Abstract
Introduction:
The impact of vision debilitating diseases is a global public health concern, which will continue until effective preventative and management protocols are developed. Two retinal diseases responsible for the majority of vision loss in the working age adults and elderly populations are diabetic retinopathy (DR) and age-related macular degeneration (AMD), respectively. Model systems, which recapitulate aspects of human pathology, are valid experimental modalities that have contributed to the identification of signaling pathways involved in disease development and consequently potential therapies.
Areas covered:
The pathology of DR and AMD, which serve as the basis for designing appropriate models of disease, is discussed. The authors also review in vitro and in vivo models of DR and AMD and evaluate the utility of these models in exploratory and pre-clinical studies.
Expert opinion:
The complex nature of non-Mendelian diseases such as DR and AMD has made identification of effective therapeutic treatments challenging. However, the authors believe that while in vivo models are often criticized for not being a ‘perfect’ recapitulation of disease, they have been valuable experimentally when used with consideration of the strengths and limitations of the experimental model selected and have a place in the drug discovery process.
Keywords: age-related macular degeneration, animal models, cell culture models, diabetic retinopathy
1. Introduction
The eye is an intricate and extraordinary organ able to translate visual light into biological signals, which are then interpreted by the brain [1]. Its sophisticated design and function during development, aging, and disease, can be influenced by multiple factors including genetics, environment, and lifestyle, all of which may play an important role in contributing to its complexity at each stage. This is clearly exemplified by retinal diseases, which have the undesirable effect of compromising vision, to varying degrees, including total blindness. Two retinal diseases responsible for a large fraction of new vision loss in working age adults (20–74 years of age) and elderly individuals (>60 years) are diabetic retinopathy (DR) and age-related macular degeneration (AMD), respectively [2,3]. Currently, it is estimated that world-wide there are approximately 126.6 million people affected by DR and an additional 187 million living with AMD [4,5]. Though treatment options are available for DR and AMD patients, they are limited, and efficacy varies significantly amongst patients. This highlights the need for identification of not only new therapeutic targets, but also animal models that present with pathology paralleling that seen in human patients.
Models of retinal diseases have been not only instrumental to identification of potential new signaling pathways involved in initiation and progression of disease, but also serve as platforms to test new therapies. Both in vitro and in vivo models have been successfully used as necessary pre-clinical modalities to test new therapies and have served as the launching point, providing compelling evidence, for initiation of clinical trials [6,7]. In spite of this, frequently, animal models of complex diseases are rejected, as they are perceived as not fully recapitulating the human condition. Arguably, creating the ‘perfect’ model of a complex disease, though laudable, may be improbable, given patients with diseases such as DR or AMD do not express all the risk factors identified to date, nor do they exhibit all the clinical phenotypic features of the disease. Herein we will review recent advances in the development of in vitro models that may be used for high through put screening of drugs as well as in vivo models that develop key phenotypic characteristics of DR and AMD.
2. Diabetic retinopathy (DR)
Currently 300 million individuals have diabetes worldwide and the prevalence is rising rapidly. Over one-third of diabetic individuals will develop DR and approximately 10% of these individuals will suffer from vision threatening disease. DR progresses in many individuals despite preventable measures such as good blood sugar, blood pressure and serum lipid control. DR pathogenesis is multifactorial including leukocyte involvement, basement membrane thickening and pericyte and endothelial loss. Early on there is vasodegeneration of capillaries that leads to retinal ischemia and non-perfusion. In addition to vascular damage the neuronal cells of the retina become compromised even before there is clear evidence of vascular compromise. While the histological and funduscopic features are well-characterized in humans and animals models of the disease, the mechanisms involved remain incompletely understood [8].
3. In vitro tools for modeling DR
Clinical manifestations of DR are microvascular, therefore historically research studies have focused on endothelial and pericyte cell culture models. As it became clear that neuroglia, as well as retinal pigment epithelial cells (RPE) contribute to the development of the disease, DR models of Müller cells, microglia, ganglion cells and RPE cells, as well as co-culture systems were developed and are now commonly used. In addition, cell culture models to determine the role of bone marrow-derived cells in retinal vascular damage and repair are used in DR research (Table 1). Below, each of these models will be discussed.
Table 1.
In vitro models used to study DR.
Assay | Cell type | Read-out |
---|---|---|
Permeability | BREC RPE HREC-pericyte co-culture |
Monolayer permeability in response to VEGF and cytokines [8,74,77,147] Tight junctions structure and function[35,84,85] |
Pro-inflammatory changes | HREC BREC Müller cells Pericytes HREC-pericyte co-culture HREC-Muller cell co-culture RPE Photoreceptors |
Cytokines and adhesion molecule expression and leukocyte adhesion in response to diabetogenic media: • lipids[30,36,42,72,90] • glucose [12,14,27,32,35,47,65,89–94] • cytokines[27,33,36,43,72,73,84–89] High glucose-induced, oxidized glycated LDL and cytokine-induced cytokine and growth factor production, ER stress[102–107]. Response of HREC (adhesion molecules etc.) to pericyte and Muller cell-produced cytokines High glucose and aldose reductase activation and hypoxia-induced cytokine and growth factor production[128–135] Hypoglycemia-induced proinflammatory changes[146] |
Apoptosis | HREC BREC Pericytes RPE |
Diabetogenic media-induced apoptosis [13,20,28,92,97–101] |
Tube formation | HREC HREC and pericyte co-culture HREC and CACs co-culture |
Tube length, width and branching points under control, pro- and anti-angiogenic and diabetogenic conditions[36,254] |
3.1. Retinal Endothelial Cells (REC)
REC isolated from bovine (BREC) [9–26] and human donor eyes (HREC) [21,27–50] are used in a significant number of studies, followed by porcine [23,51,52], rhesus monkey [53,54], feline [55], rat [56] and mouse REC [57,58]. REC cultures from bovine and human eyes are obtained by retinal digestion and filter separation of the microvessels followed by homogenization, digestion and further purification by gradient centrifugation, antibody-based purification [45], or selective culture conditions optimized for each species [34,59,60]. Porcine [23,51,52], rhesus monkey [53,54], feline [55] and rat [61] REC are isolated and cultured similar to bovine and human, however mouse cultures are notoriously difficult potentially due to different mouse osmolarity and sparse amount of starting material. The only mouse endothelial cell culture reported to date is from platelet endothelial cell adhesion molecule-1 (PECAM-1) antibody based purification of retinas isolated from immortomice [58].
To study the effect of diabetes on REC several experimental paradigms are used. Most of them involve treatment of normal control REC with diabetogenic media. The composition of diabetogenic media varies depending on the study and could include high glucose [27,46,62–66], saturated and omega 6 polyunsaturated fatty acids [67–71], ceramide, cholesterol [68,72], cytokines [42,50,68,72,73] and vascular endothelial growth factor (VEGF) [9,74] treatments. Exposure of endothelial cells to hypoxic conditions [75], or treatment with CoCl2 to induce pseudohypoxia [76] are also used. Recently the phenomenon of metabolic memory, or legacy effect was discovered and it was demonstrated that HREC isolated from diabetic patients or animal models maintain the diabetic phenotype for several passages [34,36,77–80]. HREC isolated from control and diabetic donors have also become a valuable model to examine the effects of diabetes as a whole on REC damage and repair [34,36].
Using these experimental paradigms several endpoints for the effects of diabetes and diabetogenic media on REC are tested.
Permeability
Retinal microvascular endothelial cells are part of the blood-retinal barrier and breakdown of this barrier with an increase in retinal vascular permeability are amongst the earliest diabetes-induced changes in DR. It is thus important to have reliable cell culture models to understand the effects of the diabetic metabolic abnormalities on endothelial cell barrier function. The most well characterized model for these studies are BREC [9,74]. BREC form uniform monolayers connected by tight junctions with electrical resistance above 200Ω/cm2. They also respond to VEGF and cytokines with increased permeability [9,74,81]. This model has been used to demonstrate the role of pro-inflammatory and pro-angiogenic factors in diabetes-induced tight junction abnormalities. Additionally, though HREC monolayers are leaky as they do not form a tight barrier, they can be used to determine the molecular mechanisms of the effects of diabetes on tight junction proteins [35,82,83].
Pro-inflammatory changes
Inflammation and increase in inflammatory cytokines are well accepted to play an important role in the pathogenesis of DR. Treatment of BREC and HREC with cytokines known to be increased in the diabetic retina, IL-1β and TNFα, followed by NFκB pathway activation leads to an increase in adhesion molecules ICAM-1 (intercellular) and VCAM-1 (vascular), and leukocyte adhesion [27,33,36,43,72,73,84–89]. Similar effects have also been shown after the exposure to pro-inflammatory omega 6 fatty acids [30]. These effects may be blocked by decreasing the levels of cholesterol or ceramide in the endothelial cell membrane [30,36,42,72,90]. There is a lot of controversy in the literature on the role of high glucose in controlling REC inflammatory state. Several studies have demonstrated that high glucose alone is a very weak stimulus for pure endothelial cells and that the high glucose response of endothelial cells is driven by other cell types normally present at lower levels in endothelial cell cultures, such as microglia and Müller cells [27,30]. However, most of the studies show a slight upregulation of pro-inflammatory, pro-apoptotic factors, and mitochondrial damage after prolonged exposure to high glucose [12,14,27,32,35,47,65,91–96].
Apoptosis
Endothelial cell apoptosis leads to formation of acellular capillaries, the hallmark of DR, in the retina. The role of high glucose, cytokines, fatty acids, cholesterol, sphingolipids, oxidative stress and mitochondrial damage is widely studied using BREC and HREC cultures. Endothelial cells are shown to have both intrinsic and extrinsic apoptotic pathways with activation of cytochrome C release from mitochondria and Caspase 9, Caspase 8 and FLIP pathways leading to classical Caspase 3/7 effector caspases activation and execution of apoptosis [13,20,28,92,97–101].
Tube formation
Tube formation assay is used to access both normal migratory, proliferative and repair capacity of REC, as well as REC response to neovascular stimuli. HREC isolated from diabetic donor tissue, as well as HREC and BREC treated with diabetogenic media have been shown to have defective tube formation that could be improved by inhibition of inflammatory or apoptotic factors, or introduction of reparative progenitor cells as will be discussed later [36].
3.2. Pericytes
Pericyte dropout is one of the earliest features of DR and the role of pericytes in support of retinal microvascular endothelial cell barrier through both mechanical connection as well as secretion of paracrine factors is well accepted. Pericytes isolated from human, bovine and murine tissue are widely used in DR pathway and drug discovery research. Pericytes isolated from control and diabetic tissue, as well as treated with high glucose, oxidized glycated low density lipoproteins (LDL), and cytokines are used to study the role of autophagy and ER stress in pericyte apoptosis, and paracrine factors secreted by pericytes, including ATP release and activation of P2X7R [102–108]. In addition, the role of chemomechanical contraction of pericytes in endothelial cell function can be assessed using pericytes isolated from control and diabetic donor tissue, as well as pericytes treated with high glucose, lipids and cytokines [109].
3.3. Müller cells
Müller cells are the primary glia cells in the retina that span the whole thickness of the retina and, like pericytes, have direct contact with endothelial cells, as well as neuroretina. Loss of Müller cells protective paracrine factors and increase of pro-inflammatory cytokine production by Müller cells in diabetes is studied using several cell culture models, including Müller cells isolated from human donor tissue, as well as human and rat Müller cell lines. Using these models, the role of hyperglycemia, oxidized LDL, and K channels in cytokine and VEGF production has been evaluated [110–122].
3.4. Microglia
Microglial activation occurs early in DR and is proposed to contribute to the pro-inflammatory status and pathogenesis of DR [36,123,124]. There are very few studies using microglia cell culture models of DR due to several factors. First of all, the markers that are used to identify and isolate microglia (Iba-1, CD11b, CD68) are shared between microglia and other monocytes making it challenging to separate pure populations of resident microglia from extravasated immune cells. Moreover, newborn murine retinas are required for viable cultures of microglia making it very difficult to obtain enough starting retinal material for experimental treatments [125].
