Molecular Vision 2010; 16:2121-2131 <http://www.molvis.org/molvis/v16/a227>
Received 16 March 2010 | Accepted 14 October 2010 | Published 19 October 2010

Validation of structural and functional lesions of diabetic retinopathy in mice

T.S. Kern,1,2 J. Tang,1 B.A. Berkowitz3

1Case Western Reserve University, Cleveland, OH; 2Veterans Affairs Medical Center, Cleveland, OH; 3Wayne State University, Detroit, MI

Correspondence to: Timothy S. Kern, Clinical and Molecular Endocrinology, Department of Medicine, 434 Biomedical Research Building, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106-4951; Phone: (216) 368-0800; FAX: (216) 368-5824; email: tsk@case.edu

Abstract

Diabetic retinopathy is a serious long-term complication of diabetes mellitus. There is considerable interest in using mouse models, which can be genetically modified, to understand how retinopathy develops and can be inhibited. Not all retinal lesions that develop in diabetic patients have been reproduced in diabetic mice; conversely, not all abnormalities found in diabetic mice have been studied or identified in diabetic patients. Thus, it is important to recognize which structural and functional abnormalities that develop in diabetic mice have been validated against the lesions that characteristically develop in diabetic patients. Those lesions that have been observed to develop in the mouse models to date are predominantly characteristic of the early stages of retinopathy. Identification of new therapeutic ways to inhibit these early lesions is expected to help inhibit progression to more advanced and clinically important stages of retinopathy.

Introduction

Studies in diabetic animals have provided valuable insight into the pathogenesis of diabetic retinopathy (DR). For example, studies of diabetic dogs demonstrated that improved glycemic control could inhibit the development of the retinopathy more than 16 years before a comparable demonstration in diabetic patients [1]. The possibility of genetic manipulation, and the availability of reagents and antibodies for molecular studies, has led to great interest in smaller species such as the mouse for studies of DR. In addition, the development of noninvasive, translational imaging methods that can be applied to both humans and animals has opened new opportunities for investigating the pathophysiology of DR. Several laboratory species have been compared with regard to which retinal lesions develop in diabetes, with the general conclusion that more types of lesions develop in larger, longer-lived models (primates, dogs) than in smaller, shorter-lived models (rats, mice) [2].

At present, a range of anatomic and functional retinal lesions linked with DR have been identified by a variety of methods. The identification of the lesions that are most critical to this retinopathy, as well as the methods that best demonstrate these abnormalities, has been controversial. In the present text, “robust” lesions are defined as those that occur in both human and mouse models, and have been identified by different laboratories, independent of the methods used. This category does not rule out the possibility that other retinal lesions represent real and important damage inflicted on the retina by diabetes, but indicates only that important inconsistencies and/or questions remain in the literature. To aid future efforts at translational studies, in this review, we will emphasize existing anatomic and physiologic lesions that fit this classification.

1. Diabetic retinopathy in humans

DR is a common complication of diabetes, and one of the leading causes of visual loss in working age populations in developed countries. It has been found to affect the majority of patients who have had diabetes for 1–2 decades, although not all patients develop a comparable severity of retinal disease. The major risk factors of DR are the degree and duration of hyperglycemia. The retinal lesions that develop in type 1 diabetes are not different from those that develop in type 2 diabetes, although the severity and/or incidence of the lesions may differ. For example, the severity of retinal edema and neovascularization differ between different types of diabetes [3].

1A. Characteristic structural changes

1A1. Vascular pathology-- Clinically detectable characteristics of DR have focused on damage to the retinal vasculature. Based on vascular changes, DR is subdivided into an early nonproliferative stage including progressive capillary occlusion and degeneration (NPDR), and a more advanced, proliferative or neovascular stage (PDR) [4]. In addition, macular edema (i.e., retinal edema involving or threatening the macula) can occur in both stages, can be diffuse or focal in distribution, and can have a cystoid appearance or not.

Other vascular lesions of NPDR include the appearance of microaneurysms, vascular nonperfusion, and degeneration (Figure 1). Capillary degeneration and nonperfusion are tightly associated, but it remains unclear if they occur independently of each other, or if one always causes the other. Nonetheless, degenerate capillaries are very important lesions of the retinopathy; they lead to progressive reductions in retinal perfusion, at least locally, as more and more capillaries become occluded [5-7]. Appreciable increases in capillary nonperfusion/degeneration are strongly predictive of (and likely causally related to) progression to the advanced, neovascular stages of retinopathy in patients. Selective loss of pericytes is also common in DR, but perfusion can continue in capillaries missing only some pericytes.

Microaneurysms, or dot hemorrhages, are focal dilations of individual retinal capillaries, and are detectable ophthalmoscopically in diabetic patients. Increasing numbers of microaneurysms have a strong predictive value with respect to the progression of retinopathy [8,9]. Signs of advanced NPDR include venous beading and loops, intraretinal microvascular abnormalities, and large areas of capillary nonperfusion (as assessed using fluorescein angiography).

PDR is observed to develop in areas adjacent to extensive vascular nonperfusion, which are presumably ischemic [10,11]. This advanced stage of DR is characterized by the appearance of new, fragile, and fenestrated retinal blood vessels that penetrate the inner limiting membrane and enter the vitreous (i.e., preretinal neovascularization). These delicate vessels hemorrhage easily. The resulting pooling of blood in the vitreous will reduce in the amount of light reaching the retina and potentially decrease vision. Perhaps more importantly, fibrovascular scar tissue can form around the neovascularization, producing retinal tears and tractional detachment with subsequent blindness.

1A2. Neuroglial pathology-- Diabetes can also damage nonvascular cells of the retina. In autopsy samples, retinal ganglion cells are lost, at least in part via apoptosis [12-19]. In vivo scanning laser polarimetry or optical coherence tomography (OCT) studies have also measured a thinning of the nerve fiber layer in some diabetic patients [20-26]. It is not yet known if ganglion cell loss in diabetic patients is severe enough to contribute to impaired vision.

1A3. Retinal thickness-- OCT studies have also identified retinal thickening and serous retinal detachment in some diabetic patients [27-31]. However, some authors report that foveal thickness is similar among diabetics and nondiabetics when data are controlled for age, race, and sex [32].

1A4. Permeability and edema-- In early NPDR, apparent focal increases in vascular permeability (as assessed by focal accumulation of fluorescein near retinal vessels) are clearly associated with well defined microaneurysms [33-35]. Diffuse accumulation also occurs, but is not specifically associated with microaneurysms. These findings do not rule out an alternative possibility that a functional defect leading to the impaired removal of fluorescein contributes to the accumulation of the contrast agent as well as to the edema [36,37]. Baseline fluorescein accumulation in patients with DR was predictive of progression to photocoagulation for clinically significant macular edema [38].

1A5. Glial activation-- Induction of glial fibrillary acidic protein (GFAP) is a marker of glial activation, and upregulation of this protein occurs in Muller cells from the retinas of diabetic patients [39]. Activation of macro- and microglia occurs in the retinas of diabetic patients, but is of unknown significance [39-43]. Vascular endothelial growth factor, which is strongly implicated in retinal neovascularization and permeability in diabetes, is produced in retinal Muller cells [40].

