes12 weeks of diabetesRat retina showed increased GFAP IR and increased glutamate levels compared to controls.
Agardh, E., Bruun, A., Agardh, C.D. (2001) [85]STZRetinaImmunohistochemistryYes24 weeks of diabetesRat retina showed increased GFAP and glial cell immunoreactivity, and horizontal cell numbers with decreased branching of terminals compared to controls.
Once outcomes such as visual acuity decreases, reduced contrast sensitivity, ERG changes, NFL-GCL thinning, RGC and amacrine cell neurodegeneration, and gliosis have developed, they persist and intensify with the duration of DM, with the exception of leukostasis and inflammation [17]. Although a variety of markers have been used throughout the literature to study the role of inflammation in DR, some of the more common markers included the elevated number of leukocytes, leukocyte adhesion, microglial infiltration, production of cytokines, eNOS, and several caspases [20,32,33,38,43,44,45]. Every murine model of DR shows the progression of the disease, i.e., the extent and nature of the damage of the retina increases, but not the transition from NPDR to PDR.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cubillos, S.; Kazlauskas, A. Manifestation of Pathology in Animal Models of Diabetic Retinopathy Is Delayed from the Onset of Diabetes. Int. J. Mol. Sci. 2024, 25, 1610. https://doi.org/10.3390/ijms25031610

AMA Style

Cubillos S, Kazlauskas A. Manifestation of Pathology in Animal Models of Diabetic Retinopathy Is Delayed from the Onset of Diabetes. International Journal of Molecular Sciences. 2024; 25(3):1610. https://doi.org/10.3390/ijms25031610

Chicago/Turabian Style

Cubillos, Samuel, and Andrius Kazlauskas. 2024. "Manifestation of Pathology in Animal Models of Diabetic Retinopathy Is Delayed from the Onset of Diabetes" International Journal of Molecular Sciences 25, no. 3: 1610. https://doi.org/10.3390/ijms25031610

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop