Open access peer-reviewed chapter

Animal Models of Diabetic Retinopathy (Part 2)

Written By

Larissa H.C. Tang, Ian Y.H. Wong and Amy C.Y. Lo

Reviewed: 16 August 2017 Published: 20 December 2017

DOI: 10.5772/intechopen.70589

From the Edited Volume

Experimental Animal Models of Human Diseases - An Effective Therapeutic Strategy

Edited by Ibeh Bartholomew

Chapter metrics overview

1,407 Chapter Downloads

View Full Metrics

Abstract

Diabetic retinopathy (DR) is one of the leading causes of preventable vision impairment and blindness in the working-age population worldwide. Numerous animal models have been developed for therapeutic drug screening and to further increase our understanding of the molecular and cellular pathological processes involved in DR. Following our discussion of mouse models in “Animal Models of Diabetic Retinopathy Part 1,” we describe the cellular, molecular, and morphological features of both rodent and nonrodent models of DR and their respective advantages and limitations in this chapter. To date, no animal model can holistically reproduce the pathological progression of human DR; most only display early or advanced lesions of DR. However, a thorough understanding of genotypic and phenotypic expressions of existing models will facilitate researchers’ selection of the appropriate model to simulate their desired clinical scenarios.

Keywords

  • animals
  • blood glucose
  • blindness
  • diabetic complications
  • diabetes mellitus/pathology/physiopathology
  • neovascularization
  • proliferative
  • retinal vessels

1. DR features of animal models

Among all the existing animal models of diabetic retinopathy (DR), mice and rats are most commonly used, possibly due to their small size, availability, genetic tractability, and relatively faster development of DR lesions as compared with larger animals. Tables 13 summarize the cellular, molecular, and morphological features of rat and nonrodent models of DR. Features of mouse models are detailed in the previous chapter (Animal Models of Diabetic Retinopathy Part 1).

Rat modelType of diabetesHyperglycemia onsetCellular, morphological, and vascular features of human DR displayed in rat models
(Age at which correlates are first reported unless otherwise specified)
(+Time post treatment: diabetes, galactosemia, or induction of VEGF overexpression)
NPDR featuresPDR featuresFunctional changes (ERG)
STZ injection1Within one wk• 1 wk+: reduced total retinal thickness [93]
• 2 wks+: increased retinal apoptosis [94]
  BRB breakdown [5, 93] (16 wks+ [6])
• 4 wks+: Müller cell gliosis [2, 3] (6 wks+ [4]) (12 wks+ [5])
  Reduced NeuN-positive cells in GCL [3] (4 mo reduced number of cells in GCL [1])
  Reduced astrocyte number [5] (6 wks+ [2])
  Increased number of microglia [5] (4 mo+ [3])
  Reduced number of cells in ONL [93]
• 6 wks+: reduced astrocyte processes [2]
• 8 wks+: increased adherent leukocytes [7]
• 16 wks+: increased acellular capillaries [6] (8 mo+ [1])
  Decreased number of pericytes [6]
• 4 mo+: reduced NeuN-positive cells in INL [3]
• 8 mo+: pericyte ghosts [1]
• 12 mo+: capillary BM thickening [8]
• 2 wks+: decreased b-wave amplitude [4] (11 wks+ [9]) (16 wks+ [6])
  Decreased OP amplitude [4]   (16 wks+ [6])
• 8 wks+: decreased OPs (not observed at 11 wks) [95]
• 11 wks+: increased a-wave implicit time [9]
  Decreased a-wave amplitude [9]   (10 wks+ [4])
  Delayed OPs [9]
Alloxan injection1• 6 wks+: BRB breakdown [96]
• 5 mo+: capillary occlusion with endothelial cell swelling [11]
• 8 mo+: retinal microvascular cell death [10]
• 9 mo+: IRMAs in lower capillary layer [11]
• 12 mo+: pericyte ghosts [12]
   Increased no. of acellular capillaries [12] (18 mo+ [10])
  Capillary BM thickening [12]
• 2 mo+: neovascularization in mid-periphery region [11]
• 5 mo+: neovascularization in center and mid-periphery regions [11]
• 9 mo+: neovascularization in all regions [11]
Galactose-fed//• 4 mo+: increased retinal microvascular cell apoptosis^ [10]
• 12 mo+: pericyte ghosts [12] (23 mo+)*^ [13]
  Increased no. of acellular capillaries [12] (23 mo+)*^ [13]
  Capillary BM thickening [12] (23 mo+)*^ [13] (28 mo+)^ [14]
• 24 mo+: dilated, hypercellular vessels*^ [13]
  IRMAs* [13]
• 28 mo+: disrupted retinal layers^ [14]
  Gliosis^ [14]
  Capillary dilatation^ [14]
  Microaneurysms (OPL, INL)^ [14]
  Increased number of endothelial cells^ [14]
*50% galactose diet; ^30% galactose diet
BB rat160–120 days (90% hyper-glycemic by 90–120 days) [15]• 6 mo: increased retinal capillary BM thickness [1518]
• 7.7 ± 1.1 mo: reduced and deranged basal infoldings of the RPE basal plasmalemma [16]
• 8 mo+: decreased pericyte/endothelial cell ratio [17]
  Decreased number of pericytes [17]
  Microinfarctions with nonperfused areas [19]
WBN/ Kob rat29–12 months of age [20]• 5 mo: decreased outer segment thickness [21]
• 10 mo: clustering of capillaries into tortuous knots [22]
• 11 mo: decreased OPL and ONL thickness [21]
• 14 mo: capillary BM thickening [21]
  Complete photoreceptor cell nuclei and ONL loss ([21, 22], 169)
• 15 mo: decreased number of capillaries [22]
  Capillary loop formation [22]
• 19 mo: acellular capillaries [24]
24 mo:
 Intravitreal and choroidal neovascularization [23, 24]
 Increased hyalinization of intraretinal vessels [23]
 Increased intravitreal proliferation of fibrovascular elements [23]
ZDF rat26–7 wks of age [25]• 14 wks: increased VEGF-beta mRNA levels in retina [26]
• 27 wks: capillary BM thickening [28] (6–7 mo) [27]
  Increased capillary hypercellularity [28] (6–7 mo) [27]
• 33–34 wks: increased pericyte apoptosis and presence of pericyte ghosts [29]
  Increased endothelial cell apopotosis [29]
  Increased number of acellular capillaries [29]
OLEFT218 wks of age [30]• 24 wks: increased leukocyte entrapment [38]
• 28 wks: reduced total retinal and RNFL thickness [32]
• 36 wks: reduced number of RGCs [32]
  Increased apoptosis in RNFL [32]
• 40 wks: increased GFAP immunoreactivity in Müller cells [33]
• 14 mo: capillary BM thickening [31, 34]
  Endothelial cell degeneration [31, 34]
  Reduced pericyte area to total capillary cross-sectional area ratio [34]
  Caliber irregularity [34] (64 wks) [35]
  Capillary tortuosity [34] (64 wks) [35] (17 mo) [31]
  Capillary loop formation [34] (64 wks) [35] (17 mo) [31]
• 64 wks: capillary narrowing [35]
  Microaneurysms [35] (17 mo) [31]
• 19 mo: decreased INL and photoreceptor thickness [31]
  Decreased RPE height [31]
  Poorly developed basal infoldings [31]
60 wks:
 Prolonged peak latencies of OPs [36]
GK rat2 (non-obese)4–6 wks of age [37] (glucose in-tolerance as early as at 2 wks of age)• 1 mo+: increased retinal mean circulation time [38]
• 3 mo+: increased BRB permeability [39]
• 28 wks: increased ocular VEGF concentrations [41]
• 8 mo: Increased endothelial/pericyte ratio [40]
• 12 mo+: Activated microglia [42]
1 mo (post hyperglycemia)
• 1 mo+: reduced retinal segmental blood flow [38]
• 12 mo: migration and accumulation of microglia/macrophages in the subretinal space [42]
SDT2 (non-obese)25 wks of age (male rats) (100% diabetic by 40 wks of age) [44]• 40 wks: increased cellular apoptosis in GCL and INL [45]
  Increased GFAP immunoreactivity expression across the whole retina [45]
• 44 wks: increased retinal leukostasis [46]
• 52–60 wks: increased INL, ONL, PL, choroidal and total retinal thickness [47]
• 68 wks: fluorescein leakage around the optic disc [48]
  Venous dilation [48]
  Distorted retina [48] (70 wks) [44]
  Protrusion of optic disc into the vitreous
• 70 wks: retinal thickening [44]
• 77 wks: anterior chamber hemorrhage with fibrovascular changes around the iris [44, 49]
• 49–82 wks: acellular capillaries [49]
  Pericyte loss [49]
  Tortuous vessels [49]
  Capillary nonperfusion [49]
• 20 mo: increased VEGF immunoreactivity in vascular endothelial cells, GCL, INL, border between ONL and OS [50]
• 70 wks: tractional retinal detachment with fibrous proliferation [44, 49]
• 20 mo: neovascularization (53%) without ischemia [50]
 Tractional changes without retinal detachment [50]
32 weeks
 Prolonged OP peak latencies [48]
44 weeks
 Reduced a-, b-wave and OP1, OP2, OP3 amplitudes [51]
 Increased OP2, OP3, OP4 implicit time [51]
TetO rat [52]27–8 days after incorporation of doxy-cycline in drinking water4–5 weeks+ [52]
 RGC loss
 Reduced number of pericytes
 Increased number of stenotic vessels
 Increased number of veins with abnormal vessel caliber (deep microvascular plexus)
 BRB breakdown
 Increased GFAP expression
 Reactive gliosis
 Increased inflammatory markers
P18
 Intravitreal neovascularization in peripheral retina [54, 58]
ERG Changes 4–5 weeks after hyperglycemia onset
 Reduced a-waves, b-waves, and c-wave amplitude
 Elevated b/a-wave ratio
OIR//• P13: perivascular astrocyte loss [54]
• P14–16: increased vascular permeability (ceased at p18) [55]
• P18: reduced peripheral INL(and central), IPL and total retinal thickness [56]
 Increased Müller cell gliosis in peripheral retina [54, 56]
  Increased tortuosity of arterioles [57]
 Decreased vessel profiles in outer and inner plexuses in peripheral retina [54, 58]
 Altered pericyte-endothelial interactions [54]
 Shortened retinal outer segment length [97]
ERG changes at p18
 Reduced a-wave and b-wave amplitude [57]

Table 1.

Summary of the cellular, molecular, and morphological features displayed in rat models of DR.

This table has been modified from a review by Lai and Lo [19].

ModelType of diabetesHyperglycemia onsetCellular, morphological, and vascular features of human DR displayed in non-rodent models
(Age at which correlates are first reported unless otherwise specified)
(+Time posttreatment: diabetes, galactosemia, or induction of VEGF overexpression)
NPDR featuresPDR featuresFunctional changes (ERG)
DogGalactose-fed//• 19 mo+: pericyte loss [62] (36 mo+: increased endothelial/pericyte ratio [63])
• 24 mo+: uneven distribution of endothelial cells [62]
  Acellular capillaries [98] (36 mo+ [63])
  Endothelial proliferation (in areas with pericyte loss) [62] (36 mo+ [63])
• 27 mo+: microaneurysm [62] (32–36 mo+ [64]) (36 mo+ [63])
• 33 mo+: dot and blot hemorrhages [62, 64]
  Capillary varicose enlargement [62]
  Areas of nonperfusion [99] (36 mo+ [65] (56 mo+ [62])
• 36–48 mo+: hemorrhage [62]
• 56 mo+: preretinal hemorrhage [62]
  IRMAs [62]
• 60 mo+: AV shunts [62]
  Arterial node formation [62]
  Gliosis [62]
• 60 mo+: neovascularization [65]
• 84 mo+: intravitreal retinal vascular growth [62]
Partial detachment of posterior vitreous [62]
STZ injection [60]• 1–3 yrs.+: capillary BM thickening
• 4 yrs.+: pericyte loss
  Smooth muscle cell loss
• 7 yrs.+: microaneurysms
  Acellular capillaries
  IRMAs
Alloxan injection• 5 yrs.+: microaneurysm [61]
  Pericyte ghosts [61]
  Increased endothelial/pericyte ratio [61]
  Increased acellular capillaries [61]
  BM thickening [61]
  Vascular leakage, increased permeability of retinal blood vessels [100]
CatPancreatotomy11–2 weeks post surgery• 3–10 mo+: capillary BM thickening [70]
• 5 yrs.+: microaneurysm [69] (7.7–9.4 yrs.+ [71])
• 6.5 yrs.+: small intraretinal hemorrhages [69] (7.7–9.4 yrs.+ [71])
• 7.5 yrs.+: IRMAs [69]
  Capillary non-perfusion areas [69] (9.4 yrs.+ [71])
• 8.5 yrs.+: microvessel dilation and tortuosity
• 8.5 yrs.+: small foci of neovascularization9.4 yrs.+: decreased b-wave (trend) [71]
RabbitSTZ injection1135 days+:
 10% eyes: “Moderate vasculopathy with hard or soft exudates, widespread hemorrhages” [75]
 40% eyes: “Serious vasculopathy with serious retinal and preretinal hemorrhages, vascular lesions, hemovitreous, venous thrombosis” [75]
• 135 days+: 50% of eyes: proliferative retinopathy [75]
Diet-induced [72]212–24 weeks• 12 wks+: increased number of microaneurysms
VEGF implant//• 7 days+: increased dilation and tortuosity of vessels [73]
• 14 days+: vascular leakage [73]
• 14 days+: neovascularization (decreased after 21 days with almost complete regression by 35 days) [73]
VEGF + bFGF
• 48 hrs+: grade + 1 neovascularization [74]
• 4 days+: grade 4 neovascularization [74]
• 2 wks+: total traction retinal detachment with significant hemorrhage [74]
SwineSTZ injection11 wk [79]• 18 wks+: capillary BM thickening with rarefaction [79]
Alloxan injection115–30 days [76]• 90 days+: increased GFAP in Müller cells [76]
• 20 wks+: capillary BM thickening [77]
• 20 wks (age): pericyte loss [78]
  Reduced total number of BRB capillaries [78]
  Capillary collapse [78]
Zebra fishGlucose-induced11–2 days [90]• Day 28: decreased IPL and INL [90]
Glucose-induced (flk:EGFP transgenic zebrafish)• 6 dpf (3 days of glucose tx): increased diameter of hyaloid-retinal vessels [89]
Hypoxia-induced//• 3 days+: increased capillary sprouts in arterioles and veins [91]
  Increased number of branch points [91]
  Reduced intercapillary distance [91]
3 days+: neovascularization (increased vascular area) (plateau at day 12) [91]
vhl mutants//• 5.75 dpf: increased hyaloid and choroidal vascular networks [101]
• 6 dpf: vascular leakage
dpf = days post fertilization
7.5 dpf:
 Severe macular edema [101]
 Retinal detachment [101]
 Intraretinal neovascularization (predominantly in IPL) [101]

Table 2.

Summary of the cellular, molecular, and morphological features displayed in nonrodent models of DR.

This table has been modified from a review by Lai and Lo [19].