1B. Characteristic functional defects

The above catalog of structural lesions, while critical in the clinical management of DR, provides little insight into the underlying mechanisms or pathophysiology of the disease, especially during its emerging, clinically silent phase. The pathophysiology of DR has also been extensively studied to better understand the effects of diabetes on the retina. These investigations offer a different perspective on retinal disease caused by diabetes.

1B1. Perfusion and autoregulation-- Much research has been directed at measuring diabetes-induced alterations in retinal perfusion, although there has been substantial disagreement in the literature regarding whether perfusion is increased, decreased, or unchanged [44-48]. The use of multiple different methods for monitoring plasma flow and erythrocyte velocity and transit time has likely contributed to the disparate findings. Nevertheless, steps toward understanding these differences come with evidence that perfusion changes are linked to levels of hyperglycemia and increasing duration of diabetes, along with the severity of retinopathy [47,49]. Retinal perfusion abnormalities are not detectable in patients with tight glycemic control [50].

Measurements of retinal blood flow or perfusion have commonly been made in the unchallenged retina, but since the retinovascular system must constantly adapt to changes in blood pressure and metabolic demand, it is rarely found at steady-state. Dynamic studies of autoregulation, unlike perfusion measures [50], have robustly shown dysfunction of the vascular system in DR [51-54] (Figure 2). Importantly, retinal autoregulatory abnormalities can be detected in patients with tight glycemic control [50]. Whether or not the vascular autoregulation defects or retinal perfusion abnormalities in diabetes are of pathogenic importance is not yet known.

1B2. Retinal function-- Electrophysiological assessments of the retinal function in DR have been made using electroretinogram (ERG), multifocal ERG (mfERG), and visual evoked potential. Diabetes-induced dysfunction includes decreased b-wave amplitude, reduction of oscillatory potential amplitude, and delay in oscillatory potential latency [55-64]. Changes in ERG and mfERG have been used to predict progression of the retinopathy [55-58,65]. ERG and visual evoked potential integrate the response across the whole retina and do not allow the detection of localized abnormalities. Seemingly more useful is mfERG, which allows for functional assessment of multiple small areas of the central retina. Importantly, mfERG studies have been able to analytically predict the retinal locations of new nonproliferative DR development [65]. Another noninvasive method for analytically evaluating intraretinal function with good panretinal spatial resolution involves the measurement of retinal ionic regulation using manganese-enhanced magnetic resonance imaging (MEMRI) [66]. A Food Drug Administration (FDA)-approved manganese-based contrast agent (Teslascan) is beginning to be used to assess clinical applications of MEMRI in DR [67]

Diabetes also negatively impacts psychophysical processes related to vision, such as contrast sensitivity, color vision, and dark adaptation rates [68-71]. The reduction in contrast sensitivity in patients with diabetes, despite normal visual acuity, has been well documented [72,73], and significant losses of contrast sensitivity have been observed in patients with insulin-dependent diabetes mellitus with no evidence of retinopathy when compared with nondiabetic controls [74-81]. Spatial frequencies in the mid to high range are especially sensitive to diabetes [74,76,77,79,81]. These defects can occur before overt evidence of retinopathy is present [82]. Such deficits are not necessarily retina-specific, and could reflect changes anywhere in the visual system (from anterior chamber to visual cortex and higher central processing aspects in the brain). Diabetic patients take longer to dark adapt than age-matched controls [68]; this prolongation is likely caused by alterations in the photoreceptor/pigment epithelial complex.

2. Mice as models of diabetic retinopathy

A variety of different animal species (including primates, dogs, cats, pigs, rats, and mice) have been used to investigate the pathogenesis of retinopathy. The advantages and disadvantages of those species have been summarized elsewhere [2], and here we will focus only on mice, which offer considerable promise as models due largely to the availability of molecular tools to investigate the pathogenesis of the disease. To date, the effects of diabetes on the development of retinopathy in mice have been studied only in a small number of strains, so the following summary likely will need to be amended in the future.

2A. Characteristic structural changes

2A1. Vascular pathology-- Over a period of 6–9 months of diabetes, several strains of mice (including the C57Bl/6J, Ins2Akita, db/db, KK.Cg-Ay/J(KKAY), and Non-Obese Diabetic strains) develop similar microvascular changes of the retina that are consistent with the early clinical stages of NPDR (capillary degeneration, pericyte loss, thickening of the capillary basement membrane) [83-88]. C57Bl/6J mice made diabetic with streptozotocin begin to show capillary degeneration and pericyte loss histologically at about six months after the onset diabetes (Figure 1); these lesions become more numerous with increasing duration of the diabetes [88-90]. Males of the Ins2Akita strain develop a spontaneous type 1 diabetes (females are less severely affected), with subsequent vascular histopathology [85] that is comparable in severity and progression rate to that in streptozotocin-treated C57Bl/6 mice. Likewise, genetically diabetic db/db mice (type 2 diabetes) develop strand-like and degenerate capillaries, as well as an increase in the ratio of endothelial cells to pericytes (interpreted as a loss of pericytes) relative to nondiabetic controls [87,91]. Crossing db/db mice with apolipoprotein E–deficient mice results in both hyperglycemia and hyperlipidemia [86], and these crossed mice exhibit accelerated degeneration of the retinal capillaries and pericytes compared with littermate controls [86]. The KK mouse strain exhibits pericyte ghosts, degenerate capillaries, and occasional microaneurysms between 20 and 64 weeks of age [92]. Thus, diabetic mice develop a similar vascular histopathology at prolonged progression rates to patients with early DR.

Genetically modified diabetic mice, however, can develop these structural lesions at an accelerated rate. For example, diabetic mice deficient in endothelial nitric oxide synthase are reported to have an earlier onset and increased number of acellular capillaries, sustained gliosis, and increased capillary basement membrane thickness than that reported for diabetic C57BL/6 mice [93]. In another example, KKAY mice have changes in their retinal capillaries after only three months of diabetes [84]. The changes reported were relatively nonspecific (endothelial hyperplasia, basement membrane thickening, and some edema and vacuolar degeneration of capillary cells), whereas none of the more characteristic lesions of DR (microaneurysms, capillary degeneration, pericyte loss, etc.) were discussed. Since the lesions detected were present after only short durations of diabetes, it is possible that they might be unrelated to diabetes.

Microaneurysms comparable to the saccular capillary lesions characteristic of diabetes in patients are not robustly reported in C57Bl/6 or Ins2Akita mice [85,88,89,94]. Some KK mice have been reported to show occasional microaneurysm-like vascular abnormalities in old animals [92].

Thickening of the vascular basement membrane has been detected in retinal capillaries in mice with chemically induced [88,95] and spontaneous diabetes [96-98] after about six months diabetes. This change is not specific for the retina (as it is also found in kidney, brain, and other sites [99,100]), or even for diabetes [101].

A common criticism of the mouse model of DR is that it apparently does not develop preretinal neovascularization or other advanced lesions (retinal edema, hemorrhages, microaneurysms) of retinopathy [94,102]. The relatively short duration of diabetes (most studies are <1 year) and mouse lifespan, as well as the resulting modest extent of capillary degeneration that develops during this short interval, are probable reasons that neovascularization has not been observed in these models.

Nevertheless, some reports have claimed finding retinal neovascularization. Fifteen-month-old db/db mice showed increased density of retinal capillaries in the inner nuclear layer, which was interpreted as evidence of vessel proliferation [87]. However, these vessels did not extend into the vitreous body. J129sv/B16 mice diabetic for four months have been reported to develop neovascularization; the claimed new vessels were demonstrated by injecting a contrast medium (black ink) into the vascular lumen [103]. The “neovascularization” was poorly demonstrated, and could possibly be increased retinal vascular density within the retina instead.

2A2. Neuroglial pathology-- Neuronal cells of the retina also are affected by diabetes, resulting in dysfunction and even degeneration of some neuronal cells in humans and rats. Findings in mice, however, are more controversial, and have not necessarily been in agreement with findings in rats [88,104-107].

The spontaneous development of diabetes resulted in loss of cells in the ganglion cell layer in Ins2Akita mice [85,108]. After 5–6 months of hyperglycemia, there was a significant reduction in the number of cell bodies in the retinal ganglion cell layer, which was accompanied by a significant reduction in the thickness of the inner plexiform layer in these animals. By crossing Ins2Akita with mice that express fluorescent proteins under the regulation of the Thy1 promoter, Gastinger et al. [109] demonstrated that diabetes caused 16% depletion of ganglion cells from the peripheral retina, but not from the central region. Dopaminergic and cholinergic amacrine cells are also lost from the retina in diabetes [110].

The C57Bl/6J mouse strain has also been evaluated for diabetes-induced neurodegeneration in the retina. Some investigators reported a 20% to 25% reduction in cells of the ganglion cell layer of the retina at as early as 14 weeks of diabetes [104,107,111]. Others did not detect any ganglion cell loss at diabetes durations of up to one year [88,105,106,112]. This difference between findings remains unexplained. Fifteen-month-old db/db mice were reported to have increased apoptosis of retinal ganglion cells and other cells of the neural retina [87]. After one month of diabetes, the numbers of apoptotic cells in the retinal ganglion cells and inner nuclear layers were significantly greater in the diabetic KKAY mice than in the control group, and the rate of cell death increased with duration of diabetes [84].

2A3. Retinal thickness-- Studies of diabetic mice to date have shown thinning of the retina [85,90], in contrast to the retinal thickening seen in diabetic patients with retinal edema. The relative thickness of retinal layers has been found to be reduced in Ins2Akita mice after 22 weeks of hyperglycemia [85], and in some (but not all) studies of C57Bl/6 mice made diabetic using streptozotocin [88,90,104,113]. As diabetic C57Bl/6 mice aged, MEMRI-derived measurements of retinal thickness in vivo decreased in a linear fashion [66].

2A4. Permeability-- A variety of techniques measuring accumulation of material from plasma in the neural retina of diabetic mice have been investigated to assess permeability [85,89,114-118]. Such accumulation seems diffuse in nature, and focal defects have not been reproducibly described in diabetic mice. Importantly, interpretations of techniques involving such tracer accumulation have not been validated in terms of the permeability surface area product (the gold standard metric for assessment of vascular leakage). Furthermore, whether or not the detection sensitivity and “lesion-type” (i.e., paracellular versus intracellular path defects) sensitivity of these assays are similar has not been established, making comparisons between laboratories/methods uncertain. Despite the indication of increased permeability in diabetes, edema has not been demonstrated in the retina of diabetic mice based on retinal thickness measurements (see section 2A3) [66,90]. Diabetic endothelial nitric oxide synthase−/− mice exhibit accelerated and more severe retinal vascular permeability than age-matched diabetic control mice [93].

2A5. Glial activation-- Diabetes has not been found to result in upregulation of GFAP in Muller glial cells in retinas of C57Bl/6 mice [88,105] or Ins2Akita diabetic mice [85]. In contrast, db/db diabetic mice were reported to show GFAP induction in diabetes, and this glial activation was inhibited in animals lacking aldose reductase [87]. Activation of microglia (based on shape change) also occurs in the retinas of diabetic mice [106], but is of unknown significance.

2B. Characteristic functional defects

2B1. Perfusion and autoregulation-- Only a few studies have been performed in diabetic mice [119-121], but there is controversy over changes in steady-state retinal perfusion. Nonetheless, diabetic mice demonstrate robust alterations in retinal autoregulatory ability in response to a provocation, similar to the defect reported in patients and other experimental models [119-123] (Figure 2). Correction of the retinal autoregulatory defect in diabetic mice is tightly linked with normalization of biochemical abnormalities, and is predictive of subsequent treatment efficacy [90,123,124].

2B2. Retinal function-- The primary means of assessing visual function in the mouse has been using electrophysiology methods such as ERG [86,90,125,126]. These ERG data provide evidence for dysfunction of both the outer retina and inner retina in DR. However, because the ERG measures a summed response from the entire retina, the relationship between such early dysfunction and later vascular histopathology remains unclear.

Assessment of intraretinal function in diabetes using MEMRI has also been reported, and genetic modifications that inhibited development of the vascular lesions of DR have been found to normalize the MEMRI response [66], suggesting a link between these events.

A method of assessing visual processes themselves (contrast sensitivity and visual acuity) in mice has recently become available [127-129] via measurement of the optokinetic response. Using this method, diabetes-induced reductions in contrast sensitivity and visual acuity have been demonstrated in mice ( [130], Kern et al. submitted, and Berkowitz et al. submitted).

3. Robust endpoints of retinopathy for use in diabetic mice

DR consists of a spectrum of retinal structural and functional lesions. Since DR is a composite of several different lesions, the presence of any one type of lesion is not sufficient to claim that it is present, nor is the correction of any one lesion sufficient to claim that the retinopathy has been inhibited. The fact that a particular species or strain does not develop the full spectrum of those lesions of retinopathy is obviously unfortunate, but likely is due, at least in part, to important differences between humans and rodents in the duration over which they are exposed to diabetes. The lesions which do develop in the animal models are characteristic of the early stages of retinopathy, and are worthy of continued study to identify promising new therapeutic targets at which to inhibit progression of the retinopathy to more advanced and clinically important stages (Table 1).

Characteristics of DR in patients that have not been reproducibly replicated to date in diabetic mice are preretinal neovascularization, saccular microaneurysms, retinal hemorrhage, and retinal thickening due to edema.

Nevertheless, several abnormalities regarded as important in the development of DR (or at least characteristic of it) in humans have been robustly identified in diabetic mouse models. These are the early diabetes–induced degeneration of retinal capillaries (Figure 1), loss of capillary pericytes and neuroglia, impairment in vessel autoregulation (Figure 2), and deterioration of nonvascular retinal function. The capillary degeneration/nonperfusion seems to be especially important, because it is found in patients with NPDR and is clinically linked to the eventual development of PDR. A variety of validated techniques have been used to assess the capillary degeneration and the results of using different methods to isolate or view the retinal vessels (trypsin digest method, elastase method, whole-mount immunohistochemistry) have yielded similar conclusions. In contrast, a range of more or less validated (against standard physiologic methods) techniques have been use to, for example, evaluate retinal blood flow, but with less agreement between the methods. Thus, it is unclear if such disagreements are methodological or biologic in nature. Better attempts to validate and improve the physiologic accuracy of a chosen technique should help in addressing this issue.