3.5. RPE cells
The retinal pigment epithelial layer is known for its role in diabetic macular edema, as loss of pigment epithelial-derived factor (PEDF), a trophic factor produced by RPE cells, has been shown to contribute to pericyte loss and endothelial damage in DR. Thus RPE cell cultures are important tools in DR research. It is advantageous that human, bovine, porcine and murine RPE cells are relatively easy to isolate and grow in culture. Additionally, although not without important limitations, the spontaneously immortalized RPE cell line, ARPE-19 [126], has been used extensively in RPE cell research. This RPE cell model has been reported to have low barrier function due to rudimentary tight junctions and variable expression of claudins [127]. However, it maintains a number of characteristics of human RPE cells, not all, is easy to grow in culture, and provides a useful and convenient model for DR and macula edema research.
Multiple studies have examined the effect of metabolic factors, including high glucose and lipids on glucose transport and glucose metabolism in RPE cells [128–135]. RPE cells in culture have been shown to have high levels of aldose reductase [136]. High glucose conditions lead to an increase in aldose reductase activity in RPE cells [136], and inhibition of aldose reductase prevented oxidative stress, cytokine production and apoptosis in high glucose treated RPE cells [136]. The utility of RPE cells has further been demonstrated in studies of PEDF, which have confirmed the role of diabetogenic conditions in inhibition of the production of this trophic factor using RPE cell culture models [128,137,138].
3.6. Ganglion cells
Ganglion cell death is a well accepted part of DR pathogenesis and production of VEGF by ganglion cells in response to succinate has been shown to contribute to inner blood retina barrier (BRB) breakdown in DR. Early papers used retinal ganglion cell line 5 (RGC5) to study the effects of hyperglycemia on ganglion cells, however the origin of RGC5 cells was later questioned and studies on this cell line were not verified [139]. Ganglion cells isolated from embryonic rat retinas by panning protocol are viable and can be obtained at high purity [140]. These cells have been used to determine the role of TNFα [141], PEGF [142], metabolic activity [143], and sigma receptor 1 [144] as a model of diabetes-induced damage.
3.7. Photoreceptor cells
Recent studies have revealed the contribution of photoreceptor damage to DR pathogenesis. Although there are several models for “shaving” or isolating photoreceptors, they are notoriously difficult, and the cells do not grow in culture. Most of the cell culture studies rely on a mouse SV-40 T antigen transformed photoreceptor cell line 661W [145]. The effect of hypoglycemia on photoreceptor cells has been tested using this model [146].
3.8. Co-culture studies
As the retina is a complex organ, most of the effects involve multiple cell types. To capture this complexity several co-culture models have been applied. These include media transfer models where pericytes, Müller or RPE cells are cultured in high glucose or lipid mediators and the media with the factors produced by these cells is used to treat endothelial cells. Co-culture of pericytes and endothelial cells has also been used to test the role of cell-cell interactions in fluctuating high/low glucose-induced breakdown on permeability barrier [147] and to test chemomechanical contraction of pericytes in endothelial cell proliferation [109,148].
In addition to retinal cells, bone marrow-derived circulating angiogenic cells (CACs) are known to both incorporate into the vessels and produce paracrine factors that aid in endothelial repair. Co-culture models of HREC isolated from control and diabetic patients combined with CD34+ CACs isolated from control and diabetic donors may be used to develop the strategies for improved cell-based therapy for DR [36].
4. In vivo models of DR
There are many animal models of diabetes. While it is well known that rodent models develop only the early stages of DR, they still remain the most popular models. This is largely because of their cost and well-described histological and functional characteristics. In addition to the economic considerations, rodent models have the benefit of their short generation time. In this section, selected and highly popular models will be briefly described with the specific intent of describing the histological and functional features of the model that can be reliably used to validate a pharmacological intervention, taking into account the models advantages/disadvantages. From the perspective of pre-clinical efficacy, a model must have key DR endpoints and that the “endpoints” have a reproducible time line. Specific models include type 1 diabetes (T1D) and type 2 diabetes (T2D) models and models that have “mixed” features such as the type-2 diabetes in rats that is induced by a high fat diet and streptozotocin (STZ).
Regarding the endpoints, perhaps the “classic” gold standard of DR is enumeration of acellular capillaries. This histological endpoint is representative of vasodegeneration and considers both the loss of pericytes and endothelial cells. While the pathology implicit in this histological endpoint is the culmination of many deleterious events including inflammatory cell activation, enhanced cytokine expression, oxidative injury and many more. Acellular capillaries are the “hallmark” feature of DR. Prior to popular use of acellular capillaries, the enumeration of pericyte loss (pericyte ghosts) was also considered an early and reproducible event in DR. Similarly, vascular permeability is a critical and assessable endpoint for DR [149]. A model that recapitulates many of the histological features of DR is the model of ischemia/reperfusion [150]. This model has acellular capillaries and increases in permeability (Figure 1). Although ischemia/reperfusion is different from diabetes-induced pathogenesis as it does not involve hyperglycemic environment, the endpoints of this easy economical model provide a fast way to evaluate retinal vascular changes and to test interventions.
Figure 1. Histology of retinal vascular permeability and acellular capillaries in diabetic rodents.
Vascular permeability in (A) wild-type control versus (B) STZ-induced diabetic mouse after 8 weeks of diabetes. Acellular capillaries are rare in control C57BL mice (C) or Wistar rats (E), but evident 9 months after STZ induction of diabetes (D-red arrows) or 7 days after ischemia-reperfusion (F-black arrows). This panel includes original images.
Diabetes models are generated typically through drugs, diet, or chemical damage. Genetic models are produced by selective breeding and gene editing. Mice have been the emphasis of most genetic studies, with the discovery of inherited obesity or hyperglycemia resulting in a diabetic phenotype [151–153]. However, other species have advances over rodent DR models. As put forth by Engerman, dog models appear to be most similar to human DR [154]. Pigs are preferred for the similarity of their eye structure to humans and Zebra fish for their short life span and large breeding sizes [155]. Unfortunately, nonhuman primates have proven relatively resistant to induced DR [156]. Although no single animal model represents the complete range of vascular and neural complications of human DR from both early and late stages, the murine models described below and summarized in Table 2 have been instrumental in establishing mechanisms responsible for DR and have provided reproducible pre-clinical data.
Table 2.
Animal models that exhibit DR pathology.
DR Pathology | Animal Model |
---|---|
Acellular capillaries | STZ induced T1D [158] db/db [169,170] Galactose [163] |
Pericyte ghosts [149] | STZ induced T1D [158] db/db [169,170] Galactose [163] Alloxan [157] |
Microglial activation | STZ induced T1D [158] Alloxan [157] |
Loss of RGCs, INL and ONL thinning | STZ induced T1D [158] Ins2Akita[166,167] Alloxan [157] |
Microaneurysms | Galactose fed mice |
Astrocytes/gliosis | STZ; galactose [163] Ins2Akita [166] |
Permeability | STZ induced T1D [158] db/db [169,170] Galactose[163] Ins2Akita [167] |
Leukocytosis | STZ induced T1D [158] db/db [169,170] Galactose [163] Ins2Akita [167] |
Capillary basement thickening | NOD mice [167] STZ induced T1D [158] |
RGC=retinal ganglion cells; INL=inner nuclear layer; ONL=outer nuclear layer; STZ=streptozotocin; NOD=non-obese diabetic; T1D=type 1 diabe
4.1. Drug-induced models of DR
Alloxan has been used to induce DR in a large variety of animals including mice. Alloxan causes damage to pancreatic β cells and when given to mice 8–10 weeks of age can result in hyperglycemia following a single injection. Alloxan induces pericyte ghosts and loss of RGCs within 7 days and microaneurysms with increased acellular capillaries by 21 days in mice from the FOT_FB strain [157] and microglial changes, with thicker cell bodies and shorter dendrites by 3 months of age [123].
STZ administration is the most commonly used drug to initiate diabetes and causes disruption of pancreatic beta cells and leads to hyperglycemia. Hyperglycemia onset typically occurs within 2 weeks, regardless of dosage and can be maintained for up to 22 months [158]. DR phenotypes observed in STZ mice and rats [147,159] include increased numbers of astrocytes and gliosis 4–5 weeks after onset of hyperglycemia [158], RGC loss at 6 weeks, permeability at 8 weeks (Figure 1) retinal inner nuclear layer (INL) and outer nuclear layer (ONL) thinning at 10 weeks and acellular capillaries and pericyte ghosts at 6–9 months (Figure 1) [160]. Additional changes such as increased mRNA expression of adhesion molecules have been observed as early as one week after the induction of diabetes [161,162].
Kern and Engerman first reported an animal model of DR induced by a galactose-heavy diet [163]. Mice developed hyperglycemia by 6 weeks of age. After 15 months of hyperglycemia, endothelial cell loss and increased acellular capillaries were observed. After 21 months, lesions including pericyte ghosts, microaneurysms, and retinal thickening were observed. While retinopathy takes longer to develop in these mice, the mice live longer than other models, allowing them to be observed over a longer period of time, up to 26 months [164]. Similarly, rats have been kept on high-galactose diets for over 2 years. However, allaxon, the STZ or galactose models do not result in retinal neovascularization.
4.2. Models of neovascularization
Typically, the “non diabetic” oxygen induced retinopathy (OIR) [165] is utilized to assess anti-angiogenic agents for potential translation to the clinics. The OIR model exploits that mice are born with only a partially developed vasculature and manipulation of oxygen levels post natally (postnatal days P7-P12) can influence vascular behavior, and as such are a model of retinal development. In normal mouse pups, formation of the deep vascular plexus (DVP) is initiated on P8; however, DVP development does not occur when pups are placed into high oxygen, but rather formation of the DVP is initiated several days after return to normal room air around P15. The classical endpoints that are evaluated are central vaso-obliteration and retinal neovascularization. Both vaso-oblieration and neovascularization can be evaluated on retinal flat mounts. Retinal crossections are also used for quantitation of neovascularization using serial sections of whole eyes cut sagittally. Typically, between two and four sections on each side of the optic nerve, 30 to 90 nm apart are counted for neovascularization as described by Smith et al [165]. Vascular cell nuclei, identified under light microscopy with hematoxylin staining typically are considered to be associated with new vessels and if found on the vitreal side of the internal limiting membrane are counted. Thus the OIR model provides a quantitative measurement and may demonstrate that a drug is particularly robust if it shows a significant reduction in neovascularization when performed as a dose-response study.
4.3. Genetic models of DR
Genetic mouse models of DR include the Ins2Akita, non-obese diabetic (NOD), db/db (Leprdb), Kimba, and Akimba, which vary in mode of inheritance, disease etiology, pathology, and progression of disease. The Ins2Akita mouse is a model for T1D that arises as a result of a missense mutation in the Insulin 2 gene that leads to a conformational change in the insulin protein, causing the protein to accumulate in pancreatic β cells, leading to β-cell death. Diabetes onset is around 8 weeks with an increase in retinal vascular permeability and reactive gliosis. Disease progression continues up to 8 months of age, with a reduction in axons and dendrites of RGCs by 12 weeks, an increase in acellular capillaries at 36 weeks, and an increase in leukocytes in the vascular wall, as a result of inflammation. Decreased numbers of cholinergic and dopaminergic amacrine cells are observed leading to a reduction in the thickness of the IPL and INL [166]. The Ins2Akita mouse is useful for studying the early progression of DR and the neuroprotective effects of treatments, as loss of RGCs can be detected in a short span of time [167].
Another model of T1D is the NOD mouse that shows apoptosis of pericytes, endothelial cells, and RGCs, as well as retinal capillary basement membrane thickening starting as early as 4 weeks. Vasoconstriction and degeneration of major vessels with abnormal microvessels can be detected approximately 4 months after hyperglycemia. However, NOD mice exhibit a gender bias towards females becoming diabetic [168].
A well-established T2D is the db/db (Leprdb) mouse that is deficient in the leptin receptor and spontaneously develops diabetes associated with obesity at 4–8 weeks of age. Six-month-old db/db mice have been shown to exhibit early features of DR, such as pericyte and endothelial cell loss, BM thickening [169] and increased blood flow in the retina. By 15 months, these mice demonstrated BRB breakdown, pericyte loss, neuroretinal apoptosis, glial reactivation, and acellular capillaries in the retina [170].