ModelType of diabetesHyperglycemia onsetCellular, morphological, and vascular features of human DR displayed in non-human primate models
(Age at which correlates are first reported unless otherwise specified)
(+Time post treatment: diabetes, galactosemia, or VEGF overexpression–induction)
NPDR featuresPDR featuresFunctional changes (ERG)
MonkeySTZ injection, spontaneous or pancreatectomy [81]1/Changes observed in diabetic monkeys with spontaneous or pharmacologically-induced hypertension
• 6–15 yrs.+: focal intraretinal capillary leakage spots
  Capillary dilatation
  Capillary dropout
  Cotton wool spots
  Microaneurysms
  Arteriolar dilatation and occlusion
  Atrophic retinopathy
Spontaneous, obese2/• 3–8 yrs.+: cotton wool spots [82]
  Capillary dropout [82]
  Microaneurysms Kim, #413}
  IRMAs [82]
  Nonperfused areas [82]
• 8 yrs.+: RGC loss [83]
  Decreased ONL, IS, OS thickness [83]
• 3–8 yrs.+: cystoid macular edema [82]8 yrs.+:
 Reduced OPs [83]
 Reduced amplitudes in mfERG
 Delayed a-wave [83]
Intra-vitreal VEGF injection//• 2 days+: tortuous vessels [85]
  Vascular leakage [85]
• 3 days+: vessel dilatation [84]
• 18 days+: microaneurysms [84]
  Intraretinal hemorrhage [84]
  Venous beading [84]
• 30 days+: intraretinal hemorrhage and edema [84]
• 2 days+: grade 2 iris neovascularization [85]
• 8 days+: grade 3 iris neovascularization [85]
• 19 days+: neovascular glaucoma [85]
VEGF implant//• 3 wks+: vascular leakage (BRB breakdown) [73]
• 2–3 wks+: dilation of retinal vessels [73]
Note: vascular changes peaked at 3 wks
• 1 wk.+: iris neovascularization (regressed between wks 2–3)
MarmosetGalactose-fed (30%) [86]//• 2.5 yrs.+: microaneurysm
  Increased acellular capillaries
  Pericyte loss
  BM thickening
  BRB breakdown
 Macular edema and thickening

Table 3.

Summary of the cellular, molecular, and morphological features displayed in nonhuman primate models of DR.

This table has been modified from a review by Lai and Lo [19].

1.1. Rat models

1.1.1. Pharmacological

1.1.1.1. Streptozotocin (STZ) injection

Streptozotocin (STZ) injection diabetic rats reproduce early symptoms of DR, though prominent differences have been identified between rat strains possibly as a result of differing genetic susceptibility to DR development. A report comparing differences in the development of DR lesions in Sprague Dawley, Lewis, and Wistar rats revealed that neuronal loss in the ganglion cell layer (GCL) occurred only in Lewis rats, while both Lewis and Wistar rats showed degeneration of capillaries with pericyte ghosts after 8 months of hyperglycemia [1]. Sprague Dawley rats did not develop any lesions.

In general, neuronal and glial changes appear to precede vascular changes in this model. Retinal apoptosis and retinal thinning developed within 1–2 weeks of hyperglycemia. Müller cell gliosis, as evidenced by increased glial fibrillary acidic protein (GFAP) expression, was prominent beginning at 4 weeks of hyperglycemia [2, 3, 4, 5]. Studies have also documented a reduction in astrocyte number and processes [2, 5], decrease in the number of cells in the GCL [1, 3], and an increase in number of microglia [1, 3] after 4–6 weeks of hyperglycemia. Blood-retina barrier (BRB) breakdown was present after 2 weeks of hyperglycemia, while other vascular changes, including acellular capillaries [1, 6], leukostasis [7], and arterial and venous capillary basement membrane (BM) thickening [8], were only present after a longer period of hyperglycemia. Pericyte loss became evident after 4 [6] or 8 months [1] of hyperglycemia. Retinal dysfunction was evidenced by decreased b-wave [4, 6, 9] and OP amplitude [4, 6] and decreased a-wave amplitude [4, 9] with delayed oscillatory potentials (OP) [9] as early as at 2 weeks and 10 weeks, respectively.

1.1.1.2. Alloxan injection

Studies using alloxan injections for diabetes-induction are limited, and none have assessed for neurodegenerative lesions of DR. Alloxan-induced diabetic rats were found to have BRB breakdown within 6 weeks of hyperglycemia. With a longer duration of diabetes, there was retinal microvascular cell death [10], intraretinal microvascular abnormalities (IRMA) [11], pericyte ghosts [12], acellular capillaries [10, 12], and capillary BM thickening [12]. Neovascularization began in the mid-peripheral retina after 2 months of hyperglycemia and extended to all regions of the retina by 9 months [11].

1.1.2. Diet induced

Galactose-fed rats display many retinal microangiopathy features that resemble non-proliferative diabetic retinopathy (NPDR). Four months of 30% galactose-fed diet resulted in increased retinal microvascular cell apoptosis [10]. With a longer duration of galactosemia, pericyte ghosts, acellular capillaries, vessel dilation, capillary BM thickening, and IRMAs became evident in both 30 and 50% galactose-fed rats [12, 13, 14]. Long-term galactosemia of 28 months further resulted in gliosis, displacement or disruption of retinal layers, an increase in the number of endothelial cells, and microaneurysm formation in the outer plexiform layer (OPL) and inner nuclear layer (INL) [14].

1.1.3. Transgenic

1.1.3.1. Biobreeding (BB) rats

The diabetes-prone biobreeding (BB) rat is a T1D model derived from outbred Wistar rats. BB rats develop insulitis with selective β-cell destruction, hyperglycemia, hyperketonemia, ketonuria, and hypoinsulinemia. Rapid progression to fatal diabetic ketoacidosis results if exogenous insulin is not administered. The majority of rats develop hyperglycemia by the age of 90–120 days [15]. Existing reports have demonstrated reduced and deranged basal infoldings of the retinal pigment epithelium (RPE) basal plasmalemma [16], pericyte loss, reduced pericyte to endothelial cell ratios [17], capillary BM thickening [15, 18], and microinfarctions [19]. Microaneurysm formation and neovascularization were absent even after 11 months of diabetes [19]. Characterization studies of DR using this model remain limited. Subsequent inbreeding and outbreeding have produced various genetically distinct substrains (named after the origin of the colony), including the BBDP/Wor, BB/OK, and BB/Pfd rat. Care should be taken when comparing results of studies utilizing rats of different substrains due to potential genetic and phenotypic differences.

1.1.3.2. Wistar Bonn/Kobori (WBN/Kob) rats

The Wistar Bonn/Kobori (WBN/Kob) rat is a nonobese T2D strain of Wistar rats that develop exocrine pancreatic insufficiency characterized by α- and β-cell destruction. Male rats develop hyperglycemia at around 9–12 months of age, while females remain unaffected [20].

The retinal changes in this rat model have long been argued to be more degenerative than diabetic. At the age of 5 months, prior to the development of hyperglycemia, the rats showed signs of retinal degeneration with the loss of rods and cones and thinning of the photoreceptor layers [21]. Although retinal capillary changes, such as capillary BM thickening [21], reduced capillary number, formation of capillary loops, presence of acellular capillaries, and clustering of capillaries into tortuous knots [22], were observed after hyperglycemic development, it is uncertain whether these changes arose from the development of diabetes or resulted from further progression of retinal degeneration. WBN/Kob rats survive for a longer duration under diabetic conditions compared to other models. Perhaps, this prolonged survival and hyperglycemia duration had led to the development of intravitreal and choroidal neovascularization, which was accompanied by the hyalinization of intraretinal vessels and increased proliferation of fibrovascular elements in the vitreous at 24 months of age [23, 24]. Such features had not been previously reported in other rats. The proliferative changes were mostly absent in nondiabetic age-matched female counterparts [23], suggesting that the WBN/Kob rat may serve as a diabetic angiopathy model. Nevertheless, the retinal degenerative changes in the WBN/Kob rats are arguably different from that of human DR, making it a less suitable model for DR.

1.1.3.3. Zucker diabetic fatty (ZDF) rats

The Zucker diabetic fatty rat (ZDF/Gmi, fa/fa) is a T2D model derived from the partial inbreeding of the fa/fa line. The rats carry an inherited mutation in the leptin receptor and need to be maintained on a specialized diet to ensure consistent diabetes development. Male rats display marked hyperglycemia, hyperinsulinemia, insulin resistance, and obesity at 6–7 weeks of age [25]. Studies have reported increased vascular endothelial growth factor (VEGF) expression in the retina that correlated with retinal levels of inflammatory markers at 14 weeks of age [26], capillary BM thickening with increased retinal capillary hypercellularity at 27 weeks [27, 28], and increased pericyte apoptosis, endothelial apoptosis, presence of pericyte ghosts and number of acellular capillaries as compared to the lean controls by 33–34 weeks [29]. The rats have a relatively short lifespan of 1 year but do not require insulin treatment while exhibiting consistently marked hyperglycemia with glucose levels averaging at 500 mg/dL when fed with the appropriate diet. To date, few retinal morphological and therapeutic drug studies have been performed on this rat model.

1.1.3.4. Otsuka Long-Evans Tokushima fatty (OLETF) rats

The Otsuka Long-Evans Tokushima fatty (OLETF) rat is a T2D model originally generated from selective breeding of the Long-Evans rats that spontaneously exhibited polyuria, polydipsia, and obesity. The OLETF rat is characterized by mild obesity, a late onset of chronic hyperglycemia after the age of 18 weeks, male inheritance, pancreatic islet hyperplasia, and the development of T2D as a result of metabolic syndrome [30]. Significant weight gain occurs between the age of 1 and 6 months followed by significant weight loss after the age of 9 months [31].

The rats display an array of retinal cellular morphological changes corresponding to the early stage of DR. Increased leukocyte entrapment was first reported in 24-week-old rats. Spectral domain optical coherence tomography (SD-OCT) revealed significant thinning of the total retinal and retinal nerve fiber layer (RNFL) at the age of 28 weeks [32]. There was retinal ganglion cells (RGC) loss with nonuniform distribution of remaining RGCs and increased apoptosis in the RNFL at 36 weeks [32]. GFAP and VEGF immunoreactivity were upregulated, with VEGF expression extending across all layers of the retina into the OPL [33]. By 14 months of age, there were endothelial cell degeneration, capillary BM thickening, and pericyte loss [31, 34]. A longer duration of hyperglycemia resulted in the reduction in INL thickness, photoreceptor thickness, and RPE height with poorly developed basal infoldings, suggestive of neurodegeneration. Rats aged 14–17 months have consistently exhibited marked microvascular changes, including vessel tortuosity, caliber irregularity, capillary loop formation, capillary narrowing, and microaneurysms [31, 34, 35]. One study further revealed prolonged peak latencies of OPs on ERG, particularly OP1 and OP3, [36], although experimental conditions may have accounted for such abnormalities.

Existing characterization studies have only been performed by a selective number of research groups. Despite the similarities in the pathophysiology of T2D in OLETF rats and humans, the use of OLETF rats in DR studies is limited by the model’s late onset of hyperglycemia and absence of late-stage DR features. The presence of hemorrhages, exudates, and acellular capillaries has not been reported in rats up to 19 months of age [31], signifying its unsuitability as an angiopathic DR model.

1.1.3.5. Nonobese Goto-Kakizaki (GK) rats

The GK rat is a nonobese T2D model that develops glucose intolerance at 2 weeks of age and significant hyperglycemia by the age of 4–6 weeks [37]. The rats were generated from repeated selective inbreeding of normal Wistar rats based on their glucose intolerance indices [37]. After 1 month of hyperglycemia, the rats displayed significantly increased retinal mean circulatory time and reduced retinal segmental blood flow [38]. BRB permeability was increased after 3 months of hyperglycemia [39]. Eight-month-old rats presented with an increased endothelial/pericyte ratio that increased with the duration of hyperglycemia [40]. At 28 weeks, there were increased ocular VEGF concentrations, with significant immunoreactivity in the choroid [41]. This corresponds to the VEGF localization in the eyes of diabetic patients [41]. With 12 months of chronic hyperglycemia, microglia were activated and migrated into the subretinal space between the outer segment and RPE [42], as seen in human proliferative diabetic retinopathy (PDR) with diabetic maculopathy [43]. These retinal microcirculatory changes, however, were not accompanied with retinal vessel diameter or morphological changes. The stable diabetic state of this model allows for the investigation of retinal changes over an extended duration of diabetes.

1.1.3.6. Spontaneously diabetic Torii (SDT) rats

The spontaneously diabetic Torii (SDT) rat is a type 2 diabetic inbred strain of the Sprague-Dawley rat generated by selective breeding of SD rats with polyuria and glucosuria. The rats display glucosuria, hypoinsulinemia, and hyperglycemia without signs of ketonuria. There are prominent sex differences in diabetes development in these rats. Female and male rats develop hyperglycemia by 10 and 20 weeks, respectively, but 100% cumulative incidence of diabetes was only observed in male 40-week-old rats [44]. Merely, 33.3% of female rats developed diabetes by 65 weeks of age [44].

The SDT rat is one of the few rodent models that display advanced lesions and features resembling PDR with prolonged hyperglycemia. Increased cellular apoptosis in the GCL and INL and increased GFAP expression spanning the entire retina [45] were evident at the age of 40 weeks. There was increased retinal leukostasis [46] and retinal thickening [47] by the age of 44 and 52–60 weeks, respectively. Long-standing hyperglycemia in rats older than 60 weeks of age has been shown to result in the distortion of the retina, fluorescein leakage around the optic disc, vessel tortuosity, capillary nonperfusion, neovascularization, and tractional retinal detachment accompanied by proliferative fibrous formations [48] (70 weeks) [44, 49]. Anterior chamber hemorrhage with fibrous proliferation around the iris and posterior synechia formation were also noted in a few cases [44, 49]. A major discrepancy between the rat model and human DR is the development of retinal neovascularization and proliferative DR features in the SDT rat in the absence of retinal ischemia [50]. The presence of microaneurysms and areas of vascular nonperfusion were also rarely reported in this model. Retinal dysfunction was evidenced by delayed OP peak latencies at 32 weeks of age [48] and reduced a-wave, b-wave, and OP amplitudes with increased OP implicit times at the age of 44 weeks [51].

The SDT rat has a relatively long lifespan without requiring exogenous insulin administration. Hence, it may serve as a unique rodent model for PDR, considering that the SDT rate retinae can be exposed to high concentrations of VEGF for a longer period of time. Some researchers have further introduced the fa mutation of the leptin receptor gene found in ZF rats into the SDT rat to create a SDT fatty rat that develops diabetes and diabetes-associated complications at a younger age.

1.1.3.7. TetO rat

The NTac:SD-Tg(H1/tetO-RNAi:Insr) (TetO) transgenic rat is a newly reported rat model of T2D exhibiting both hyperglycemia and hyperinsulinemia [52]. The insulin receptor (INSR) gene is reversibly knocked down through the use of tetracycline-inducible small hairpin RNA (shRNA) expression systems [53]. INSR expression is inhibited upon doxycycline administration in drinking water, resulting in insulin resistance. After 4–5 weeks of hyperglycemia, RGC and pericyte loss were observed. The rats displayed an increase in the number of stenotic vessels and veins with abnormal caliber. Blood-retina barrier (BRB) disruption was evidenced by albumin extravasation and reduced zonula occludens-1 expression in the RPE. Increased expression of GFAP and inflammatory markers was also observed in the rat retinae. Functionally, ERG studies revealed significantly decreased scotopic a-waves, b-waves, and c-waves, and an elevated b/a-wave ratio.

The reversible nature of the tetracycline-inducible shRNA system allows for fine modulation of the degree of hyperglycemia to be induced in the rats. This may be useful for the reproduction of clinical situations in an animal model. The rats also project an array of early and late features of human DR within a shorter time span as compared with other rodent models of DR. Given that only one study to date has documented retinopathy changes in the TetO rat, no detailed reports of the timeframe of pathophysiological changes in the retinae are currently available.

1.1.4. Angiogenesis

1.1.4.1. Oxygen-induced Retinopathy (OIR)

In Sprague-Dawley rats, the OIR model predominantly induced vascular changes in the peripheral retina. Two days after pups were transferred to normoxia, there was evident perivascular astrocyte loss [54]. This was followed by significantly increased vascular permeability until P18 [55]. At P18, there was significant reduction in INL, inner plexiform layer (IPL), and total retinal thickness [56] and increased arteriole tortuosity [57]. Increased Müller cell gliosis, as demonstrated by increased GFAP immunoreactivity, was particularly prominent in avascular areas of the retina [56]. Although pericyte apoptosis, a prominent feature of DR, was not observed, pericyte-endothelial interactions were abnormal [54]. This may likewise compromise vascular wall stability. Intravitreal neovascularization was accompanied with decreased blood vessel profiles in the outer and inner plexuses and abnormal vessel tufts extending from the superficial plexus in an extraretinal direction [54, 58]. Functionally, a- and b-wave amplitudes on ERG were reduced in OIR rats [55]. It has been suggested that the degree of vascular hyperpermeability, development of vascular pathologies, and area of avascularity differs, according to the strain of mice used and the degree of VEGF overexpression [55, 59]. Researchers should be meticulous in their selection of rat strains to ensure that desired phenotypes are displayed.

1.2. Nonrodent models

Nonrodent models, including dogs, cats, rabbits, pigs, and zebrafish, have been used for DR studies. Larger animals have eyes that are more similar to human eyes in terms of size and structure. Larger eyes also allow for the extraction of retinal cells and components of the vitreous for further in vitro studies. Nonhuman primates are ultimately most similar to humans anatomically, particularly in regard to the presence of the macula. However, significant costs, ethical challenges, genetic intractability, and feasibility issues regarding the housing and handling of such large animals greatly limit their use. Further studies are required for the establishment of reported features in the following nonrodent models due to the small sample sizes and large intra-species variations in existing reports.

1.2.1. Dog models

Diabetes has been induced in dogs via STZ injections or galactose feeding. Both diabetic and galactose-fed galactosemic dogs display retinal lesions similar to those of human DR, though a long duration of galactose feeding is required for the development of vessel changes associated with PDR. Diabetic dogs displayed capillary BM thickening within 1 year of hyperglycemia, pericyte and smooth muscle cell loss at 4 years, and microaneurysms, acellular capillaries, and IRMAs after 7 years of diabetes [60]. Similar lesions were found in alloxan-induced diabetic dogs after 5 years of diabetes, though such changes were significantly more evident in galactosemic dogs [61]. Galactosemic dogs have been reported to develop features of both NPDR and PDR [62, 63, 64, 65], including intravitreal retinal vascular growth and partial detachment of the posterior vitreous after 84 months of 30% galactose feeding [62]. Some have suggested that pericyte destruction and microaneurysm formation are age dependent, with younger animals developing such symptoms at a faster rate than older animals [63].

1.2.2. Cat models

The cat eye has a retinal and choroidal circulation comparable to that of the human eye [66]. The feline lens advantageously does not develop diabetic cataracts, thus allowing for clear visualization of the fundus [67]. The majority of studies of diabetic cats involve induction of diabetes via pancreatectomy. Among the few studies published, there are discrepancies regarding the presence of reported lesions. Within the small sample size, some observed microaneurysms, capillary BM thickening, increased vessel tortuosity, decreased b-waves, capillary nonperfusion, and small intraretinal hemorrhages [68, 69, 70, 71], while other cats did not develop microaneurysms or hemorrhage [68]. It is worth noting that the inner half of the diabetic retinae becomes hypoxic prior to the development of apparent capillary nonperfusion. This may be comparable with hypoxia-stimulated VEGF expression in humans prior to the development of DR [67]. The use of cats for DR studies is less feasible due to the slow development of lesions and high costs.

1.2.3. Rabbit models

Although the rabbit globe is significantly larger than that of rodents, rabbits possess a merangiotic retinal circulation, where the inner retina is supplied by choriocapillaris [66]. Humans, rodents, dogs, cats, pigs, and primates have a dual holangiotic circulation, where the inner retina is supplied by central and cilioretinal arteries, and the outer retina is supplied by the choriocapillaris. Pharmacological, diet-induced hyperglycemia and hypercholesterolemia (lard, sucrose, and cholesterol-added chow [72]), and VEGF injection models (implantation of a human recombinant VEGF pellet into the vitreous [73, 74]) have been used in rabbits for DR studies. However, the presence of DR lesions is highly variable between and within models. Rabbits with STZ-induced diabetes demonstrated varying degrees of retinopathy after 135 days of hyperglycemia. Some showed proliferative retinopathy, while others only demonstrated moderate vasculopathy with hemorrhages and hard or soft exudates [75]. Diet-induced hyperglycemic and hypercholesterolemic rabbits only exhibited an increased number of microaneurysms after 12–24 weeks of special feeding. The development of vascular pathologies and neovascularization in the VEGF implant model was limited by the gradual depletion of VEGF [73]. Neovascularization developed 14 days after the implant was inserted, but there was almost complete regression by 35 days. The addition of basic fibroblast growth factor (bFGF) to the pellet in another study demonstrated a faster induction of retinopathologies [74], but angiogenic responses may vary depending on the strain of rabbits used.

1.2.4. Porcine models

The retinal structure of the pig eye, with its holangiotic retinal circulation and area centralis comparable to the human macula, closely resembles that of the human eye [66]. Few studies have examined morphological changes in the pig retinae. Alloxan-induced pigs developed Müller cell gliosis at 90 days of hyperglycemia [76]. By 20 weeks, pigs developed capillary BM thickening [77], pericyte loss, and capillary collapse with a reduction in the total number of BRB capillaries [78]. STZ-induced pigs showed similar results, with capillary BM thickening and lamellation and rarefaction on electron microscopy at 20-week post-STZ treatment [79]. A proliferative vitreoretinopathy swine model has also been developed, whereby vitreal and retinal detachments were induced surgically prior to the intravitreal injection of RPE cells [80]. However, it is not a suitable angiogenesis model for DR.

1.2.5. Nonhuman primates

1.2.5.1. Monkey models

Monkeys are the most common primate models used for DR studies. Investigations in 39 STZ-induced, pancreatectomy-induced, or spontaneously hyperglycemic T1D monkeys revealed only mild ocular changes after 6–15 years of diabetes [81]. There were no signs of PDR. Only diabetic monkeys that spontaneously developed hypertension or those that were pharmacologically induced to develop hypertension displayed ischemic retinopathy changes, including microaneurysms, capillary dilatation and dropout, focal intraretinal capillary leakage spots, cotton wool spots, and arteriolar lesions. Spontaneously obese type 2 diabetic monkeys developed various retinal lesions and displayed signs of retinal functional loss on ERG, but lesion development was highly variable among individual monkeys [82, 83]. Occurrence of retinopathy was significantly correlated with the presence of hypertension, as seen in the type 1 diabetic monkey models. Angiogenesis monkey models have also been reported, where repeated intravitreal VEGF injections resulted in vessel tortuosity, dilation, leakage, microaneurysm formation, venous beading, capillary nonperfusion, and preretinal neovascularization [84]. Both VEGF injections and implants resulted in iris neovascularization [73, 85]. Despite their anatomical similarities to human eyes, DR development is highly variable and exceedingly slow, making it an impractical model for DR studies.