In addition to the “standard” techniques that have been used to demonstrate changes in diabetic patients and rodents, new techniques (including adaptive optics, OCT, oxygenation mapping techniques, and MRI-based methods) are offering exciting new and potentially noninvasive ways to assess these (and other) DR lesions in the future.

Acknowledgments

Supported by NIH EY00300 (T.S.K.) and EY010221 (B.A.B.), by a Merit grant from the Medical Research Service of the Department of Veteran Affairs (T.S.K.), and by grants from the Juvenile Diabetes Research Foundation (B.A.B.); National Institutes of Health Animal Models of Diabetic Complications Consortium and Mouse Metabolic Phenotyping Centers Pilot and Feasibility Programs (B.A.B.), and by an unrestricted grant from Research to Prevent Blindness (Kresge Eye Institute). We thank LP Aiello, M.D., Ph.D., for his comments and suggestions related to a draft of this manuscript.

References

  1. Engerman R, Bloodworth JMB, , Jr Nelson S. Relationship of microvascular disease in diabetes to metabolic control. Diabetes. 1977; 26:760-9. [PMID: 885298]
  2. Kern TS. In vivo models of diabetic retinopathy. In: Duh EJ, editor. Diabetic Retinopathy. Totowa, New Jersey: Humana Press; 2008. p. 137–156.
  3. Klein R, Davis MD, Moss SE, Klein BE, DeMets DL. The Wisconsin Epidemiologic Study of Diabetic Retinopathy. A comparison of retinopathy in younger and older onset diabetic persons. Adv Exp Med Biol. 1985; 189:321-35. [PMID: 4036719]
  4. Davis MD, Kern TS, Rand LI. Diabetic Retinopathy. In: Alberti KGMM, Zimmet P, DeFronzo RA, editors. International Textbook of Diabetes Mellitus. 2 ed. New York: John Wiley & Sons; 1997. p. 1413–46.
  5. Kohner EM, Henkind P. Correlation of fluorescein angiogram and retinal digest in diabetic retinopathy. Am J Ophthalmol. 1970; 69:403-14. [PMID: 4907465]
  6. De Venecia G, Davis MD, Engerman RL. Clinicopathologic correlations in diabetic retinopathy. 1. Histology and fluorescein angiography of microaneurysms. Arch Ophthalmol. 1976; 94:1766-73. [PMID: 973822]
  7. Shimizu K, Kobayashi Y, Muraoka K. Midperipheral fundus involvement in diabetic retinopathy. Ophthalmology. 1981; 88:601-12. [PMID: 6167923]
  8. Kohner EM, Sleightholm M. Does microaneurysm count reflect severity of early diabetic retinopathy? Ophthalmology. 1986; 93:586-9. [PMID: 3725317]
  9. Kohner EM, Stratton IM, Aldington SJ, Turner RC, Matthews DR. Microaneurysms in the development of diabetic retinopathy (UKPDS 42). UK Prospective Diabetes Study Group. Diabetologia. 1999; 42:1107-12. [PMID: 10447523]
  10. Bresnick GH, Engerman R, Davis MD, de Venecia G, Myers FL. Patterns of ischemia in diabetic retinopathy. Trans Sect Ophthalmol Am Acad Ophthalmol Otolaryngol. 1976; 81:OP694-709. [PMID: 822560]
  11. Bresnick GH, De Venecia G, Myers FL, Harris JA, Davis MD. Retinal ischemia in diabetic retinopathy. Arch Ophthalmol. 1975; 93:1300-10. [PMID: 1200895]
  12. Wolter JR. Diabetic retinopathy. Am J Ophthalmol. 1961; 51:1123-41. [PMID: 13786453]
  13. Bloodworth JM, Jr. Diabetic retinopathy. Diabetes. 1962; 11:1-22. [PMID: 13870120]
  14. Bloodworth JM, , Jr Molitor DL. Ultrastructural aspects of human and canine diabetic retinopathy. Invest Ophthalmol. 1965; 4:1037-48. [PMID: 5848759]
  15. Kerrigan LA, Zack DJ, Quigley HA, Smith SD, Pease ME. TUNEL-positive ganglion cells in human primary open-angle glaucoma. Arch Ophthalmol. 1997; 115:1031-5. [PMID: 9258226]
  16. Barber AJ, Lieth E, Khin SA, Antonetti DA, Buchanan AG, Gardner TW. Neural apoptosis in the retina during experimental and human diabetes. Early onset and effect of insulin. J Clin Invest. 1998; 102:783-91. [PMID: 9710447]
  17. Abu-El-Asrar AM, Dralands L, Missotten L, Al-Jadaan IA, Geboes K. Expression of apoptosis markers in the retinas of human subjects with diabetes. Invest Ophthalmol Vis Sci. 2004; 45:2760-6. [PMID: 15277502]
  18. Abu El-Asrar AM, Dralands L, Missotten L, Geboes K. Expression of antiapoptotic and proapoptotic molecules in diabetic retinas. Eye. 2007; 21:238-45. [PMID: 16424911]
  19. Oshitari T, Yamamoto S, Hata N, Roy S. Mitochondria- and caspase-dependent cell death pathway involved in neuronal degeneration in diabetic retinopathy. Br J Ophthalmol. 2008; 92:552-6. [PMID: 18369072]
  20. Chihara E, Matsuoka T, Ogura Y, Matsumura M. Retinal nerve fiber layer defect as an early manifestation of diabetic retinopathy. Ophthalmology. 1993; 100:1147-51. [PMID: 8341494]
  21. Takahashi H, Goto T, Shoji T, Tanito M, Park M, Chihara E. Diabetes-associated retinal nerve fiber damage evaluated with scanning laser polarimetry. Am J Ophthalmol. 2006; 142:88-94. [PMID: 16815255]
  22. Chihara E, Zhang S. Analysis of diabetic optic neuropathy with a topographic laser scanning system. Nippon Ganka Gakkai Zasshi. 1998; 102:431-5. [PMID: 9720364]
  23. Ozdek S, Lonneville YH, Onol M, Yetkin I, Hasanreisoglu BB. Assessment of nerve fiber layer in diabetic patients with scanning laser polarimetry. Eye. 2002; 16:761-5. [PMID: 12439673]
  24. Lopes de Faria JM, Russ H, Costa VP. Retinal nerve fibre layer loss in patients with type 1 diabetes mellitus without retinopathy. Br J Ophthalmol. 2002; 86:725-8. [PMID: 12084737]
  25. van Dijk HW, Kok PH, Garvin M, Sonka M, Devries JH, Michels RP, van Velthoven ME, Schlingemann RO, Verbraak FD, Abràmoff MD. Selective loss of inner retinal layer thickness in type 1 diabetic patients with minimal diabetic retinopathy. Invest Ophthalmol Vis Sci. 2009; 50:3404-9. [PMID: 19151397]
  26. van Dijk HW, Verbraak FD, Kok PH, Garvin MK, Sonka M, Lee K, Devries JH, Michels RP, van Velthoven ME, Schlingemann RO, Abràmoff MD. Decreased retinal ganglion cell layer thickness in patients with type 1 diabetes. Invest Ophthalmol Vis Sci. 2010; 51:3660-5. [PMID: 20130282]
  27. Lang GE. Optical coherence tomography findings in diabetic retinopathy. Dev Ophthalmol. 2007; 39:31-47. [PMID: 17245077]
  28. Gaucher D, Tadayoni R, Erginay A, Haouchine B, Gaudric A, Massin P. Optical coherence tomography assessment of the vitreoretinal relationship in diabetic macular edema. Am J Ophthalmol. 2005; 139:807-13. [PMID: 15860284]
  29. Browning DJ, Glassman AR, Aiello LP, Bressler NM, Bressler SB, Danis RP, Davis MD, Ferris FL, Huang SS, Kaiser PK, Kollman C, Sadda S, Scott IU, Qin H, Diabetic Retinopathy Clinical Research Network.. Optical coherence tomography measurements and analysis methods in optical coherence tomography studies of diabetic macular edema. Ophthalmology. 2008; 115:1366-71. [PMID: 18675696]
  30. Cho HY, Lee DH, Chung SE, Kang SW. Diabetic retinopathy and peripapillary retinal thickness. Korean J Ophthalmol. 2010; 24:16-22. [PMID: 20157409]
  31. Koleva-Georgieva DN, Sivkova NP. Optical coherence tomography for the detection of early macular edema in diabetic patients with retinopathy. Folia Med (Plovdiv). 2010; 52:40-8. [PMID: 20380286]
  32. Kashani AH, Zimmer-Galler IE, Shah SM, Dustin L, Do DV, Eliott D, Haller JA, Nguyen QD. Retinal thickness analysis by race, gender, and age using Stratus OCT. Am J Ophthalmol. 2010; 149:496-502. [PMID: 20042179]
  33. Lobo CL, Bernardes RC, Santos FJ, Cunha-Vaz JG. Mapping retinal fluorescein leakage with confocal scanning laser fluorometry of the human vitreous. Arch Ophthalmol. 1999; 117:631-7. [PMID: 10326960]
  34. Sander B, Larsen M, Moldow B, Lund-Andersen H. Diabetic macular edema: passive and active transport of fluorescein through the blood-retina barrier. Invest Ophthalmol Vis Sci. 2001; 42:433-8. [PMID: 11157879]
  35. Knudsen ST, Bek T, Poulsen PL, Hove MN, Rehling M, Mogensen CE. Macular edema reflects generalized vascular hyperpermeability in type 2 diabetic patients with retinopathy. Diabetes Care. 2002; 25:2328-34. [PMID: 12453981]
  36. Grimes PA. Fluorescein distribution in retinas of normal and diabetic rats. Exp Eye Res. 1985; 41:227-38. [PMID: 4065247]
  37. Grimes PA. Carboxyfluorescein distribution in ocular tissues of normal and diabetic rats. Curr Eye Res. 1988; 7:981-8. [PMID: 2976362]
  38. Sander B, Thornit DN, Colmorn L, Strøm C, Girach A, Hubbard LD, Lund-Andersen H, Larsen M. Progression of diabetic macular edema: correlation with blood retinal barrier permeability, retinal thickness, and retinal vessel diameter. Invest Ophthalmol Vis Sci. 2007; 48:3983-7. [PMID: 17724176]
  39. Mizutani M, Gerhardinger C, Lorenzi M. Muller cell changes in human diabetic retinopathy. Diabetes. 1998; 47:445-9. [PMID: 9519752]
  40. Amin RH, Frank RN, Kennedy A, Eliott D, Puklin JE, Abrams GW. Vascular endothelial growth factor is present in glial cells of the retina and optic nerve of human subjects with nonproliferative diabetic retinopathy. Invest Ophthalmol Vis Sci. 1997; 38:36-47. [PMID: 9008628]
  41. Carrasco E, Hernandez C, Miralles A, Huguet P, Farres J, Simo R. Lower somatostatin expression is an early event in diabetic retinopathy and is associated with retinal neurodegeneration. Diabetes Care. 2007; 30:2902-8. [PMID: 17704349]
  42. Zeng XX, Ng YK, Ling EA. Neuronal and microglial response in the retina of streptozotocin-induced diabetic rats. Vis Neurosci. 2000; 17:463-71. [PMID: 10910112]
  43. Zeng HY, Green WR, Tso MO. Microglial activation in human diabetic retinopathy. Arch Ophthalmol. 2008; 126:227-32. [PMID: 18268214]
  44. Bertram B, Wolf S, Fiehofer S, Schulte K, Arend O, Reim M. Retinal circulation times in diabetes mellitus type I. Br J Ophthalmol. 1991; 75:462-5. [PMID: 1873263]
  45. Kohner EM. The retinal blood flow in diabetes. Diabete Metab. 1993; 19:401-4. [PMID: 8056118]
  46. Feke GT, Yoshida A, Ogasawara H, Konno S, Goger DG, Buzney SM, McMeel JW. Retinal blood flow increases following short-term aspirin usage in type I diabetics with no or minimal retinopathy. Ophthalmic Res. 1996; 28:108-16. [PMID: 8792361]
  47. Konno S, Feke GT, Yoshida A, Fujio N, Goger DG, Buzney SM. Retinal blood flow changes in type I diabetes. A long-term follow-up study. Invest Ophthalmol Vis Sci. 1996; 37:1140-8. [PMID: 8631628]
  48. Bursell SE, Clermont AC, Aiello LP, Aiello LM, Schlossman DK, Feener EP, Laffel L, King GL. High-dose vitamin E supplementation normalizes retinal blood flow and creatinine clearance in patients with type 1 diabetes. Diabetes Care. 1999; 22:1245-51. [PMID: 10480765]
  49. Feke GT, Buzney SM, Ogasawara H, Fujio N, Goger DG, Spack NP, Gabbay KH. Retinal circulatory abnormalities in type 1 diabetes. Invest Ophthalmol Vis Sci. 1994; 35:2968-75. [PMID: 8206714]
  50. Lorenzi M, Feke GT, Cagliero E, Pitler L, Schaumberg DA, Berisha F, Nathan DM, McMeel JW. Retinal haemodynamics in individuals with well-controlled type 1 diabetes. Diabetologia. 2008; 51:361-4. [PMID: 18026927]
  51. Grunwald JE, Brucker AJ, Braunstein SN, Schwartz SS, Baker L, Petrig BL, Riva CE. Strict metabolic control and retinal blood flow in diabetes mellitus. Br J Ophthalmol. 1994; 78:598-604. [PMID: 7918284]
  52. Rassam SM, Patel V, Kohner EM. The effect of experimental hypertension on retinal vascular autoregulation in humans: a mechanism for the progression of diabetic retinopathy. Exp Physiol. 1995; 80:53-68. [PMID: 7734138]
  53. Berkowitz BA, Kowluru RA, Frank RN, Kern TS, Hohman TC, Prakash M. Subnormal retinal oxygenation response precedes diabetic-like retinopathy. Invest Ophthalmol Vis Sci. 1999; 40:2100-5. [PMID: 10440266]