Similar to mouse models, there are several spontaneously diabetic rat models, the Zucker diabetic fatty rat, the WBN/K0b rat, the Otsuka Long-evans Tokushima fatty rat, the Goto-Kakizaki rat, the Torii rat [167] and the BBZ/Wor [171]. Each of these models recapitulates the major phenotypic characteristics of DR and are reviewed in detail in Robinson et al [167].
5. Age-related macular degeneration (AMD)
AMD is the leading cause of irreversible vision loss in individuals over 65 years of age. It is estimated that over 187 million people worldwide are affected by the disease [172,173]. It is characterized by a progressive and chronic degeneration of the macula, a 5.5 mm diameter region central in the retina, responsible for high acuity vision. Tissues and cells, which are affected and thus ‘vulnerable’ in AMD include, photoreceptors, RPE, Bruch’s membrane, and the outer blood supply of the eye including the choriocapillaries and choroidal endothelial cells [174]. Clinically, the pathogenesis of early dry AMD involves accumulation of lipid and protein rich extracellular deposits (e.g. sub-RPE deposits, basal laminar and linear deposits, drusen), below the RPE, the cells that support the overlying neural retina (Figure 2) [175–180]. As the disease progresses to the advanced stages it can manifest as either geographic atrophy/”late dry”, or exudative/”wet” AMD. Geographic atrophy is characterized by areas of RPE atrophy that can coalesce to become larger, along with loss of photoreceptor cells in the central macula, leading to vision loss [174,181]. Neovascular AMD is characterized by abnormal growth of choroidal blood vessels through Bruch’s membrane, generally confined below the RPE and/or retina, though it can also penetrate beyond the sub-retinal space and within the retina, progressing to becoming retinal angiomatous proliferation. These ‘new’ vessels are leaky and proliferative in nature and eventually may lead to fibrosis and scarring, contributing to significant vision loss [181,182]. Currently, there are no effective therapies available for treatment of early dry AMD or geographic atrophy, and the anti-VEGF strategy commonly used in clinics to treat “wet” AMD has been shown to be effective in only a subset of patients [183]. This highlights the critical unmet need of developing more effective therapeutic strategies for the treatment of all clinical sub-types of AMD.
Figure 2. Pathology of sub-RPE deposits in human AMD samples.
Transmission electron microscopy images reveal details of the complexity of sub-RPE deposits including the presence of basal lamina deposits (A-red double arrowhead), fibrous long spacing collagen (B-red arrow), and membranous debris (C-green double arrowhead). This panel includes original images.
6. In vitro modeling of AMD
Cell-based modeling of chronic neurodegenerative diseases of aging such as AMD is challenging, as it requires patience and time. One concern often raised, is that the use of just one or even two cell types in a culture dish over simplifies pathogenic mechanisms that would occur in an otherwise complex tissue. However, this may in and of itself be an advantage as it allows for investigating the response of isolated cells under various conditions and challenges, which may reveal important insights into disease mechanisms. Furthermore, if standardized, cell cultures may serve as a platform for high throughput screening of pharmacological agents as potential therapies. RPE cells derived from human eyes have been used most frequently for modeling AMD phenotypic features and specifically with the goal of developing sub-RPE deposits such as drusen, the key pathology in early dry AMD, in a culture dish (Table 3). Recent successes include the culturing of fetal human RPE cells on to porous supports, which over time produce extracellular deposits containing apolipoprotein E (apoE) and complement proteins, molecules also identified in human drusen [184]. The presence of components of drusen including apoE, hydroxyapatite and neutral lipids have also been observed below porcine RPE cells cultured for up to 6 months on uncoated transwells [185]. Finally, patient-derived human-induced pluripotent stem cell RPE derived from patients with the dominant macular degenerations, Sorsby’s fundus dystrophy, Doyne honeycomb retinal dystrophy and malattia Leventinese, have been used as a resource for modeling sub-RPE deposit formation in vitro [186]. Of significance, cultures from patients with macular degenerations developed a greater number of deposits, with a different composition, when compared to normal aged individuals, highlighting the potential value of using these model systems to identify disease specific mechanisms that lead to drusen formation.
Table 3.
In vitro models used to study AMD.
In vitro models | Advantages | Disadvantages |
---|---|---|
Human RPE from adult eyes | • Easy to isolate and culture from post mortem eyes. • Can be cryopreserved and cultured. • Can be used to study molecular pathways implicated in AMD, e.g., lipid transport, metabolism, etc. • Can be used to study the effect of various insults, e.g cigarette smoke, oxidized lipids, etc. • Amenable to transfection (siRNA and plasmid) |
• Lacks tissue complexity; e.g. choroid and photoreceptors. • Lacks immune system components. • Sub-RPE deposits and known constituents of drusen are generally absent. • Loss of RPE characteristics with passage. |
Human RPE from fetal eyes | • Easy to grow on porous support. • Can be cryopreserved and cultured. • Form sub-RPE structures which stain positive for known drusen components including apoE, vitronectin, clusterin, amyloid, etc. • Can be used to study the role of component system as apoE positive deposits co-localize with component system molecules in the presence of serum in growth conditions. • Good platform to test the effect of drugs on deposit formation as well as complement activation. |
• Lacks tissue complexity; e.g. choroid and photoreceptors. • It takes about 1-3 months for deposit formation. |
iPSC-RPE derived from human | • Provides a suitable platform to investigate the RPE-autonomous molecular events. • Can be used to study patient specific effects like age and genetics (point mutations or polymorphisms) on drusen formation and response to therapy. • Conducive to genome editing by CRISPR-Cas9 system. • Starting material is convenient to obtain, e.g. skin biopsies, blood. |
• Lacks tissue complexity; e.g. choroid and photoreceptors. • Reprogramming is an expensive and time-consuming process. • Extensive characterization is required at each step of dedifferentiation and differentiation. |
Porcine RPE cells | • More readily available than human post mortem eyes. • Cultures produce focal and diffuse sub-RPE deposits similar to the ones seen in human AMD macula. • Can be used to test the effect of drugs on deposit formation |
• Lacks tissue complexity; e.g. choroid and photoreceptors. • Long culture time |
7. In vivo modeling of AMD
Animal models have proven to be invaluable in investigating the pathobiology of an aging disease as well as in testing of new therapies. An ideal model of AMD would be inexpensive, recreate the histological and functional changes, but progress in a rapid time course to allow for effective studies. Developing an efficient model of AMD that mimics both the early and late characteristics of AMD has been challenging due to several hurdles. First, development of AMD is a complex process, which involves genetic and environmental factors. Not only have numerous genetic polymorphisms and loci been reported to contribute to increased risk for AMD, but multiple biological pathways have also been shown to be dysregulated in the pathogenesis of AMD [175]. These include but are not limited to, oxidative stress, inflammation, angiogenesis, lipid and carbohydrate metabolism. Second, is the issue of anatomical differences between the species, in which the models have been developed, and the human retina. Despite these obstacles, models of AMD have been created in mice, rats, rabbits, pigs and non-human primates, which collectively have led to the discovery of multiple facets of the pathology of the disease [187–193]. Rodents are advantageous due to their relatively short life-span, low cost and rapid disease progression. Importantly, they are amenable to genetic manipulation. However, as mentioned, both mice and rats lack a distinct anatomical macula and fovea, unlike humans in which the characterizing features of the disease are defined as occurring in the cone-rich macula. Non-human primates, on the other hand, form the other end of spectrum. They are closest to humans in terms of the anatomy of the macula but are expensive to maintain, difficult to manipulate genetically, and have a slow disease progression.
In general, the goal in developing an animal model of AMD is to recreate classical histological features, which have been reported from the eyes of patients with AMD. These include sub-RPE basal laminar deposits and basal linear deposits, and drusen; thickening of Bruch’s membrane, RPE atrophy, accumulation of immune cells in the inner and outer retina, photoreceptor atrophy, choriocapillary atrophy, choroidal neovascularization and fibrosis (Figure 2 and 3) [194]. Decrease in functional activity as measured by electroretinography (ERG), is also observed in AMD patients and a desired outcome [195]. We will review several select animal models, which were developed following manipulation of genetic or environmental factors impacting reported AMD pathogenic pathways, and highlight the disease relevant pathology they exhibit (Table 4).
Figure 3. Abundance of immune cells in human AMD.
Cryosections from human donor eyes were probed with antibodies to Iba1, which stains microglia and macrophages (green), SV2, a synaptic vesicle marker (red) and Hoechst, which highlights cell nuclei (blue). Iba1 positive cells are absent from the retina of an aged donor without eye pathology (A), but present in the retina of a patient with wet AMD both in regions where CNV is absent (B) and present (C). Furthermore, an abundance of Iba1 positive cells are seen throughout the CNV lesion itself (C-CNV lesion boarder is indicated by yellow broken line). This panel includes original images.
Table 4.
Testing of potential therapies on various AMD pathologies in mice.
Models of AMD Pathologies | Candidate drugs | Effect on AMD pathogenesis | References |
---|---|---|---|
Oxidative stress | |||
Sod−/− | α-Lipoic acid | Increase in serum SOD | [255] |
AhR−/− | AhR agonists | Ameliorate CNV formation | Unpublished |
Immune regulation | |||
Ccl2/Cx3cr1−/− |
Nutrient supplementation with lutein and zeaxanthin Naloxone |
Lesion regression Lower VEGF, TNFα, iNos, Cox2 expression in retina Lower number of microglia in outer retina |
[256] [257] |
Ccr2−/− | TNFα-stimulated gene/protein (TSG-6) | Lowered VEGF expression Lower infiltration of CCR2+ cells in CNV lesions |
[258] |
Complement pathway | IBI302 Eculizumab Anti-factor D (lampalizumab) |
Novel bispecific decoy receptor fusion protein with a VEGF inhibition domain and a complement cascade inhibition domain Binds C3b/C4b Anti-mouse complement component 5 antibody Inhibit CNV Inhibit infiltration of F4/80-positive cells into CNV lesions Humanized IgG Fab fragment directed against factor D Inhibit systemic Alternative Complement activity |
[259] [260] [261] |
Lipid transport and metabolism | |||
Pparβ/δ−/− | GSK0660 | Ameliorate CNV formation Regulate extracellular matrix deposition in CNV lesions |
[234] |
AhR=aryl hydrocarbon receptor; SOD=superoxide dismutase; CNV=choroidal neovascularization; VEGF=vascular endothelial growth factor; TNFa=tumor necrosis factor alpha; iNos=inducible nitric oxide synthase; Cox2=cytochrome C oxidase; Ccl2=CC chemokine ligand 2; Ccr2=cc chemokine receptor; PPAR=peroxisome proliferator activated receptor.
Oxidative injury
The retina is susceptible to oxidative damage due to its high metabolic demand, high concentration of oxidation prone polyunsaturated fatty acids and photosensitive molecules such as rhodopsin and lipofuscin, which can produce reactive oxygen species when exposed to light [196]. Oxidative damage from sunlight or cigarette smoking has been shown to be AMD risk factors in several epidemiological studies [197–199]. These observations have been the impetus to the development of animal models by using mice that lack intrinsic anti-oxidant mechanisms or exposing animals to various oxidant insults.
Super oxide dismutase (SOD) is a powerful anti-oxidant and is expressed in three forms, SOD1, SOD2 and SOD3. SOD1, is primarily cytosolic and is highly expressed in retina, SOD2 and SOD3, localize primarily to the mitochondria and extracellular space, respectively [200]. Sod1−/− mice, after 7 months of age display ‘drusen’-like deposits between the RPE and Bruch’s membrane. These deposits express biomarkers of drusen including: vitronectin, carboxymethyl lysine and tissue inhibitor metalloproteinase 3 [201,202]. Light exposure in these mice can accelerate the appearance of ‘drusen’-like deposits in a dose dependent manner. Interestingly, 10% of aged Sod1−/− mice display evidence of spontaneous CNV by histology and fluorescein imaging, making this model one of the few to date to develop features of both dry and wet AMD. Sod1−/− mice also exhibit progressive visual decline, reflected by a decrease in their a- and b- wave amplitudes and corroborated by an increase in the occurrence of necrotic cell death in the INL as compared to wild-type mice [203]. On the other hand, Sod2−/− mice, die soon after birth due to dilated cardiomyopathy, which limits their use as a model. This is unfortunate as polymorphisms in Sod2 gene have been associated with increased risk of AMD development [204,205]. To counter this, mice in which their RPE and retina have been transfected with an adenovirus expressing a ribozyme capable of degrading Sod2, have been produced. These mice exhibit increased expression of markers of oxidative damage, thickening of Bruch’s membrane, RPE degeneration, increased autofluorescence and photoreceptor atrophy, all characteristic features of dry AMD [206].
Cigarette smoking, second to aging, is a significant preventable environmental risk factor for the development of AMD as cigarette smoke contains over 5000 potential harmful compounds including many pro-oxidants. The role of cigarette smoke to induce or exacerbate AMD has been investigated in several animal models, both in isolation as well as in combination with a high fat diet and blue light exposure [207–210]. In these models, it has been demonstrated that when mice are stressed with oxidants including cigarette smoke, blue light, and/or high fat diet, they develop a thickened Bruch’s membrane and basal laminar deposits, making them good models for investigating either how oxidants contribute to basal deposit formation or testing therapies to target deposit formation. The potential role of cigarette smoke and xenobiotic metabolism in AMD pathology has further been corroborated by investigating the phenotype of the aryl hydrocarbon receptor (AhR) knock out mice (Ahr−/−). AhR is a transcription factor responsible for xenobiotic metabolism and detoxification and can be activated by components of cigarette smoke. Furthermore, AhR activity in human RPE cells decreases with increasing age, suggesting a decline in the ability of RPE cells to effectively clear out toxins through this pathway [211]. Ahr−/− mice exhibit decreased visual function and develop several phenotypic features of dry AMD including disrupted RPE cell junctions, lipid- and collagen-rich basal deposits, Bruch’s membrane thickening and progressive choroidal atrophy (Figure 4) [211,212]. Interestingly, aged Ahr−/− mice are also susceptible to developing large and complex neovascular lesions following laser injury in comparison to age-matched wild-type mice [213]. These studies have revealed a novel pathway contributing to development of AMD pathology and support investigating the role of AhR agonists and specifically acute activation of the AhR as a potential therapeutic strategy for early and dry AMD.
Figure 4. Pathology of sub-RPE deposits in a mouse model.
Aged AhR knockout mice develop clinical phenotypic features of dry AMD including accumulation of neutral lipids with in sub-RPE deposits as evident by staining of cryosections with oil red o (A-wildtype, B and C-AhR knockouts). Transmission electron microscopy reveals that while wild-type mice do not develop sub-RPE deposits (D), thick and continuous deposits occure frequently in AhR knockout mice (E, F-red double arrowhead). This panel includes original images
Lipid transport and metabolism
Several parallels have been drawn between the deposition of cholesterol and lipids in atherosclerotic plaques and the deposits that accumulate under the RPE and within Bruch’s membrane [173,214–216]. Lipids and cholesterol accumulate within Bruch’s membrane with aging and their hydrophobic nature may interfere with the conventional passage of metabolites between the RPE and the outer retinal blood supply [216–218]. They have also been reported to contribute to accumulation of basal laminar and basal linear deposits [219]. Finally, a higher consumption of cholesterol, monosaturated, polyunsaturated and saturated fats; and genetic polymorphisms in apolipoproteins, which regulate lipid transport, have also been shown to be risks associated with AMD development [220–223]. These observations have served as the inspiration for the creation of various animal models, which explore the relationship between lipid transport dysregulation and AMD.
Basal laminar deposit accumulation has been demonstrated to occur in old (16 months) C57BL/6 mice fed high-fat diets [207]. In combination with an additional stressor such as exposure to blue-green light and/or estrogen depletion, the number and severity of deposits has been shown to increase, supporting the role of diet as a risk for AMD development [207,224]. Apolipoprotein molecules, which mediate lipid and cholesterol transport, are found in drusen and as such, the retinal morphology of apolipoprotein knockout mice have been examined extensively. For example, apoE deficient mice develop high levels of circulating cholesterol [225] and histological evaluation of the retinas of 8 month old ApoE−/− mice has revealed they develop a thicker Bruch’s membrane containing membranous material when compared to age-matched controls [226]. The retinal morphology of transgenic animals where the mouse ApoE was replaced with different human APOE alleles has also shed light on their role in AMD pathology development [227–229]. For example, aged APOE4 targeted replacement mice, when fed a high fat diet over 8 months, develop Bruch’s membrane thickening, RPE pigmentary changes, and sub-RPE deposits. A small percentage of these aged APOE4 mice also displayed areas of CNV and photoreceptor loss. Another apolipoprotein found in abundance in drusen is apolipoprotein B100, a major component of low-density lipoprotein (LDL) cholesterol-carriers [230]. Transgenic mice expressing human APOB100, when fed a high fat diet and exposed to oxidative blue-green light have been shown to develop basal laminar deposits [231], a phenotype that is exacerbated with age resulting in loss of RPE basal infoldings, basal laminar and linear deposits and Bruch’s membrane thickening [232,233].
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ), is a ligand-activated transcription factor, which is in involved in the regulation of lipid metabolism, extracellular matrix remodeling and inflammation, pathways also associated with AMD development. Aged PPARβ/δ−/− mice have been shown to develop several features of dry AMD including, Bruch’s membrane thickening, discontinuous sub-RPE deposits, loss of RPE basal infoldings, RPE degenerative changes and lipofuscin accumulation when compared to age matched wild-type mice [234]. Interestingly, absence of the PPARβ/δ resulted in less severe choroidal neovascular lesions forming following laser injury, in comparison to mice expressing the receptor. Importantly, treatment with PPARβ/δ antagonist demonstrated its therapeutic potential in the experimental mouse model of CNV [234]. Collectively, these studies highlight the importance of studying regulators of lipid metabolism in the pathobiology of AMD. Furthermore, the models discussed are suitable for further in-depth investigation of potential mechanisms contributing to- and/or development of candidate therapeutic strategies that specifically target RPE degenerative changes, sub-RPE development or CNV formation, with consideration of a patient’s dietary habits or systemic lipid levels.
Immune dysregulation
Several genome-wide association studies have established a role of immune system dysregulation in AMD progression. These studies revealed an association between single nucleotide polymorphisms (SNPs) encoding components of the immune system and increased risk for developing advanced AMD [235–239]. Additionally, multiple histological studies examining human donor tissue have identified various components of the immune system within drusen, the choriocapillaries and choroid [240].
Complement factor H (CFH) was the first complement gene associated with increased risk of developing AMD, with polymorphisms including Y402H, increasing the risk of advanced disease development [236–239]. Additionally, inheritance of Y402H has been reported to confer resistance to anti-VEGF and thus has potentially negative implications for treatment outcomes in patients with CNV [241,242]. CFH acts to regulate the alternative complement pathway and several CFH-based mouse models have been generated to study its impact on AMD pathology. Cfh−/− mice display retinal dysfunction, RPE autofluorescence and progressive C3 deposition [243,244]. By contrast, CfhY402H transgenic mice, which express the human Y402H variant in the RPE and liver, driven by the apoE promoter, exhibit basal laminar deposits and accumulation of lipofuscin-like material in the RPE [245]. Interestingly, this phenotype is accompanied by accumulation of macrophages and immune cells in the outer retina. Additionally, comparison of pathology in aged Cfh−/− versus Cfh+/− mice when challenged with a high fat diet, has revealed greater RPE damage and visual function impairment in mice with decreased levels of Cfh rather than complete absence of the gene [246]. In a follow-up study it has been shown that mice that express the full length human CFH when crossed with Cfh−/− mice demonstrate improved visual function, supporting the therapeutic potential of increasing CFH levels [247]. Similarly, low levels of CFH have been associated with development of a more severe laser induced CNV lesion in mice [248,249]. Collectively, these studies have begun to shed light on the complexity of the complement pathway in the development of early dry AMD. It is important to note, that alterations in CFH levels have not resulted in spontaneous expression of features of either GA or CNV in mice, perhaps reflecting differences in immune regulation between mice and humans.
Chemokines or their receptors have also been associated with an increased risk of AMD. Monocyte chemoattractant protein 1, also known as CCL2 and its receptor CCR2 are involved in recruitment of immune cells at the sites of injury, and CCL2 levels are elevated in the aqueous humor, and urine of AMD patients [250]. Additionally, loss of function in CX3CR1 due to T280M mutation has been implicated in the development of AMD [251]. However, to date, chemokine knockout mouse models have failed to convincingly demonstrate primary AMD pathology such as drusen, RPE atrophy and/or spontaneous CNV. For example, Cx3cr1−/− display hypertrophic microglia in the sub-retinal space. It has been hypothesized that loss of Cx3cr1 function results in death of photoreceptors, which are then phagocytosed, contributing to drusen formation. Tuo et al developed an accelerated model of AMD by crossing Cx3cr1−/− and Ccl2−/− mice, which developed thickening of Bruch’s membrane, RPE changes, photoreceptor atrophy and neovascularization extending into the retina, in as early as 6 weeks of age [252]. It was later discovered that these findings may have been complicated by the retinal expression of the Crbrd8 mutation [253]. Beyond chemokines, there are several other models in which immune cell recruitment into the sub-retinal space has been reported, following laser induced experimental CNV, including in aged Ahr−/− and PPARβ/δ−/− mice [213,234]. Collectively, these studies support a contributing role of chemokines and the involvement of immune cells in the development of AMD and highlight some of the models that may be used to test therapies targeting accumulation of immune cells below the retina or within CNV lesions.
8. Conclusions
The challenges we continue to face in understanding the pathogenesis of complex retinal diseases such as DR and AMD, are dependent on a better appreciation of the various nuances of human pathology and attempting to recreate these features in in vitro and in vivo models. This may be in part dependent on better characterization of disease phenotypes in association with patient genotypes. The in vivo models that have been produced so far, take advantage of the risk factors that have been identified to date and each develop some characteristics of disease, but not all of them. As such they are often criticized as not being the ‘perfect’ model such that the search continues. Yet it remains to be seen whether or not developing a ‘perfect’ model of a complex retinal disease such as DR or AMD, though meritorious, would be an achievable goal, given the clinical and epidemiological information we have so far, which indicate that there are no patients that express all risk factors identified for the disease nor are there patients that present with all the clinical phenotypic features. It is arguable that the quest for the ‘perfect’ model of DR and AMD until better phenotypic-genotypic associations and sub-classifications of each of these complex diseases are available, may be a futile one. Nevertheless, this does not diminish the value of the models currently available, each of which are credible resources and given an appreciation of the disease relevant pathology they exhibit, may be used to not only identify new signaling pathways contributing to initiation and progression of disease but also new therapeutic targets to pursue. Given the availability of possible drugs for therapeutic testing, these models would be valid platforms to investigate drug efficacy, in a targeted manner, and on specific pathological features. Thus it is critical during experimental design to be aware of the disease relevant endpoints and utilize the appropriate model that will facilitate an accurate interpretation of the therapeutic potential of a drug on disease outcome.
9. Expert opinion
As mentioned earlier, the multi-factorial nature of complex non-Mendelian retinal diseases such as DR and AMD has made understanding the pathogenesis and identification of effective therapeutic treatments challenging. However, as highlighted in our review, there are a number of useful in vitro and in vivo tools that are highly relevant and have already led to breakthroughs and clinically approved therapies. Critical to this goal, has been identification of the appropriate model to use in order to test a potential therapy, identifying the appropriate endpoint by using the model, as well as appreciating possible drawbacks of the model. Currently, the use of a specific model to target one particular aspect of the disease in question has garnered considerable popularity; however it may also add cost, should additional aspects of the disease and thus additional experimental models need to be investigated. Validation of pharmacological agents follows this same logic necessitating the selection of an in vivo model that exhibits the pathology being addressed by the drug. Similarly, in vitro models can mimic features of a disease and allow for extensive dose-response and time-course studies to be performed efficiently and in a cost effective manner. For DR, the combination of type 1 and 2 diabetes animal models that progress to non-proliferative stages of disease as well as the OIR animal model, which features neovascularization during development, rather than associated with diabetes, has already promoted significant advances in our understanding of DR pathogenesis and led to recent success of steroids and anti-VEGF treatments. Analogously, the most popular model used to identify potential therapies for the wet clinical sub-type of AMD has been the laser induced choroidal neovascularization mouse model and though it diverges in some respects from the human form, pre-clinical studies with this model led to clinical studies that eventually approved anti-VEGF therapies for treatment of neovascular AMD. This model continues to serve as the ‘go to’ model to test emerging new therapies.
It is important to note, that though the etiology of DR and AMD are quite different they share common pathogenic mechanisms associated with their initiation and progression. Additionally, there is overlap in the number of cells that are affected in both diseases. The RPE cells serve a good example, in that they constitute a common denominator in the disease pathology of both DR and AMD, yet are characterized by their ability to regulate the cytokine milieu in the outer retina, their interactions with overlying photoreceptors, as well as the underlying Bruch’s membrane and choroidal vasculature. With this in mind, should a therapy be identified that could strengthen ailing RPE cells, it would be useful to consider its potential for both diseases. Likewise, improving aberrant endothelial cells (retinal in origin in the case of DR and choroidal in the case of AMD) offer a common target for treating the neovascular components of both retinal diseases. The use of anti-VEGF approaches to treat neovascularization in both DR and AMD presents the text book example of taking advantage of the common pathogenic pathways involved in two different retinal diseases. After the identification of VEGF as an angiogenic molecule, anti-VEGF therapy was first shown to be effective in inhibiting retinal neovascularization in models of retinal ischemia and retinopathy of prematurity, which was subsequently tested as a treatment for choroidal neovascularization associated with advanced AMD. Additional examples of drugs currently under investigation for both DR and AMD include targeting various growth factors as well as components of the complement pathway. Thus, it is safe to say that future studies and breakthroughs in identifying potential therapies for complex diseases will benefit from an awareness of common pathways and cell-types affected, as they may present new avenues for drug discovery including repurposing existing drugs for new indications.
Article highlights box.
Current treatments for complex retinal diseases including diabetic retinopathy (DR) and age-related macular degeneration (AMD), major causes of vision loss in the young and elderly populations, are limited.
In vitro and in vivo models are valuable tools to not only study the pathogenesis of disease, but also identify new therapeutic targets.
The complexity of retinal diseases such as DR and AMD, may preclude development of the ‘perfect’ model, which features all the phenotypic and genotypic characteristics of the human disease.
Rational design should be employed when using models currently available in pre-clinical studies to validate the efficacy of drugs; cognizant of the strengths and weaknesses of the model, as well as the phenotype or pathogenic pathway being targeted.
New avenues for drug discovery may arise from breakthroughs in identifying potential therapies for other complex diseases, with which DR and AMD share common pathways and/or cell-types affected.
Acknowledgments
Funding
The authors acknowledge the following funding agencies for their support of our ongoing research projects: the National Eye Institute grants: EY02868 (to G Malek), EY016077 (to JV Busik), EY025383 (to MB Grant and JV Busik), and P30 EY005722 (to the Duke Eye Center); the Edward N. & Della L. Thome Memorial Foundation Award; BrightFocus Macular Degeneration Grant and the Research to Prevent Blindness, Inc (RPB) Core grant (to the Duke Eye Center).
List of abbreviations:
- AMD
Age-related Macular Degeneration
- DR
Diabetic Retinopathy
- RPE
Retinal Pigment Epithelium
- REC
Retinal Endothelial Cells
- BREC
Bovine Retinal Endothelial Cells
- HREC
Human Retinal Endothelial Cells
- PECAM
Platelet Endothelial Cell Adhesion Molecule
- ERG
Electroretinography
- VEGF
Vascular Endothelial Growth Factor
- ICAM-1
Intercellular Adhesion Molecule-1
- VCAM-1
Vascular Cell Adhesion Molecule-1
- LDL
Low Density Lipoproteins
- PEDF
Pigment Epithelial-Derived Factor
- CACs
Circulating Angiogenic Cells
- RGSs
Retinal Ganglion Cells
- STZ
Streptozotocin
- INL
Inner Nuclear Layer
- ONL
Outer Nuclear Layer
- IPL
Inner Plexiform Layer
- NOD
Non-Obese Diabetic
- BRB
Blood Retina Barrier
- SOD
Superoxide Dismutase
- CNV
Choroidal NeoVascularization
- AhR
Aryl Hydrocarbon Receptor
- Apo
Apolipoprotein
- PPAR
Peroxisome Proliferator Activated Receptor
- CFH:
Complement Factor H
- GA
Geographic Atrophy
- SNP
Single Nucleotide Polymorphism
- T1D
Type 1 Diabetes
- T2D
Type 2 Diabetes
- OIR
Oxygen Induced Retinopathy
- DVP
Deep Vascular Plexus
Footnotes
Declaration of Interest:
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. Peer reviewers on this manuscript have no relevant financial or other relationships to disclose
Bibliography
Papers of special note have been indicated as either of interest (*) or considerable interest (**) to readers.
- 1.Parr T, Friston KJ. The active construction of the visual world. Neuropsychologia (2017) 104(92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Fine SL, Berger JW, Maguire MG, et al. Age-related macular degeneration. The New England journal of medicine (2000) 342(7):483–492. [DOI] [PubMed] [Google Scholar]
- 3.Fong DS, Aiello L, Gardner TW, et al. Diabetic retinopathy. Diabetes Care (2003) 26(1):226–229. [DOI] [PubMed] [Google Scholar]
- 4.Wong WL, Su X, Li X, et al. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: A systematic review and meta-analysis. The Lancet Global health (2014) 2(2):e106–116. [DOI] [PubMed] [Google Scholar]
- 5.Zheng Y, He M, Congdon N. The worldwide epidemic of diabetic retinopathy. Indian J Ophthalmol (2012) 60(5):428–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Eyetech Study G Preclinical and phase 1a clinical evaluation of an anti-vegf pegylated aptamer (eye001) for the treatment of exudative age-related macular degeneration. Retina (2002) 22(2):143–152. [DOI] [PubMed] [Google Scholar]
- 7.Meyer CH, Holz FG. Preclinical aspects of anti-vegf agents for the treatment of wet amd: Ranibizumab and bevacizumab. Eye (2011) 25(6):661–672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Antonetti DA, Klein R, Gardner TW. Diabetic retinopathy. The New England journal of medicine (2012) 366(13):1227–1239. [DOI] [PubMed] [Google Scholar]
- 9.Antonetti DA, Barber AJ, Khin S, et al. Vascular permeability in experimental diabetes is associated with reduced endothelial occludin content: Vascular endothelial growth factor decreases occludin in retinal endothelial cells. Penn state retina research group. Diabetes (1998) 47(12):1953–1959. [DOI] [PubMed] [Google Scholar]
- 10.Bullard SR, Hatchell DL, Cohen HJ, et al. Increased adhesion of neutrophils to retinal vascular endothelial cells exposed to hyperosmolarity. Exp Eye Res (1994) 58(6):641–647. [DOI] [PubMed] [Google Scholar]
- 11.Chibber R, Molinatti PA, Rosatto N, et al. Toxic action of advanced glycation end products on cultured retinal capillary pericytes and endothelial cells: Relevance to diabetic retinopathy. Diabetologia (1997) 40(2):156–164. [DOI] [PubMed] [Google Scholar]
- 12.Connell P, Walshe T, Ferguson G, et al. Elevated glucose attenuates agonist- and flow-stimulated endothelial nitric oxide synthase activity in microvascular retinal endothelial cells. Endothelium : journal of endothelial cell research (2007) 14(1):17–24. [DOI] [PubMed] [Google Scholar]
- 13.Elayappan B, Ravinarayannan H, Pasha SP, et al. Pedf inhibits vegf- and epo- induced angiogenesis in retinal endothelial cells through interruption of pi3k/akt phosphorylation. Angiogenesis (2009) 12(4):313–324. [DOI] [PubMed] [Google Scholar]
- 14.Fernandes R, Girao H, Pereira P. High glucose down-regulates intercellular communication in retinal endothelial cells by enhancing degradation of connexin 43 by a proteasome-dependent mechanism. J Biol Chem (2004) 279(26):27219–27224. [DOI] [PubMed] [Google Scholar]
- 15.Gardner TW, Lieth E, Khin SA, et al. Astrocytes increase barrier properties and zo-1 expression in retinal vascular endothelial cells. Invest Ophthalmol Vis Sci (1997) 38(11):2423–2427. [PubMed] [Google Scholar]
- 16.Giannini S, Cresci B, Manuelli C, et al. Insulin-like growth factor binding protein production in bovine retinal endothelial cells. Metabolism: clinical and experimental (1997) 46(12):1367–1379. [DOI] [PubMed] [Google Scholar]
- 17.King GL, Buzney SM, Kahn CR, et al. Differential responsiveness to insulin of endothelial and support cells from micro- and macrovessels. J Clin Invest (1983) 71(4):974–979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Knott RM, Robertson M, Forrester JV. Regulation of glucose transporter (glut 3) and aldose reductase mrna inbovine retinal endothelial cells and retinal pericytes in high glucose and high galactose culture. Diabetologia (1993) 36(9):808–812. [DOI] [PubMed] [Google Scholar]
- 19.Lee TS, MacGregor LC, Fluharty SJ, et al. Differential regulation of protein kinase c and (na,k)-adenosine triphosphatase activities by elevated glucose levels in retinal capillary endothelial cells. J Clin Invest (1989) 83(1):90–94. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 20.Li W, Liu X, Yanoff M, et al. Cultured retinal capillary pericytes die by apoptosis after an abrupt fluctuation from high to low glucose levels: A comparative study with retinal capillary endothelial cells. Diabetologia (1996) 39(5):537–547. [DOI] [PubMed] [Google Scholar]
- 21.Lyons TJ, Li W, Wells-Knecht MC, et al. Toxicity of mildly modified low-density lipoproteins to cultured retinal capillary endothelial cells and pericytes. Diabetes (1994) 43(9):1090–1095.* This was the first study to demonstrate the role of oxydized and glycated low-density lipoproteins in REC and pericyte damage.
- 22.Mamputu JC, Renier G. Advanced glycation end products increase, through a protein kinase c-dependent pathway, vascular endothelial growth factor expression in retinal endothelial cells. Inhibitory effect of gliclazide. Journal of diabetes and its complications (2002) 16(4):284–293. [DOI] [PubMed] [Google Scholar]
- 23.Sheikpranbabu S, Haribalaganesh R, Banumathi E, et al. Pigment epithelium-derived factor inhibits advanced glycation end-product-induced angiogenesis and stimulates apoptosis in retinal endothelial cells. Life sciences (2009) 85(21–22):719–731. [DOI] [PubMed] [Google Scholar]
- 24.Sone H, Deo BK, Kumagai AK. Enhancement of glucose transport by vascular endothelial growth factor in retinal endothelial cells. Invest Ophthalmol Vis Sci (2000) 41(7):1876–1884. [PubMed] [Google Scholar]
- 25.Thieme H, Aiello LP, Takagi H, et al. Comparative analysis of vascular endothelial growth factor receptors on retinal and aortic vascular endothelial cells. Diabetes (1995) 44(1):98–103. [DOI] [PubMed] [Google Scholar]
- 26.Wu H, Xia X, Jiang C, et al. High glucose attenuates insulin-induced vegf expression in bovine retinal microvascular endothelial cells. Eye (2010) 24(1):145–151. [DOI] [PubMed] [Google Scholar]
- 27.Busik JV, Mohr S, Grant MB. Hyperglycemia-induced reactive oxygen species toxicity to endothelial cells is dependent on paracrine mediators. Diabetes (2008) 57(7):1952–1965.* This study compared the response to hyperglycemia and cytokines in RPE, Muller and REC and demonstrated that REC cells have the lowest sensitivity to high glucose among retinal cells.
- 28.Cai J, Ahmad S, Jiang WG, et al. Activation of vascular endothelial growth factor receptor-1 sustains angiogenesis and bcl-2 expression via the phosphatidylinositol 3-kinase pathway in endothelial cells. Diabetes (2003) 52(12):2959–2968. [DOI] [PubMed] [Google Scholar]
- 29.Chen W, Esselman WJ, Jump DB, et al. Anti-inflammatory effect of docosahexaenoic acid on cytokine-induced adhesion molecule expression in human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci (2005) 46(11):4342–4347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chen W, Jump DB, Grant MB, et al. Dyslipidemia, but not hyperglycemia, induces inflammatory adhesion molecules in human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci (2003) 44(11):5016–5022. *This is the first demonstration of the role of pro-inflammatory lipids in REC damage.
- 31.Ding Y, Hu Z, Luan J, et al. Protective effect of mir-200b/c by inhibiting vasohibin-2 in human retinal microvascular endothelial cells. Life sciences (2017). [DOI] [PubMed] [Google Scholar]
- 32.Fernandes R, Hosoya K, Pereira P. Reactive oxygen species downregulate glucose transport system in retinal endothelial cells. American journal of physiology Cell physiology (2011) 300(4):C927–936. [DOI] [PubMed] [Google Scholar]
- 33.Grant MB, Caballero S, Millard WJ. Inhibition of igf-i and b-fgf stimulated growth of human retinal endothelial cells by the somatostatin analogue, octreotide: A potential treatment for ocular neovascularization. Regulatory peptides (1993) 48(1–2):267–278. [DOI] [PubMed] [Google Scholar]
- 34.Grant MB, Guay C. Plasminogen activator production by human retinal endothelial cells of nondiabetic and diabetic origin. Invest Ophthalmol Vis Sci (1991) 32(1):53–64. [PubMed] [Google Scholar]
- 35.Ibrahim AS, Saleh H, El-Shafey M, et al. Targeting of 12/15-lipoxygenase in retinal endothelial cells, but not in monocytes/macrophages, attenuates high glucose-induced retinal leukostasis. Biochimica et biophysica acta (2017) 1862(6):636–645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kady N, Yan Y, Salazar T, et al. Increase in acid sphingomyelinase level in human retinal endothelial cells and cd34+ circulating angiogenic cells isolated from diabetic individuals is associated with dysfunctional retinal vasculature and vascular repair process in diabetes. Journal of clinical lipidology (2017) 11(3):694–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ma J, Liu T, Dong X. Advanced glycation end products of bovine serum albumin-induced endothelial-to-mesenchymal transition in cultured human and monkey endothelial cells via protein kinase b signaling cascades. Molecular vision (2010) 16(2669–2679. [PMC free article] [PubMed] [Google Scholar]
- 38.Maines LW, French KJ, Wolpert EB, et al. Pharmacologic manipulation of sphingosine kinase in retinal endothelial cells: Implications for angiogenic ocular diseases. Invest Ophthalmol Vis Sci (2006) 47(11):5022–5031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mammadzada P, Gudmundsson J, Kvanta A, et al. Differential hypoxic response of human choroidal and retinal endothelial cells proposes tissue heterogeneity of ocular angiogenesis. Acta ophthalmologica (2016) 94(8):805–814. [DOI] [PubMed] [Google Scholar]
- 40.Miller EC, Capps BE, Sanghani RR, et al. Regulation of igf-i signaling in retinal endothelial cells by hyperglycemia. Invest Ophthalmol Vis Sci (2007) 48(8):3878–3887. [DOI] [PubMed] [Google Scholar]
- 41.Nawaz MI, Van Raemdonck K, Mohammad G, et al. Autocrine ccl2, cxcl4, cxcl9 and cxcl10 signal in retinal endothelial cells and are enhanced in diabetic retinopathy. Exp Eye Res (2013) 109(67–76. [DOI] [PubMed] [Google Scholar]
- 42.Opreanu M, Lydic TA, Reid GE, et al. Inhibition of cytokine signaling in human retinal endothelial cells through downregulation of sphingomyelinases by docosahexaenoic acid. Invest Ophthalmol Vis Sci (2010) 51(6):3253–3263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Spoerri PE, Afzal A, Li Calzi S, et al. Effects of vegfr-1, vegfr-2, and igf-ir hammerhead ribozymes on glucose-mediated tight junction expression in cultured human retinal endothelial cells. Molecular vision (2006) 12(32–42. [PubMed] [Google Scholar]
- 44.Steinle JJ, Chin VC, Williams KP, et al. Beta-adrenergic receptor stimulation modulates inos protein levels through p38 and erk1/2 signaling in human retinal endothelial cells. Exp Eye Res (2008) 87(1):30–34. [DOI] [PubMed] [Google Scholar]
- 45.Su T, Gillies MC. A simple method for the in vitro culture of human retinal capillary endothelial cells. Invest Ophthalmol Vis Sci (1992) 33(10):2809–2813. [PubMed] [Google Scholar]
- 46.Sun J, Xu Y, Sun S, et al. Intermittent high glucose enhances cell proliferation and vegf expression in retinal endothelial cells: The role of mitochondrial reactive oxygen species. Molecular and cellular biochemistry (2010) 343(1–2):27–35. [DOI] [PubMed] [Google Scholar]
- 47.Trudeau K, Molina AJ, Guo W, et al. High glucose disrupts mitochondrial morphology in retinal endothelial cells: Implications for diabetic retinopathy. Am J Pathol (2010) 177(1):447–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xiaozhuang Z, Xianqiong L, Jingbo J, Shuiqing H, Jie Y, Yunbin C: Isolation and characterization of fetus human retinal microvascular endothelial cells. Ophthalmic Res (2010) 44(2):125–130. [DOI] [PubMed] [Google Scholar]
- 49.Yamagishi S, Okamoto T, Amano S, et al. Palmitate-induced apoptosis of microvascular endothelial cells and pericytes. Molecular medicine (2002) 8(4):179–184. [PMC free article] [PubMed] [Google Scholar]
- 50.Ye EA, Steinle JJ. Mir-15b/16 protects primary human retinal microvascular endothelial cells against hyperglycemia-induced increases in tumor necrosis factor alpha and suppressor of cytokine signaling 3. Journal of neuroinflammation (2015) 12(44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sheikpranbabu S, Kalishwaralal K, Venkataraman D, et al. Silver nanoparticles inhibit vegf-and il-1beta-induced vascular permeability via src dependent pathway in porcine retinal endothelial cells. Journal of nanobiotechnology (2009) 7(8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Takamura Y, Tomomatsu T, Kubo E, et al. Role of the polyol pathway in high glucose-induced apoptosis of retinal pericytes and proliferation of endothelial cells. Invest Ophthalmol Vis Sci (2008) 49(7):3216–3223. [DOI] [PubMed] [Google Scholar]
- 53.Caprnda M, Kubatka P, Saxena S, et al. The impact of hyperglycemia on vegf secretion in retinal endothelial cells. Folia medica (2017) 59(2):183–189. [DOI] [PubMed] [Google Scholar]
- 54.Mondragon AA, Betts-Obregon BS, Moritz RJ, et al. Bigh3 protein and macrophages in retinal endothelial cell apoptosis. Apoptosis : an international journal on programmed cell death (2015) 20(1):29–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Frank RN, Kinsey VE, Frank KW, et al. In vitro proliferation of endothelial cells from kitten retinal capillaries. Invest Ophthalmol Vis Sci (1979) 18(11):1195–1200. [PubMed] [Google Scholar]
- 56.Fan Y, Qiao Y, Huang J, et al. Protective effects of panax notoginseng saponins against high glucose-induced oxidative injury in rat retinal capillary endothelial cells. Evidence-based complementary and alternative medicine : eCAM (2016) 2016(5326382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Boosani CS, Nalabothula N, Sheibani N, et al. Inhibitory effects of arresten on bfgf-induced proliferation, migration, and matrix metalloproteinase-2 activation in mouse retinal endothelial cells. Current eye research (2010) 35(1):45–55. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 58.Su X, Sorenson CM, Sheibani N. Isolation and characterization of murine retinal endothelial cells. Molecular vision (2003) 9(171–178.** The method for isolation and culture of mouse REC is described in this paper.
- 59.Antonetti DA, Wolpert EB. Isolation and characterization of retinal endothelial cells. Methods in molecular medicine (2003) 89(365–374. [DOI] [PubMed] [Google Scholar]
- 60.Schor AM, Schor SL. The isolation and culture of endothelial cells and pericytes from the bovine retinal microvasculature: A comparative study with large vessel vascular cells. Microvascular research (1986) 32(1):21–38. [DOI] [PubMed] [Google Scholar]
- 61.Matsubara TA, Murata TA, Wu GS, et al. Isolation and culture of rat retinal microvessel endothelial cells using magnetic beads coated with antibodies to pecam-1. Current eye research (2000) 20(1):1–7. [PubMed] [Google Scholar]
- 62.Chen X, Li J, Li M, et al. Kh902 suppresses high glucose-induced migration and sprouting of human retinal endothelial cells by blocking vegf and pigf. Diabetes, obesity & metabolism (2013) 15(3):224–233. [DOI] [PubMed] [Google Scholar]
- 63.Duffy A, Liew A, O’Sullivan J, et al. Distinct effects of high-glucose conditions on endothelial cells of macrovascular and microvascular origins. Endothelium : journal of endothelial cell research (2006) 13(1):9–16. [DOI] [PubMed] [Google Scholar]
- 64.Giannini S, Cresci B, Pala L, et al. Igfbps modulate igf-i- and high glucose-controlled growth of human retinal endothelial cells. The Journal of endocrinology (2001) 171(2):273–284. [DOI] [PubMed] [Google Scholar]
- 65.Li J, Chen X, Xiao W, et al. Mitochondria-targeted antioxidant peptide ss31 attenuates high glucose-induced injury on human retinal endothelial cells. Biochemical and biophysical research communications (2011) 404(1):349–356. [DOI] [PubMed] [Google Scholar]
- 66.Thakran S, Zhang Q, Morales-Tirado V, et al. Pioglitazone restores igfbp-3 levels through DNA pk in retinal endothelial cells cultured in hyperglycemic conditions. Invest Ophthalmol Vis Sci (2014) 56(1):177–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Capozzi ME, McCollum GW, Cousins DB, et al. Linoleic acid is a diabetes-relevant stimulator of retinal inflammation in human retinal muller cells and microvascular endothelial cells. Journal of diabetes & metabolism (2016) 7(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Chen W, Jump DB, Esselman WJ, et al. Inhibition of cytokine signaling in human retinal endothelial cells through modification of caveolae/lipid rafts by docosahexaenoic acid. Invest Ophthalmol Vis Sci (2007) 48(1):18–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Delton-Vandenbroucke I, Grammas P, Anderson RE. Polyunsaturated fatty acid metabolism in retinal and cerebral microvascular endothelial cells. Journal of lipid research (1997) 38(1):147–159. [PubMed] [Google Scholar]
- 70.Delton-Vandenbroucke I, Grammas P, Anderson RE. Regulation of n-3 and n-6 fatty acid metabolism in retinal and cerebral microvascular endothelial cells by high glucose. Journal of neurochemistry (1998) 70(2):841–849. [DOI] [PubMed] [Google Scholar]
- 71.Lecomte M, Paget C, Ruggiero D, et al. Docosahexaenoic acid is a major n-3 polyunsaturated fatty acid in bovine retinal microvessels. Journal of neurochemistry (1996) 66(5):2160–2167. [DOI] [PubMed] [Google Scholar]
- 72.Hammer SS, Beli E, Kady N, et al. The mechanism of diabetic retinopathy pathogenesis unifying key lipid regulators, sirtuin 1 and liver x receptor. EBioMedicine (2017) 22(181–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kowluru RA, Odenbach S. Role of interleukin-1beta in the pathogenesis of diabetic retinopathy. Br J Ophthalmol (2004) 88(10):1343–1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Antonetti DA, Barber AJ, Hollinger LA, et al. Vascular endothelial growth factor induces rapid phosphorylation of tight junction proteins occludin and zonula occluden 1. A potential mechanism for vascular permeability in diabetic retinopathy and tumors. J Biol Chem (1999) 274(33):23463–23467.** This study identified the role of VEGF-induced occludin and zonula occluden 1 phosphorylation in the loss of tight junction integrity leading to increased vascular permeability in REC.
- 75.Xie J, Liu X, Li Y, et al. Validation of rt-qpcr reference genes and determination of robo4 expression levels in human retinal endothelial cells under hypoxia and/or hyperglycemia. Gene (2016) 585(1):135–142. [DOI] [PubMed] [Google Scholar]
- 76.Wang D, Wang L, Gu J, et al. Scutellarin inhibits high glucose-induced and hypoxia-mimetic agent-induced angiogenic effects in human retinal endothelial cells through reactive oxygen species/hypoxia-inducible factor-1alpha/vascular endothelial growth factor pathway. J Cardiovasc Pharmacol (2014) 64(3):218–227. [DOI] [PubMed] [Google Scholar]
- 77.Chan PS, Kanwar M, Kowluru RA. Resistance of retinal inflammatory mediators to suppress after reinstitution of good glycemic control: Novel mechanism for metabolic memory. Journal of diabetes and its complications (2010) 24(1):55–63.** An important concept of metabolic memory was introduced in the paper
- 78.Madsen-Bouterse SA, Mohammad G, Kanwar M, et al. Role of mitochondrial DNA damage in the development of diabetic retinopathy, and the metabolic memory phenomenon associated with its progression. Antioxidants & redox signaling (2010) 13(6):797–805.* The role of mitochondrial damage in diabetes-induced REC pathology is demonstrated in this study
- 79.Santos JM, Kowluru RA. Impaired transport of mitochondrial transcription factor a (tfam) and the metabolic memory phenomenon associated with the progression of diabetic retinopathy. Diabetes/metabolism research and reviews (2013) 29(3):204–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhong Q, Kowluru RA. Epigenetic changes in mitochondrial superoxide dismutase in the retina and the development of diabetic retinopathy. Diabetes (2011) 60(4):1304–1313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Diaz-Coranguez M, Chao DL, Salero EL, et al. Cell autonomous sonic hedgehog signaling contributes to maintenance of retinal endothelial tight junctions. Exp Eye Res (2017) 164(82–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Samra YA, Saleh HM, Hussein KA, et al. Adenosine deaminase-2-induced hyperpermeability in human retinal vascular endothelial cells is suppressed by microrna-146b-3p. Invest Ophthalmol Vis Sci (2017) 58(2):933–943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Xie J, Gong Q, Liu X, et al. Transcription factor sp1 mediates hyperglycemia-induced upregulation of roundabout4 in retinal microvascular endothelial cells. Gene (2017) 616(31–40. [DOI] [PubMed] [Google Scholar]
- 84.Castellon R, Caballero S, Hamdi HK, et al. Effects of tenascin-c on normal and diabetic retinal endothelial cells in culture. Invest Ophthalmol Vis Sci (2002) 43(8):2758–2766. [PubMed] [Google Scholar]
- 85.Kielczewski JL, Jarajapu YP, McFarland EL, et al. Insulin-like growth factor binding protein-3 mediates vascular repair by enhancing nitric oxide generation. Circulation research (2009) 105(9):897–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kielczewski JL, Li Calzi S, Shaw LC, et al. Free insulin-like growth factor binding protein-3 (igfbp-3) reduces retinal vascular permeability in association with a reduction of acid sphingomyelinase (asmase). Invest Ophthalmol Vis Sci (2011) 52(11):8278–8286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Li Calzi S, Purich DL, Chang KH, et al. Carbon monoxide and nitric oxide mediate cytoskeletal reorganization in microvascular cells via vasodilator-stimulated phosphoprotein phosphorylation: Evidence for blunted responsiveness in diabetes. Diabetes (2008) 57(9):2488–2494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Mizutani M, Kern TS, Lorenzi M. Accelerated death of retinal microvascular cells in human and experimental diabetic retinopathy. J Clin Invest (1996) 97(12):2883–2890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Munjal ID, McLean NV, Grant MB, et al. Differences in the synthesis of secreted proteins in human retinal endothelial cells of diabetic and nondiabetic origin. Current eye research (1994) 13(4):303–310. [DOI] [PubMed] [Google Scholar]
- 90.Tikhonenko M, Lydic TA, Opreanu M, et al. N-3 polyunsaturated fatty acids prevent diabetic retinopathy by inhibition of retinal vascular damage and enhanced endothelial progenitor cell reparative function. PLoS One (2013) 8(1):e55177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Du XL, Edelstein D, Rossetti L, et al. Hyperglycemia-induced mitochondrial superoxide overproduction activates the hexosamine pathway and induces plasminogen activator inhibitor-1 expression by increasing sp1 glycosylation. Proc Natl Acad Sci U S A (2000) 97(22):12222–12226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Du Y, Miller CM, Kern TS. Hyperglycemia increases mitochondrial superoxide in retina and retinal cells. Free radical biology & medicine (2003) 35(11):1491–1499. [DOI] [PubMed] [Google Scholar]
- 93.Gao J, Zheng Z, Gu Q, et al. Deacetylation of mnsod by parp-regulated sirt3 protects retinal capillary endothelial cells from hyperglycemia-induced damage. Biochemical and biophysical research communications (2016) 472(3):425–431. [DOI] [PubMed] [Google Scholar]
- 94.Lopez-Quintero SV, Cancel LM, Pierides A, et al. High glucose attenuates shear-induced changes in endothelial hydraulic conductivity by degrading the glycocalyx. PLoS One (2013) 8(11):e78954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Perrone L, Devi TS, Hosoya K, et al. Thioredoxin interacting protein (txnip) induces inflammation through chromatin modification in retinal capillary endothelial cells under diabetic conditions. Journal of cellular physiology (2009) 221(1):262–272. [DOI] [PubMed] [Google Scholar]
- 96.Zhang Y, Qu Y, Niu T, et al. O-glcnac modification of sp1 mediates hyperglycaemia-induced icam-1 up-regulation in endothelial cells. Biochemical and biophysical research communications (2017) 484(1):79–84. [DOI] [PubMed] [Google Scholar]
- 97.Castilho A, Aveleira CA, Leal EC, et al. Heme oxygenase-1 protects retinal endothelial cells against high glucose- and oxidative/nitrosative stress-induced toxicity. PLoS One (2012) 7(8):e42428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Dagher Z, Gerhardinger C, Vaz J, et al. The increased transforming growth factor-beta signaling induced by diabetes protects retinal vessels. Am J Pathol (2017) 187(3):627–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.el-Remessy AB, Bartoli M, Platt DH, et al. Oxidative stress inactivates vegf survival signaling in retinal endothelial cells via pi 3-kinase tyrosine nitration. Journal of cell science (2005) 118(Pt 1):243–252. [DOI] [PubMed] [Google Scholar]
- 100.Leal EC, Aveleira CA, Castilho AF, et al. High glucose and oxidative/nitrosative stress conditions induce apoptosis in retinal endothelial cells by a caspase-independent pathway. Exp Eye Res (2009) 88(5):983–991. [DOI] [PubMed] [Google Scholar]
- 101.Li J, Wang P, Yu S, et al. Calcium entry mediates hyperglycemia-induced apoptosis through ca(2+)/calmodulin-dependent kinase ii in retinal capillary endothelial cells. Molecular vision (2012) 18(2371–2379. [PMC free article] [PubMed] [Google Scholar]
- 102.Betts-Obregon BS, Mondragon AA, Mendiola AS, et al. Tgfbeta induces bigh3 expression and human retinal pericyte apoptosis: A novel pathway of diabetic retinopathy. Eye (2016) 30(12):1639–1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Devi TS, Hosoya K, Terasaki T, et al. Critical role of txnip in oxidative stress, DNA damage and retinal pericyte apoptosis under high glucose: Implications for diabetic retinopathy. Experimental cell research (2013) 319(7):1001–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ding L, Cheng R, Hu Y, et al. Peroxisome proliferator-activated receptor alpha protects capillary pericytes in the retina. Am J Pathol (2014) 184(10):2709–2720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Fu D, Wu M, Zhang J, et al. Mechanisms of modified ldl-induced pericyte loss and retinal injury in diabetic retinopathy. Diabetologia (2012) 55(11):3128–3140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Fu D, Yu JY, Yang S, et al. Survival or death: A dual role for autophagy in stress-induced pericyte loss in diabetic retinopathy. Diabetologia (2016) 59(10):2251–2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Song W, Barth JL, Lu K, et al. Effects of modified low-density lipoproteins on human retinal pericyte survival. Annals of the New York Academy of Sciences (2005) 1043(390–395. [DOI] [PubMed] [Google Scholar]
- 108.Platania CBM, Giurdanella G, Di Paola L, et al. P2×7 receptor antagonism: Implications in diabetic retinopathy. Biochem Pharmacol (2017) 138(130–139. [DOI] [PubMed] [Google Scholar]
- 109.Durham JT, Dulmovits BM, Cronk SM, et al. Pericyte chemomechanics and the angiogenic switch: Insights into the pathogenesis of proliferative diabetic retinopathy? Invest Ophthalmol Vis Sci (2015) 56(6):3441–3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Chavira-Suarez E, Sandoval A, Felix R, et al. Expression and high glucose-mediated regulation of k+ channel interacting protein 3 (kchip3) and kv4 channels in retinal muller glial cells. Biochemical and biophysical research communications (2011) 404(2):678–683. [DOI] [PubMed] [Google Scholar]
- 111.Cheng L, Bu H, Portillo JA, et al. Modulation of retinal muller cells by complement receptor c5ar. Invest Ophthalmol Vis Sci (2013) 54(13):8191–8198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Fu D, Yu JY, Connell AR, et al. Beneficial effects of berberine on oxidized ldl-induced cytotoxicity to human retinal muller cells. Invest Ophthalmol Vis Sci (2016) 57(7):3369–3379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Jayaguru P, Mohr S. Nuclear gapdh: Changing the fate of muller cells in diabetes. Journal of ocular biology, diseases, and informatics (2011) 4(1–2):34–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Kida T, Ikeda T, Nishimura M, et al. Renin-angiotensin system in proliferative diabetic retinopathy and its gene expression in cultured human muller cells. Japanese journal of ophthalmology (2003) 47(1):36–41. [DOI] [PubMed] [Google Scholar]
- 115.Kusner LL, Sarthy VP, Mohr S. Nuclear translocation of glyceraldehyde-3-phosphate dehydrogenase: A role in high glucose-induced apoptosis in retinal muller cells. Invest Ophthalmol Vis Sci (2004) 45(5):1553–1561. [PubMed] [Google Scholar]
- 116.Li Q, Puro DG. Diabetes-induced dysfunction of the glutamate transporter in retinal muller cells. Invest Ophthalmol Vis Sci (2002) 43(9):3109–3116. [PubMed] [Google Scholar]
- 117.Puro DG. Diabetes-induced dysfunction of retinal muller cells. Transactions of the American Ophthalmological Society (2002) 100(339–352. [PMC free article] [PubMed] [Google Scholar]
- 118.Shen X, Zhong Y, Xie B, et al. Pigment epithelium derived factor as an anti-inflammatory factor against decrease of glutamine synthetase expression in retinal muller cells under high glucose conditions. Graefes Arch Clin Exp Ophthalmol (2010) 248(8):1127–1136. [DOI] [PubMed] [Google Scholar]
- 119.Tien T, Zhang J, Muto T, et al. High glucose induces mitochondrial dysfunction in retinal muller cells: Implications for diabetic retinopathy. Invest Ophthalmol Vis Sci (2017) 58(7):2915–2921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Tretiach M, Madigan MC, Gillies MC. Conditioned medium from mixed retinal pigmented epithelium and muller cell cultures reduces in vitro permeability of retinal vascular endothelial cells. Br J Ophthalmol (2004) 88(7):957–961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Wang LL, Chen H, Huang K, et al. Elevated histone acetylations in muller cells contribute to inflammation: A novel inhibitory effect of minocycline. Glia (2012) 60(12):1896–1905. [DOI] [PubMed] [Google Scholar]