1.2.5.2. Marmoset models

An alternate primate model is the marmoset. The marmoset has large eyeballs in respect to its body size with anatomical features closely resembling those of the human eye. The only study to date characterizing the development of retinal DR lesions in these animals reported the development of microaneurysms with increased acellular capillaries, pericyte loss, BM thickening, BRB breakdown, and macular edema in marmosets that were put on a 30% galactose-rich diet for two and a half years [86].

1.2.6. Zebrafish models

Zebrafish (Danio rerio) uniquely offers the tractable genetics of rodent models and a retinal structure resembling that of the human eye. They have the distinctive five-layered human retinal structure, though they lack the subretinal plexi found in the inner and outer retina of human retinae [87, 88]. Despite lacking a macula, the density and number of cones in the zebrafish are comparable to that of humans.

Researchers have developed high-glucose diabetic and angiogenic models to mimic DR. Hyperglycemia was induced by immersing zebrafish into glucose-added water [89, 90]. This resulted in reduced IPL and INL thickness [90] and increased diameter of hyaloid retinal vessels [33]. Zebrafish subjected to a hypoxia-induced retinopathy model developed neovascular features after 3 days [91]. One group developed an angiogenesis model using vhl (von Hippel-Lindau tumor suppressor gene) mutant fish. By inactivating the vhl tumor suppressor gene, hypoxia-inducible factor was upregulated, resulting in overproduction of VEGF. Extensive neovascularization and PDR features were observed in these vhl mutant zebrafish, but such a model is not commercially available.

The short lifespan, rapid development, and capability of zebrafish to breed in exceedingly large numbers allow for high-throughput screening [88], genetic screening, and significantly shorter experimental turnover times as compared with that of rodents. Techniques for developing transgenic lines and gene-targeting mutations have also been established. Although the presence of DR-like lesions and exact models ideal for the simulation of DR have not yet been well established, the zebrafish represents a promising short-term model for future DR drug screening.

Advertisement

2. Next-generation DR models and conclusion

The ideal animal model for DR would mimic the complete pathophysiological process of DR in humans, with initial development of NPDR features and gradual progression to PDR with or without macular edema in the presence of prolonged hyperglycemia. However, the majority of existing animal models only display early or advanced lesions of DR. DR has a complex etiology, with various genetic and environmental factors implicated in its disease susceptibility and progression. Animal models are particularly important for the screening of novel therapeutic interventions. The clinical application of gene therapy for DR has been garnering increasing interest, and this may call for animal models that better reflect the intricacies of DR pathogenesis. Mice and rats are highly genetically tractable, but current transgenic models are based on isolated crosses [92]. Increasing development and use of resources such as The Collaborative Cross [92] may be required to enhance future identification and development of mouse strains with complex traits and epigenetics that are more reflective of the clinical scenario. Only by identifying and manipulating genes that mediate clinically relevant phenotypes can we realize and exploit the full genetic tractability of the mouse to better model DR.