  54. Berkowitz BA, Ito Y, Kern TS, McDonald C, Hawkins R. Correction of early subnormal superior hemiretinal ΔPO2 predicts therapeutic efficacy in experimental diabetic retinopathy. Invest Ophthalmol Vis Sci. 2001; 42:2964-9. [PMID: 11687543]
  55. Bresnick GH, Korth K, Groo A, Palta M. Electroretinographic oscillatory potentials predict progression of diabetic retinopathy. Preliminary report. Arch Ophthalmol. 1984; 102:1307-11. [PMID: 6383303]
  56. Bresnick GH, Palta M. Oscillatory potential amplitudes. Relation to severity of diabetic retinopathy. Arch Ophthalmol. 1987; 105:929-33. [PMID: 3606452]
  57. Bresnick GH, Palta M. Predicting progression to severe proliferative diabetic retinopathy. Arch Ophthalmol. 1987; 105:810-4. [PMID: 3579713]
  58. Bresnick GH, Palta M. Temporal aspects of the electroretinogram in diabetic retinopathy. Arch Ophthalmol. 1987; 105:660-4. [PMID: 3619742]
  59. Juen S, Kieselbach GF. Electrophysiological changes in juvenile diabetics without retinopathy. Arch Ophthalmol. 1990; 108:372-5. [PMID: 2310337]
  60. Holopigian K, Seiple W, Lorenzo M, Carr R. A comparison of photopic and scotopic electroretinographic changes in early diabetic retinopathy. Invest Ophthalmol Vis Sci. 1992; 33:2773-80. [PMID: 1526726]
  61. Fortune B, Schneck ME, Adams AJ. Multifocal electroretinogram delays reveal local retinal dysfunction in early diabetic retinopathy. Invest Ophthalmol Vis Sci. 1999; 40:2638-51. [PMID: 10509661]
  62. Greenstein VC, Holopigian K, Hood DC, Seiple W, Carr RE. The nature and extent of retinal dysfunction associated with diabetic macular edema. Invest Ophthalmol Vis Sci. 2000; 41:3643-54. [PMID: 11006263]
  63. Klemp K, Sander B, Brockhoff PB, Vaag A, Lund-Andersen H, Larsen M. The multifocal ERG in diabetic patients without retinopathy during euglycemic clamping. Invest Ophthalmol Vis Sci. 2005; 46:2620-6. [PMID: 15980256]
  64. Tyrberg M, Ponjavic V, Lovestam-Adrian M. Multifocal electroretinogram (mfERG) in patients with diabetes mellitus and an enlarged foveal avascular zone (FAZ). Doc Ophthalmol. 2008; 117:185-9. [PMID: 18324430]
  65. Bearse MA, , Jr Adams AJ, Han Y, Schneck ME, Ng J, Bronson-Castain K, Barez S. A multifocal electroretinogram model predicting the development of diabetic retinopathy. Prog Retin Eye Res. 2006; 25:425-48. [PMID: 16949855]
  66. Berkowitz BA, Gradianu M, Bissig D, Kern TS, Roberts R. Retinal ion regulation in a mouse model of diabetic retinopathy: natural history and the effect of Cu/Zn superoxide dismutase overexpression. Invest Ophthalmol Vis Sci. 2009; 50:2351-8. [PMID: 19074809]
  67. Tofts PS, Porchia A, Jin Y, Roberts R, Berkowitz BA. Toward clinical application of manganese-enhanced MRI of retinal function. Brain Res Bull. 2010; 81:333-8. [PMID: 19524028]
  68. Henson DB, North RV. Dark adaptation in diabetes mellitus. Br J Ophthalmol. 1979; 63:539-41. [PMID: 476028]
  69. Newsome DA, Negreiro M. Reproducible measurement of macular light flash recovery time using a novel device can indicate the presence and worsening of macular diseases. Curr Eye Res. 2009; 34:162-70. [PMID: 19219688]
  70. Kiser AK, Mladenovich D, Eshraghi F, Bourdeau D, Dagnelie G. Reliability and consistency of dark-adapted psychophysical measures in advanced eye disease. Invest Ophthalmol Vis Sci. 2006; 47:444-52. [PMID: 16384995]
  71. Midena E, Segato T, Giuliano M, Zucchetto M. Macular recovery function (nyctometry) in diabetics without and with early retinopathy. Br J Ophthalmol. 1990; 74:106-8. [PMID: 2310723]
  72. Hyvärinen L, Rovamo J, Laurinen P, Peltomaa A. Contrast sensitivity function in evaluation of visual impairment due to retinitis pigmentosa. Acta Ophthalmol (Copenh). 1981; 59:763-73. [PMID: 7315229]
  73. Sokol S, Moskowitz A, Skarf B, Evans R, Molitch M, Senior B. Contrast sensitivity in diabetics with and without background retinopathy. Arch Ophthalmol. 1985; 103:51-4. [PMID: 3977675]
  74. Ghafour IM, Foulds WS, Allan D, McClure E. Contrast sensitivity in diabetic subjects with and without retinopathy. Br J Ophthalmol. 1982; 66:492-5. [PMID: 7104265]
  75. Della Sala S, Bertoni G, Somazzi L, Stubbe F, Wilkins AJ. Impaired contrast sensitivity in diabetic patients with and without retinopathy: a new technique for rapid assessment. Br J Ophthalmol. 1985; 69:136-42. [PMID: 3967001]
  76. Trick GL, Burde RM, Gordon MO, Santiago JV, Kilo C. The relationship between hue discrimination and contrast sensitivity deficits in patients with diabetes mellitus. Ophthalmology. 1988; 95:693-8. [PMID: 3262851]
  77. Di Leo MA, Caputo S, Falsini B, Porciatti V, Minnella A, Greco AV, Ghirlanda G. Nonselective loss of contrast sensitivity in visual system testing in early type I diabetes. Diabetes Care. 1992; 15:620-5. [PMID: 1516480]
  78. Banford D, North RV, Dolben J, Butler G, Owens DR. Longitudinal study of visual functions in young insulin dependent diabetics. Ophthalmic Physiol Opt. 1994; 14:339-46. [PMID: 7845689]
  79. Arend O, Remky A, Evans D, Stuber R, Harris A. Contrast sensitivity loss is coupled with capillary dropout in patients with diabetes. Invest Ophthalmol Vis Sci. 1997; 38:1819-24. [PMID: 9286271]
  80. Verrotti A, Lobefalo L, Petitti MT, Mastropasqua L, Morgese G, Chiarelli F, Gallenga PE. Relationship between contrast sensitivity and metabolic control in diabetics with and without retinopathy. Ann Med. 1998; 30:369-74. [PMID: 9783835]
  81. Liska V, Dostalek M. Are contrast sensitivity functions impaired in insulin dependent diabetics without diabetic retinopathy? Acta Medica (Hradec Kralove). 1999; 42:133-8. [PMID: 10812680]
  82. Ewing FM, Deary IJ, Strachan MW, Frier BM. Seeing beyond retinopathy in diabetes: electrophysiological and psychophysical abnormalities and alterations in vision. Endocr Rev. 1998; 19:462-76. [PMID: 9715375]