References

  1. 1. Kern TS, et al. Comparison of three strains of diabetic rats with respect to the rate at which retinopathy and tactile allodynia develop. Molecular Vision. 2010;16:1629-1639
  2. 2. Ly A, et al. Early inner retinal astrocyte dysfunction during diabetes and development of hypoxia, retinal stress, and neuronal functional loss. Investigative Ophthalmology & Visual Science. 2011;52(13):9316-9326
  3. 3. Zeng XX, Ng YK, Ling EA. Neuronal and microglial response in the retina of streptozotocin-induced diabetic rats. Visual Neuroscience. 2000;17(3):463-471
  4. 4. Li Q, et al. Early retinal damage in experimental diabetes: Electroretinographical and morphological observations. Experimental Eye Research. 2002;74(5):615-625
  5. 5. Rungger-Brandle E, Dosso AA, Leuenberger PM. Glial reactivity, an early feature of diabetic retinopathy. Investigative Ophthalmology & Visual Science. 2000;41(7):1971-1980
  6. 6. Si YF, et al. Treatment with hydrogen sulfide alleviates streptozotocin-induced diabetic retinopathy in rats. British Journal of Pharmacology. 2013;169(3):619-631
  7. 7. Jariyapongskul A, et al. Long-term effects of oral vitamin C supplementation on the endothelial dysfunction in the iris microvessels of diabetic rats. Microvascular Research. 2007;74(1):32-38
  8. 8. Anderson HR, et al. Diabetic retinopathy: Morphometric analysis of basement membrane thickening of capillaries in different retinal layers within arterial and venous environments. The British Journal of Ophthalmology. 1995;79(12):1120-1123
  9. 9. Hancock HA, Kraft TW. Oscillatory potential analysis and ERGs of normal and diabetic rats. Investigative Ophthalmology & Visual Science. 2004;45(3):1002-1008
  10. 10. Kern TS, et al. Response of capillary cell death to aminoguanidine predicts the development of retinopathy: Comparison of diabetes and galactosemia. Investigative Ophthalmology & Visual Science. 2000;41(12):3972-3978
  11. 11. Schroder S, Palinski W, Schmid-Schonbein GW. Activated monocytes and granulocytes, capillary nonperfusion, and neovascularization in diabetic retinopathy. The American Journal of Pathology. 1991;139(1):81-100
  12. 12. Kowluru RA, Tang J, Kern TS. Abnormalities of retinal metabolism in diabetes and experimental galactosemia. VII. Effect of long-term administration of antioxidants on the development of retinopathy. Diabetes. 2001;50(8):1938-1942
  13. 13. Kern TS, Engerman RL. Galactose-induced retinal microangiopathy in rats. Investigative Ophthalmology & Visual Science. 1995;36(2):490-496
  14. 14. Robison WG Jr et al. Diabetes-related histopathologies of the rat retina prevented with an aldose reductase inhibitor. Experimental Eye Research. 1990;50(4):355-366
  15. 15. Greenwood MD et al. Significant retinal capillary basement membrane thickening in hyperglycemic and normoglycemic diabetic-prone (DP) BB Wistar rats. Ultrastructural Pathology. 2011;35(2):97-105
  16. 16. Blair NP, Tso MO, Dodge JT. Pathologic studies of the blood--retinal barrier in the spontaneously diabetic BB rat. Investigative Ophthalmology & Visual Science. 1984;25(3):302-311
  17. 17. Sima AA, Garcia-Salinas R, Basu PK. The BB Wistar rat: An experimental model for the study of diabetic retinopathy. Metabolism. 1983;32(7 Suppl 1):136-140
  18. 18. Chakrabarti S, Cherian PV, Sima AA. The effect of acarbose on diabetes- and age-related basement membrane thickening in retinal capillaries of the BB/W-rat. Diabetes Research and Clinical Practice. 1993;20(2):123-128
  19. 19. Lai AK, Lo AC. Animal models of diabetic retinopathy: Summary and comparison. Journal of Diabetes Research. 2013;2013:106594
  20. 20. Mori Y, et al. Diabetic strain (WBN/Kob) of rat characterized by endocrine-exocrine pancreatic impairment due to distinct fibrosis. Pancreas. 1990;5(4):452-459
  21. 21. Miyamura N, Amemiya T. Lens and retinal changes in the WBN/Kob rat (spontaneously diabetic strain). Electron-microscopic study. Ophthalmic Research. 1998;30(4):221-232
  22. 22. Bhutto IA, Miyamura N, Amemiya T. Vascular architecture of degenerated retina in WBN/Kob rats: Corrosion cast and electron microscopic study. Ophthalmic Research. 1999;31(5):367-377
  23. 23. Tsuji N, et al. Diabetic retinopathy and choroidal angiopathy in diabetic rats (WBN/Kob). Experimental Animals. 2009;58(5):481-487
  24. 24. Matsuura T, et al. Proliferative retinal changes in diabetic rats (WBN/Kob). Laboratory Animal Science. 1999;49(5):565-569
  25. 25. Mulder GB, Luo S, Gramlich P. The Zucker Diabetic Fatty (ZDF) Rat. C.R.R.m.a. Services, Editor; USA. 2010
  26. 26. Mima A, et al. Retinal not systemic oxidative and inflammatory stress correlated with VEGF expression in rodent models of insulin resistance and diabetes. Investigative Ophthalmology & Visual Science. 2012;53(13):8424-8432
  27. 27. Danis RP, Yang Y. Microvascular retinopathy in the Zucker diabetic fatty rat. Investigative Ophthalmology & Visual Science. 1993;34(7):2367-2371
  28. 28. Yang YS, et al. Acarbose partially inhibits microvascular retinopathy in the Zucker diabetic fatty rat (ZDF/Gmi-fa). Journal of Ocular Pharmacology and Therapeutics. 2000;16(5):471-479
  29. 29. Behl Y, et al. Diabetes-enhanced tumor necrosis factor-alpha production promotes apoptosis and the loss of retinal microvascular cells in type 1 and type 2 models of diabetic retinopathy. The American Journal of Pathology. 2008;172(5):1411-1418
  30. 30. Kawano K, et al. Spontaneous long-term hyperglycemic rat with diabetic complications. Otsuka Long-Evans Tokushima fatty (OLETF) strain. Diabetes. 1992;41(11):1422-1428
  31. 31. Lu ZY, Bhutto IA, Amemiya T. Retinal changes in Otsuka long-evans Tokushima fatty rats (spontaneously diabetic rat)--possibility of a new experimental model for diabetic retinopathy. Japanese Journal of Ophthalmology. 2003;47(1):28-35
  32. 32. Yang JH, et al. Retinal neurodegeneration in type ii diabetic otsuka long-evans tokushima fatty rats. Investigative Ophthalmology & Visual Science. 2013;54(6):3844-3851
  33. 33. Jung KI, et al. Neuroprotective effects of cilostazol on retinal ganglion cell damage in diabetic rats. The Journal of Pharmacology and Experimental Therapeutics. 2013;345(3):457-463
  34. 34. Miyamura N, Bhutto IA, Amemiya T. Retinal capillary changes in Otsuka long-Evans Tokushima fatty rats (spontaneously diabetic strain). Electron-microscopic study. Ophthalmic Research. 1999;31(5):358-366
  35. 35. Bhutto IA, et al. Retinal and choroidal vasculature in rats with spontaneous diabetes type 2 treated with the angiotensin-converting enzyme inhibitor cilazapril: Corrosion cast and electron-microscopic study. Ophthalmic Research. 2002;34(4):220-231
  36. 36. Segawa Y, et al. Upregulation of retinal vascular endothelial growth factor mRNAs in spontaneously diabetic rats without ophthalmoscopic retinopathy. A possible participation of advanced glycation end products in the development of the early phase of diabetic retinopathy. Ophthalmic Research. 1998;30(6):333-339
  37. 37. Goto Y, et al. Development of diabetes in the non-obese NIDDM rat (GK rat). Advances in Experimental Medicine and Biology. 1988;246:29-31
  38. 38. Miyamoto K, et al. Evaluation of retinal microcirculatory alterations in the Goto-Kakizaki rat. A spontaneous model of non-insulin-dependent diabetes. Investigative Ophthalmology & Visual Science. 1996;37(5):898-905
  39. 39. Carmo A, et al. Nitric oxide synthase activity in retinas from non-insulin-dependent diabetic Goto-Kakizaki rats: Correlation with blood-retinal barrier permeability. Nitric Oxide. 2000;4(6):590-596
  40. 40. Agardh CD, et al. Altered endothelial/pericyte ratio in Goto-Kakizaki rat retina. Journal of Diabetes and its Complications. 1997;11(3):158-162
  41. 41. Sone H, et al. Ocular vascular endothelial growth factor levels in diabetic rats are elevated before observable retinal proliferative changes. Diabetologia. 1997;40(6):726-730
  42. 42. Omri S, et al. Microglia/macrophages migrate through retinal epithelium barrier by a transcellular route in diabetic retinopathy: Role of PKCζ in the Goto Kakizaki rat model. The American Journal of Pathology. 2011;179(2):942-953
  43. 43. Zeng HY, Green WR, Tso MO. Microglial activation in human diabetic retinopathy. Archives of Ophthalmology. 2008;126(2):227-232
  44. 44. Shinohara M et al. A new spontaneously diabetic non-obese Torii rat strain with severe ocular complications. International Journal of Experimental Diabetes Research. 2000;1(2):89-100
  45. 45. Fukuda M, et al. Altered expression of aquaporins 1 and 4 coincides with neurodegenerative events in retinas of spontaneously diabetic Torii rats. Experimental Eye Research. 2010;90(1):17-25
  46. 46. Matsuoka M, et al. Leukostasis and pigment epithelium-derived factor in rat models of diabetic retinopathy. Molecular Vision. 2007;13:1058-1065
  47. 47. Toyoda F, et al. Diabetic retinal and choroidal edema in SDT rats. Journal of Diabetes Research. 2016;2016:2345141
  48. 48. Sasase T, et al. Protein kinase C beta inhibitor prevents diabetic peripheral neuropathy, but not histopathological abnormalities of retina in spontaneously diabetic Torii rat. Diabetes, Obesity & Metabolism. 2009;11(11):1084-1087