  83. Naeser P, Agren A. Morphology and enzyme activities of the retinal capillaries in mice with obese hyperglycemic syndrome. Acta Ophthalmol (Copenh). 1978; 56:607-16. [PMID: 153702]
  84. Ning X, Baoyu Q, Yuzhen L, Shuli S, Reed E, Li QQ. Neuro-optic cell apoptosis and microangiopathy in KKAY mouse retina. Int J Mol Med. 2004; 13:87-92. [PMID: 14654976]
  85. Barber AJ, Antonetti DA, Kern TS, Reiter CE, Soans RS, Krady JK, Levison SW, Gardner TW, Bronson SK. The Ins2Akita Mouse as a Model of Early Retinal Complications in Diabetes. Invest Ophthalmol Vis Sci. 2005; 46:2210-8. [PMID: 15914643]
  86. Barile GR, Pachydaki SI, Tari SR, Lee SE, Donmoyer CM, Ma W, Rong LL, Buciarelli LG, Wendt T, Hörig H, Hudson BI, Qu W, Weinberg AD, Yan SF, Schmidt AM. The RAGE axis in early diabetic retinopathy. Invest Ophthalmol Vis Sci. 2005; 46:2916-24. [PMID: 16043866]
  87. Cheung AK, Fung MK, Lo AC, Lam TT, So KF, Chung SS, Chung SK. Aldose reductase deficiency prevents diabetes-induced blood-retinal barrier breakdown, apoptosis, and glial reactivation in the retina of db/db mice. Diabetes. 2005; 54:3119-25. [PMID: 16249434]
  88. Feit-Leichman RA, Kinouchi R, Takeda M, Fan Z, Mohr S, Kern TS, Chen DF. Vascular Damage in a Mouse Model of Diabetic Retinopathy: Relation to Neuronal and Glial Changes. Invest Ophthalmol Vis Sci. 2005; 46:4281-7. [PMID: 16249509]
  89. Joussen AM, Poulaki V, Le ML, Koizumi K, Esser C, Janicki H, Schraermeyer U, Kociok N, Fauser S, Kirchhof B, Kern TS, Adamis AP. A central role for inflammation in the pathogenesis of diabetic retinopathy. FASEB J. 2004; 18:1450-2. [PMID: 15231732]
  90. Zheng L, Du Y, Miller C, Gubitosi-Klug RA, Ball S, Berkowitz BA, Kern TS. Critical role of inducible nitric oxide synthase in degeneration of retinal capillaries in mice with streptozotocin-induced diabetes. Diabetologia. 2007; 50:1987-96. [PMID: 17583794]
  91. Midena E, Segato T, Radin S, di Giorgio G, Meneghini F, Piermarocchi S, Belloni AS. Studies on the retina of the diabetic db/db mouse. I. Endothelial cell-pericyte ratio. Ophthalmic Res. 1989; 21:106-11. [PMID: 2734001]
  92. Duhault J, Lebon F, Boulanger M. KK mice as a model of microangiopathic lesions in diabetes. In: 7th Europ. Conf. Microcirculation. Aberdeen: Karger; 1973. p. 453–458.
  93. Li Q, Verma A, Han PY, Nakagawa T, Johnson RJ, Grant MB, Campbell-Thompson M, Jarajapu YP, Lei B, Hauswirth WW. Diabetic eNOS knockout mice develop accelerated retinopathy. Invest Ophthalmol Vis Sci. 2010; 51:5240-6. [PMID: 3105130]
  94. Gubitosi-Klug R, Kern T. Studies of Diabetic Retinopathy Using Mice. In: Williams LM, CaR W, editors. Eye, Retina, and Visual System of the Mouse. Cambridge, Massachusetts: The MIT Press; 2008. p. 547–557.
  95. Cuthbertson RA, Mandel TE. The effect of murine fetal islet transplants on renal and retinal capillary basement membrane thickness. Transplant Proc. 1987; 19:2919-21. [PMID: 3105130]
  96. Clements RS, , Jr Robison WG, , Jr Cohen MP. Anti-glycated albumin therapy ameliorates early retinal microvascular pathology in db/db mice. J Diabetes Complications. 1998; 12:28-33. [PMID: 9442812]
  97. Carlson EC, Audette JL, Veitenheimer NJ, Risan JA, Laturnus DI, Epstein PN. Ultrastructural morphometry of capillary basement membrane thickness in normal and transgenic diabetic mice. Anat Rec A Discov Mol Cell Evol Biol. 2003; 271:332-41. [PMID: 12629676]
  98. Miller-Lotan R, Miller B, Nakhoul F, Aronson D, Asaf R, Levy AP. Retinal capillary basement membrane thickness in diabetic mice genetically modified at the haptoglobin locus. Diabetes Metab Res Rev. 2007; 23:152-6. [PMID: 16742000]
  99. Rasch R. Prevention of diabetic glomerulopathy in streptozotocin diabetic rats by insulin treatment. Glomerular basement membrane thickness. Diabetologia. 1979; 16:319-24. [PMID: 156660]
  100. Johnson PC, Brendel K, Meezan E. Thickened cerebral cortical capillary basement membranes in diabetes. Arch Pathol Lab Med. 1982; 106:214-7. [PMID: 6896132]
  101. Fatemi SH, Antosh M, Cullan GM, Sharp JG. Late ultrastructural effects of heavy ions and gamma irradiation in the gastrointestinal tract of the mouse. Virchows Arch B Cell Pathol Incl Mol Pathol. 1985; 48:325-40. [PMID: 2861688]
  102. Kern TS, Mohr S. Nonproliferative stages of diabetic retinopathy: animal models and pathogenesis. In: Joussen A, Gardner TW, Kirchof B, Ryan SJ, editors. Retinal Vascular Disease: Springer; 2007.
  103. Ebrahimian TG, Tamarat R, Clergue M, Duriez M, Levy BI, Silvestre JS. Dual effect of angiotensin-converting enzyme inhibition on angiogenesis in type 1 diabetic mice. Arterioscler Thromb Vasc Biol. 2005; 25:65-70. [PMID: 15528473]
  104. Martin PM, Roon P, Van Ells TK, Ganapathy V, Smith SB. Death of retinal neurons in streptozotocin-induced diabetic mice. Invest Ophthalmol Vis Sci. 2004; 45:3330-6. [PMID: 15326158]
  105. Asnaghi V, Gerhardinger C, Hoehn T, Adeboje A, Lorenzi M. A role for the polyol pathway in the early neuroretinal apoptosis and glial changes induced by diabetes in the rat. Diabetes. 2003; 52:506-11. [PMID: 12540628]
  106. Gaucher D, Chiappore JA, Pâques M, Simonutti M, Boitard C, Sahel JA, Massin P, Picaud S. Microglial changes occur without neural cell death in diabetic retinopathy. Vision Res. 2007; 47:612-23. [PMID: 17267004]
  107. Smith SB, Jiang G, Mysona B, Martin-Studdard A, Roon P, Ganapathy V. The sigma receptor-1 (σR1) ligand Pentazocine (PTZ) affords marked protection against neuronal degeneration in the Ins2Akita mouse model of retinopathy. ARVO Annual Meeting; 2007 May 6–10; Fort Lauderdale (FL).
  108. Smith SB, Duplantier J, Dun Y, Mysona B, Roon P, Martin PM, Ganapathy V. In vivo protection against retinal neurodegeneration by sigma receptor 1 ligand (+)-pentazocine. Invest Ophthalmol Vis Sci. 2008; 49:4154-61. [PMID: 18469181]