  49. 49. Kakehashi A, et al. Characteristics of diabetic retinopathy in SDT rats. Diabetes/Metabolism Research and Reviews. 2006;22(6):455-461
  50. 50. Yamada H, et al. Retinal neovascularisation without ischaemia in the spontaneously diabetic Torii rat. Diabetologia. 2005;48(8):1663-1668
  51. 51. Okuno T, et al. Electroretinographic study of spontaneously diabetic Torii rats. Documenta Ophthalmologica. 2008;117(3):191-196
  52. 52. Reichhart N, et al. The TetO rat as a new translational model for type 2 diabetic retinopathy by inducible insulin receptor knockdown. Diabetologia. 2016
  53. 53. Kotnik K, et al. Inducible transgenic rat model for diabetes mellitus based on shRNA-mediated gene knockdown. PLoS One. 2009;4(4):e5124
  54. 54. Downie LE, et al. AT1 receptor inhibition prevents astrocyte degeneration and restores vascular growth in oxygen-induced retinopathy. GLIA. 2008;56(10):1076-1090
  55. 55. Zhang SX, et al. Genetic difference in susceptibility to the blood-retina barrier breakdown in diabetes and oxygen-induced retinopathy. The American Journal of Pathology. 2005;166(1):313-321
  56. 56. Downie LE, et al. Neuronal and glial cell changes are determined by retinal vascularization in retinopathy of prematurity. The Journal of Comparative Neurology. 2007;504(4):404-417
  57. 57. Liu K, et al. The retinal vasculature and function of the neural retina in a rat model of retinopathy of prematurity. Investigative Ophthalmology & Visual Science. 2006;47(6):2639-2647
  58. 58. Fletcher EL, et al. The significance of neuronal and glial cell changes in the rat retina during oxygen-induced retinopathy. Documenta Ophthalmologica. 2010;120(1):67-86
  59. 59. van Wijngaarden P, et al. Strain-dependent differences in oxygen-induced retinopathy in the inbred rat. Investigative Ophthalmology & Visual Science. 2005;46(4):1445-1452
  60. 60. Gardiner TA, et al. Selective loss of vascular smooth muscle cells in the retinal microcirculation of diabetic dogs. The British Journal of Ophthalmology. 1994;78(1):54-60
  61. 61. Kern TS, Engerman RL. Capillary lesions develop in retina rather than cerebral cortex in diabetes and experimental galactosemia. Archives of Ophthalmology. 1996;114(3):306-310
  62. 62. Kador PF, et al. Diabeteslike proliferative retinal changes in galactose-fed dogs. Archives of Ophthalmology. 1995;113(3):352-354
  63. 63. Kador PF, et al. Age-dependent retinal capillary pericyte degeneration in galactose-fed dogs. Journal of Ocular Pharmacology and Therapeutics. 2007;23(1):63-69
  64. 64. Engerman RL, Kern TS. Experimental galactosemia produces diabetic-like retinopathy. Diabetes. 1984;33(1):97-100
  65. 65. Cusick M, et al. Effects of aldose reductase inhibitors and galactose withdrawal on fluorescein angiographic lesions in galactose-fed dogs. Archives of Ophthalmology. 2003;121(12):1745-1751
  66. 66. Bertschinger DR et al. A review of in vivo animal studies in retinal prosthesis research. Graefe's Archive for Clinical and Experimental Ophthalmology. 2008;246(11):1505-1517
  67. 67. Henson MS, O'Brien TD. Feline models of type 2 diabetes mellitus. ILAR Journal. 2006;47(3):234-242
  68. 68. Linsenmeier RA, et al. Retinal hypoxia in long-term diabetic cats. Investigative Ophthalmology & Visual Science. 1998;39(9):1647-1657
  69. 69. Hatchell DL, et al. Diabetic retinopathy in a cat. Experimental Eye Research. 1995;60(5):591-593
  70. 70. Mansour SZ, et al. Reduction of basement membrane thickening in diabetic cat retina by sulindac. Investigative Ophthalmology & Visual Science. 1990;31(3):457-463
  71. 71. Budzynski E, et al. Intraretinal pH in diabetic cats. Current Eye Research. 2005;30(3):229-240
  72. 72. Helfenstein T, et al. Impaired glucose tolerance plus hyperlipidaemia induced by diet promotes retina microaneurysms in New Zealand rabbits. International Journal of Experimental Pathology. 2011;92(1):40-49
  73. 73. Ozaki H, et al. Intravitreal sustained release of VEGF causes retinal neovascularization in rabbits and breakdown of the blood-retinal barrier in rabbits and primates. Experimental Eye Research. 1997;64(4):505-517
  74. 74. Wong CG, et al. Intravitreal VEGF and bFGF produce florid retinal neovascularization and hemorrhage in the rabbit. Current Eye Research. 2001;22(2):140-147
  75. 75. Drago F, et al. Effects of sulfinpyrazone on retinal damage induced by experimental diabetes mellitus in rabbits. Pharmacological Research. 1998;38(2):97-100
  76. 76. King JL, et al. The influence of alloxan-induced diabetes on Muller cell contraction-promoting activities in vitreous. Investigative Ophthalmology & Visual Science. 2011;52(10):7485-7491
  77. 77. Hainsworth DP, et al. Retinal capillary basement membrane thickening in a porcine model of diabetes mellitus. Comparative Medicine. 2002;52(6):523-529
  78. 78. Yang Y, et al. Retinal redox stress and remodeling in cardiometabolic syndrome and diabetes. Oxidative Medicine and Cellular Longevity. 2010;3(6):392-403
  79. 79. Lee SE, et al. Ultrastructural features of retinal capillary basement membrane thickening in diabetic swine. Ultrastructural Pathology. 2010;34(1):35-41
  80. 80. Umazume K, et al. Proliferative vitreoretinopathy in the swine-a new model. Investigative Ophthalmology & Visual Science. 2012;53(8):4910-4916
  81. 81. Tso MO, et al. Microangiopathic retinopathy in experimental diabetic monkeys. Transactions of the American Ophthalmological Society. 1988;86:389-421
  82. 82. Kim SY, et al. Retinopathy in monkeys with spontaneous type 2 diabetes. Investigative Ophthalmology & Visual Science. 2004;45(12):4543-4553
  83. 83. Johnson MA, et al. Ocular structure and function in an aged monkey with spontaneous diabetes mellitus. Experimental Eye Research. 2005;80(1):37-42
  84. 84. Tolentino MJ, et al. Intravitreous injections of vascular endothelial growth factor produce retinal ischemia and microangiopathy in an adult primate. Ophthalmology. 1996;103(11):1820-1828
  85. 85. Tolentino MJ, et al. Vascular endothelial growth factor is sufficient to produce iris neovascularization and neovascular glaucoma in a nonhuman primate. Archives of Ophthalmology. 1996;114(8):964-970
  86. 86. Chronopoulos A, et al. Hyperhexosemia-induced retinal vascular pathology in a novel primate model of diabetic retinopathy. Diabetes. 2015;64(7):2603-2608
  87. 87. Gestri G, Link BA, Neuhauss SC. The visual system of zebrafish and its use to model human ocular diseases. Developmental Neurobiology. 2012;72(3):302-327
  88. 88. Chhetri J, Jacobson G, Gueven N. Zebrafish--on the move towards ophthalmological research. Eye (London, England). 2014;28(4):367-380
  89. 89. Jung SH, et al. High glucose-induced changes in hyaloid-retinal vessels during early ocular development of zebrafish: A short-term animal model of diabetic retinopathy. British Journal of Pharmacology. 2016;173(1):15-26
  90. 90. Gleeson M, Connaughton V, Arneson LS. Induction of hyperglycaemia in zebrafish (Danio Rerio) leads to morphological changes in the retina. Acta Diabetologica. 2007;44(3):157-163
  91. 91. Cao R, et al. Hypoxia-induced retinal angiogenesis in zebrafish as a model to study retinopathy. PLoS One. 2008;3(7):e2748
  92. 92. Churchill GA, et al. The collaborative cross, a community resource for the genetic analysis of complex traits. Nature Genetics. 2004;36(11):1133-1137
  93. 93. Zhang J, et al. Intravitreal injection of erythropoietin protects both retinal vascular and neuronal cells in early diabetes. Investigative Ophthalmology & Visual Science. 2008;49(2):732-742
  94. 94. Gastinger MJ, Singh RS, Barber AJ. Loss of cholinergic and dopaminergic amacrine cells in streptozotocin-diabetic rat and Ins2Akita-diabetic mouse retinas. Investigative Ophthalmology & Visual Science. 2006;47(7):3143-3150
  95. 95. Kohzaki K, Vingrys AJ, Bui BV. Early inner retinal dysfunction in streptozotocin-induced diabetic rats. Investigative Ophthalmology & Visual Science. 2008;49(8):3595-3604
  96. 96. Doczi-Keresztesi Z et al. Retinal and renal vascular permeability changes caused by stem cell stimulation in alloxan-induced diabetic rats, measured by extravasation of fluorescein. In Vivo. 2012;26(3):427-435
  97. 97. Fulton AB, et al. Rod photoreceptors in infant rats with a history of oxygen exposure. Investigative Ophthalmology & Visual Science. 1999;40(1):168-174
  98. 98. Kador PF, et al. Prevention of retinal vessel changes associated with diabetic retinopathy in galactose-fed dogs by aldose reductase inhibitors. Archives of Ophthalmology. 1990;108(9):1301-1309
  99. 99. Kobayashi T, et al. Retinal vessel changes in galactose-fed dogs. Archives of Ophthalmology. 1998;116(6):785-789
  100. 100. Wallow IH, Engerman RL. Permeability and patency of retinal blood vessels in experimental diabetes. Investigative Ophthalmology & Visual Science. 1977;16(5):447-461
  101. 101. van Rooijen E, et al. Von Hippel-Lindau tumor suppressor mutants faithfully model pathological hypoxia-driven angiogenesis and vascular retinopathies in zebrafish. Disease Models & Mechanisms. 2010;3(5-6):343-353

Written By

Larissa H.C. Tang, Ian Y.H. Wong and Amy C.Y. Lo

Reviewed: 16 August 2017 Published: 20 December 2017