  109. Gastinger MJ, Kunselman AR, Conboy EE, Bronson SK, Barber AJ. Dendrite remodeling and other abnormalities in the retinal ganglion cells of Ins2Akita diabetic mice. Invest Ophthalmol Vis Sci. 2008; 49:2635-42. [PMID: 18515593]
  110. Gastinger MJ, Singh RS, Barber AJ. Loss of cholinergic and dopaminergic amacrine cells in streptozotocin-diabetic rat and Ins2Akita-diabetic mouse retinas. Invest Ophthalmol Vis Sci. 2006; 47:3143-50. [PMID: 16799061]
  111. 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; 53:971-9. [PMID: 20162412]
  112. Kern TS, Barber AJ. Retinal ganglion cells in diabetes. J Physiol. 2008; 586:4401-8. [PMID: 18565995]
  113. Gubitosi-Klug RA, Talahalli R, Du Y, Nadler JL, Kern TS. 5-Lipoxygenase, but not 12/15-Lipoxygenase, Contributes to Degeneration of Retinal Capillaries in a Mouse Model of Diabetic Retinopathy. Diabetes. 2008; 57:1387-93. [PMID: 18346986]
  114. Joussen AM, Poulaki V, Tsujikawa A, Qin W, Qaum T, Xu Q, Moromizato Y, Bursell SE, Wiegand SJ, Rudge J, Ioffe E, Yancopoulos GD, Adamis AP. Suppression of diabetic retinopathy with angiopoietin-1. Am J Pathol. 2002; 160:1683-93. [PMID: 12000720]
  115. Muranaka K, Yanagi Y, Tamaki Y, Usui T, Kubota N, Iriyama A, Terauchi Y, Kadowaki T, Araie M. Effects of peroxisome proliferator-activated receptor gamma and its ligand on blood-retinal barrier in a streptozotocin-induced diabetic model. Invest Ophthalmol Vis Sci. 2006; 47:4547-52. [PMID: 17003451]
  116. Leal EC, Manivannan A, Hosoya K, Terasaki T, Cunha-Vaz J, Ambrósio AF, Forrester JV. Inducible nitric oxide synthase isoform is a key mediator of leukostasis and blood-retinal barrier breakdown in diabetic retinopathy. Invest Ophthalmol Vis Sci. 2007; 48:5257-65. [PMID: 17962481]
  117. Li J, Wang JJ, Chen D, Mott R, Yu Q, Ma JX, Zhang SX. Systemic administration of HMG-CoA reductase inhibitor protects the blood-retinal barrier and ameliorates retinal inflammation in type 2 diabetes. Exp Eye Res. 2009; 89:71-8. [PMID: 19254713]
  118. Kim JH, Yu YS, Cho CS, Kim KW. Blockade of angiotensin II attenuates VEGF-mediated blood-retinal barrier breakdown in diabetic retinopathy. J Cereb Blood Flow Metab. 2009; 29:621-8. [PMID: 19107135]
  119. Wright WS, Messina JE, Harris NR. Attenuation of diabetes-induced retinal vasoconstriction by a thromboxane receptor antagonist. Exp Eye Res. 2009; 88:106-12. [PMID: 18996116]
  120. 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. [PMID: 12957148]
  121. Wright WS, Harris NR. Ozagrel attenuates early streptozotocin-induced constriction of arterioles in the mouse retina. Exp Eye Res. 2008; 86:528-36. [PMID: 18262522]
  122. Berkowitz BA, Luan H, Gupta RR, Pacheco D, Seidner A, Roberts R, Liggett J, Knoerzer DL, Connor JR, Du Y, Kern TS, Ito Y. Regulation of the early subnormal retinal oxygenation response in experimental diabetes by inducible nitric oxide synthase. Diabetes. 2004; 53:173-8. [PMID: 14693712]
  123. Luan H, Leitges M, Gupta RR, Pacheco D, Seidner A, Liggett J, Ito Y, Kowluru R, Berkowitz BA. Effect of PKCbeta on retinal oxygenation response in experimental diabetes. Invest Ophthalmol Vis Sci. 2004; 45:937-42. [PMID: 14985314]
  124. Berkowitz BA, Luan H, Gupta RR, Pacheco D, Seidner A, Roberts R, Liggett J, Knoerzer DL, Connor JR, Du Y, Kern TS, Ito Y. Regulation of the early subnormal retinal oxygenation response in experimental diabetes by inducible nitric oxide synthase. Diabetes. 2004; 53:173-8. [PMID: 14693712]
  125. Horio N, Clermont AC, Abiko A, Abiko T, Shoelson BD, Bursell SE, Feener EP. Angiotensin AT(1) receptor antagonism normalizes retinal blood flow and acetylcholine-induced vasodilatation in normotensive diabetic rats. Diabetologia. 2004; 47:113-23. [PMID: 14618237]
  126. Johnsen-Soriano S, Garcia-Pous M, Arnal E, Sancho-Tello M, Garcia-Delpech S, Miranda M, Bosch-Morell F, Diaz-Llopis M, Navea A, Romero FJ. Early lipoic acid intake protects retina of diabetic mice. Free Radic Res. 2008; 42:613-7. [PMID: 18608516]
  127. Prusky GT, Alam NM, Beekman S, Douglas RM. Rapid quantification of adult and developing mouse spatial vision using a virtual optomotor system. Invest Ophthalmol Vis Sci. 2004; 45:4611-6. [PMID: 15557474]
  128. Prusky GT, Alam NM, Douglas RM. Enhancement of vision by monocular deprivation in adult mice. J Neurosci. 2006; 26:11554-61. [PMID: 17093076]
  129. Prusky GT, Douglas RM. Characterization of mouse cortical spatial vision. Vision Res. 2004; 44:3411-8. [PMID: 15536009]
  130. Barber A, Schuller K, Bridi M, Bronson S. Visual Acuity and Contrast Sensitivity Are Reduced in the Ins2Akita Diabetic Mouse. ARVO Annual Meeting; 2010 May 2–6; Fort Lauderdale (FL).
  131. de Gooyer TE, Stevenson KA, Humphries P, Simpson DA, Gardiner TA, Stitt AW. Retinopathy is reduced during experimental diabetes in a mouse model of outer retinal degeneration. Invest Ophthalmol Vis Sci. 2006; 47:5561-8. [PMID: 17122149]
  132. Nishikiori N, Osanai M, Chiba H, Kojima T, Mitamura Y, Ohguro H, Sawada N. Glial cell-derived cytokines attenuate the breakdown of vascular integrity in diabetic retinopathy. Diabetes. 2007; 56:1333-40. [PMID: 17470563]
  133. Al-Shabrawey M, Rojas M, Sanders T, Behzadian A, El-Remessy A, Bartoli M, Parpia AK, Liou G, Caldwell RB. Role of NADPH Oxidase in Retinal Vascular Inflammation. Invest Ophthalmol Vis Sci. 2008; 49:3239-44. [PMID: 17470563]
  134. Pfister F, Feng Y, vom Hagen F, Hoffmann S, Molema G, Hillebrands JL, Shani M, Deutsch U, Hammes HP. Pericyte migration: a novel mechanism of pericyte loss in experimental diabetic retinopathy. Diabetes. 2008; 57:2495-502. [PMID: 18559662]