- 122.Wu M, Yang S, Elliott MH, et al. Oxidative and endoplasmic reticulum stresses mediate apoptosis induced by modified ldl in human retinal muller cells. Invest Ophthalmol Vis Sci (2012) 53(8):4595–4604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Gaucher D, Chiappore JA, Paques M, et al. Microglial changes occur without neural cell death in diabetic retinopathy. Vision research (2007) 47(5):612–623. [DOI] [PubMed] [Google Scholar]
- 124.Portillo JC, Lopez Corcino Y, Miao Y, et al. Cd40 in retinal muller cells induces p2×7-dependent cytokine expression in macrophages/microglia in diabetic mice and development of early experimental diabetic retinopathy. Diabetes (2017) 66(2):483–493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ding X, Zhang M, Gu R, et al. Activated microglia induce the production of reactive oxygen species and promote apoptosis of co-cultured retinal microvascular pericytes. Graefes Arch Clin Exp Ophthalmol (2017) 255(4):777–788. [DOI] [PubMed] [Google Scholar]
- 126.Dunn KC, Aotaki-Keen AE, Putkey FR, et al. Arpe-19, a human retinal pigment epithelial cell line with differentiated properties. Exp Eye Res (1996) 62(2):155–169. [DOI] [PubMed] [Google Scholar]
- 127.Luo Y, Zhuo Y, Fukuhara M, et al. Effects of culture conditions on heterogeneity and the apical junctional complex of the arpe-19 cell line. Invest Ophthalmol Vis Sci (2006) 47(8):3644–3655. [DOI] [PubMed] [Google Scholar]
- 128.Calado SM, Alves LS, Simao S, et al. Glut1 activity contributes to the impairment of pedf secretion by the rpe. Molecular vision (2016) 22(761–770. [PMC free article] [PubMed] [Google Scholar]
- 129.Chang ML, Chiu CJ, Shang F, et al. High glucose activates chrebp-mediated hif-1alpha and vegf expression in human rpe cells under normoxia. Adv Exp Med Biol (2014) 801(609–621. [DOI] [PubMed] [Google Scholar]
- 130.Du M, Wu M, Fu D, et al. Effects of modified ldl and hdl on retinal pigment epithelial cells: A role in diabetic retinopathy? Diabetologia (2013) 56(10):2318–2328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Kim DI, Park MJ, Choi JH, et al. Hyperglycemia-induced glp-1r downregulation causes rpe cell apoptosis. The international journal of biochemistry & cell biology (2015) 59(41–51. [DOI] [PubMed] [Google Scholar]
- 132.Li X, Cai Y, Wang YS, et al. Hyperglycaemia exacerbates choroidal neovascularisation in mice via the oxidative stress-induced activation of stat3 signalling in rpe cells. PLoS One (2012) 7(10):e47600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Winkler J, Hoerauf H. Tgf-ss and rpe-derived cells in taut subretinal strands from patients with proliferative vitreoretinopathy. European journal of ophthalmology (2011) 21(4):422–426. [DOI] [PubMed] [Google Scholar]
- 134.Xu HZ, Song Z, Fu S, et al. Rpe barrier breakdown in diabetic retinopathy: Seeing is believing. Journal of ocular biology, diseases, and informatics (2011) 4(1–2):83–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Foresti R, Bucolo C, Platania CM, et al. Nrf2 activators modulate oxidative stress responses and bioenergetic profiles of human retinal epithelial cells cultured in normal or high glucose conditions. Pharmacol Res (2015) 99(296–307. [DOI] [PubMed] [Google Scholar]
- 136.Henry DN, Frank RN, Hootman SR, et al. Glucose-specific regulation of aldose reductase in human retinal pigment epithelial cells in vitro. Invest Ophthalmol Vis Sci (2000) 41(6):1554–1560. [PubMed] [Google Scholar]
- 137.Farnoodian M, Halbach C, Slinger C, et al. High glucose promotes the migration of retinal pigment epithelial cells through increased oxidative stress and pedf expression. American journal of physiology Cell physiology (2016) 311(3):C418–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Liu Y, Leo LF, McGregor C, et al. Pigment epithelium-derived factor (pedf) peptide eye drops reduce inflammation, cell death and vascular leakage in diabetic retinopathy in ins2(akita) mice. Molecular medicine (2012) 18(1387–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Krishnamoorthy RR, Clark AF, Daudt D, et al. A forensic path to rgc-5 cell line identification: Lessons learned. Invest Ophthalmol Vis Sci (2013) 54(8):5712–5719.** This paper describes the importance of proper controls in cell culture model studies. It provided the evidence that RGC-5 cell ine was not of ganglion cell origin.
- 140.Skaper SD. Rodent retinal ganglion cell cultures. Methods in molecular biology (2012) 846(117–129. [DOI] [PubMed] [Google Scholar]
- 141.Fuchs C, Forster V, Balse E, et al. Retinal-cell-conditioned medium prevents tnf-alpha-induced apoptosis of purified ganglion cells. Invest Ophthalmol Vis Sci (2005) 46(8):2983–2991. [DOI] [PubMed] [Google Scholar]
- 142.Unterlauft JD, Claudepierre T, Schmidt M, et al. Enhanced survival of retinal ganglion cells is mediated by muller glial cell-derived pedf. Exp Eye Res (2014) 127(206–214. [DOI] [PubMed] [Google Scholar]
- 143.McCaffery CA, Bennett MR, Dreher B. The survival of neonatal rat retinal ganglion cells in vitro is enhanced in the presence of appropriate parts of the brain. Experimental brain research (1982) 48(3):377–386. [DOI] [PubMed] [Google Scholar]
- 144.Ha Y, Saul A, Tawfik A, et al. Diabetes accelerates retinal ganglion cell dysfunction in mice lacking sigma receptor 1. Molecular vision (2012) 18(2860–2870. [PMC free article] [PubMed] [Google Scholar]
- 145.Tan E, Ding XQ, Saadi A, et al. Expression of cone-photoreceptor-specific antigens in a cell line derived from retinal tumors in transgenic mice. Invest Ophthalmol Vis Sci (2004) 45(3):764–768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Balmer D, Emery M, Andreux P, et al. Autophagy defect is associated with low glucose-induced apoptosis in 661w photoreceptor cells. PLoS One (2013) 8(9):e74162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Lupo G, Motta C, Giurdanella G, et al. Role of phospholipases a2 in diabetic retinopathy: In vitro and in vivo studies. Biochem Pharmacol (2013) 86(11):1603–1613. [DOI] [PubMed] [Google Scholar]
- 148.Watanabe S, Morisaki N, Tezuka M, et al. Cultured retinal pericytes stimulate in vitro angiogenesis of endothelial cells through secretion of a fibroblast growth factor-like molecule. Atherosclerosis (1997) 130(1–2):101–107. [DOI] [PubMed] [Google Scholar]
- 149.Wang Q, Navitskaya S, Chakravarthy H, et al. Dual anti-inflammatory and anti-angiogenic action of mir-15a in diabetic retinopathy. EBioMedicine (2016) 11(138–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Caballero S, Sengupta N, Afzal A, et al. Ischemic vascular damage can be repaired by healthy, but not diabetic, endothelial progenitor cells. Diabetes (2007) 56(4):960–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Chen H, Charlat O, Tartaglia LA, et al. Evidence that the diabetes gene encodes the leptin receptor: Identification of a mutation in the leptin receptor gene in db/db mice. Cell (1996) 84(3):491–495. [DOI] [PubMed] [Google Scholar]
- 152.Makino S, Kunimoto K, Muraoka Y, et al. Breeding of a non-obese, diabetic strain of mice. Jikken dobutsu Experimental animals (1980) 29(1):1–13. [DOI] [PubMed] [Google Scholar]
- 153.Thayer TC, Wilson SB, Mathews CE. Use of nonobese diabetic mice to understand human type 1 diabetes. Endocrinology and metabolism clinics of North America (2010) 39(3):541–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Engerman RL, Bloodworth JM, Experimental diabetic retinopathy in dogs. Arch Ophthalmol (1965) 73(205–210. [DOI] [PubMed] [Google Scholar]
- 155.Goldsmith JR, Jobin C. Think small: Zebrafish as a model system of human pathology. Journal of biomedicine & biotechnology (2012) 2012(817341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Tso MO, Kurosawa A, Benhamou E, et al. Microangiopathic retinopathy in experimental diabetic monkeys. Transactions of the American Ophthalmological Society (1988) 86(389–421. [PMC free article] [PubMed] [Google Scholar]
- 157.Weerasekera LY, Balmer LA, Ram R, et al. Characterization of retinal vascular and neural damage in a novel model of diabetic retinopathy. Invest Ophthalmol Vis Sci (2015) 56(6):3721–3730. [DOI] [PubMed] [Google Scholar]
- 158.Feit-Leichman RA, Kinouchi R, Takeda M, et al. Vascular damage in a mouse model of diabetic retinopathy: Relation to neuronal and glial changes. Invest Ophthalmol Vis Sci (2005) 46(11):4281–4287. [DOI] [PubMed] [Google Scholar]
- 159.Amadio M, Pascale A, Cupri S, et al. Nanosystems based on sirna silencing hur expression counteract diabetic retinopathy in rat. Pharmacol Res (2016) 111(713–720. [DOI] [PubMed] [Google Scholar]
- 160.Martin PM, Roon P, Van Ells TK, et al. Death of retinal neurons in streptozotocin-induced diabetic mice. Invest Ophthalmol Vis Sci (2004) 45(9):3330–3336. [DOI] [PubMed] [Google Scholar]
- 161.Joussen AM, Poulaki V, Mitsiades N, Kirchhof B, et al. Nonsteroidal anti-inflammatory drugs prevent early diabetic retinopathy via tnf-alpha suppression. FASEB J (2002) 16(3):438–440. [DOI] [PubMed] [Google Scholar]
- 162.Joussen AM, Poulaki V, Qin W, et al. Retinal vascular endothelial growth factor induces intercellular adhesion molecule-1 and endothelial nitric oxide synthase expression and initiates early diabetic retinal leukocyte adhesion in vivo. Am J Pathol (2002) 160(2):501–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Engerman RL, Kern TS. Experimental galactosemia produces diabetic-like retinopathy. Diabetes (1984) 33(1):97–100. [DOI] [PubMed] [Google Scholar]
- 164.Kern TS, Engerman RL. A mouse model of diabetic retinopathy. Arch Ophthalmol (1996) 114(8):986–990. [DOI] [PubMed] [Google Scholar]
- 165.Smith LE, Wesolowski E, McLellan A, et al. Oxygen-induced retinopathy in the mouse. Invest Ophthalmol Vis Sci (1994) 35(1):101–111. [PubMed] [Google Scholar]
- 166.Barber AJ, Antonetti DA, Kern TS, et al. The ins2akita mouse as a model of early retinal complications in diabetes. Invest Ophthalmol Vis Sci (2005) 46(6):2210–2218. [DOI] [PubMed] [Google Scholar]
- 167.Robinson R, Barathi VA, Chaurasia SS, et al. Update on animal models of diabetic retinopathy: From molecular approaches to mice and higher mammals. Disease models & mechanisms (2012) 5(4):444–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Lee S, Harris NR. Losartan and ozagrel reverse retinal arteriolar constriction in non-obese diabetic mice. Microcirculation (2008) 15(5):379–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Midena E, Segato T, Radin S, et al. Studies on the retina of the diabetic db/db mouse. I. Endothelial cell-pericyte ratio. Ophthalmic Res (1989) 21(2):106–111. [DOI] [PubMed] [Google Scholar]
- 170.Tadayoni R, Paques M, Gaudric A, et al. Erythrocyte and leukocyte dynamics in the retinal capillaries of diabetic mice. Exp Eye Res (2003) 77(4):497–504. [DOI] [PubMed] [Google Scholar]
- 171.Ellis EA, Guberski DL, Hutson B, et al. Time course of nadh oxidase, inducible nitric oxide synthase and peroxynitrite in diabetic retinopathy in the bbz/wor rat. Nitric oxide : biology and chemistry (2002) 6(3):295–304. [DOI] [PubMed] [Google Scholar]
- 172.Rein DB, Wittenborn JS, Zhang X, et al. Forecasting age-related macular degeneration through the year 2050: The potential impact of new treatments. Arch Ophthalmol (2009) 127(4):533–540. [DOI] [PubMed] [Google Scholar]
- 173.Klein R, Cruickshanks KJ, Myers CE, et al. The relationship of atherosclerosis to the 10-year cumulative incidence of age-related macular degeneration: The beaver dam studies. Ophthalmology (2013) 120(5):1012–1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Bhutto I, Lutty G. Understanding age-related macular degeneration (amd): Relationships between the photoreceptor/retinal pigment epithelium/bruch’s membrane/choriocapillaris complex. Mol Aspects Med (2012) 33(4):295–317.* Thorough review of AMD.
- 175.Malek G, Lad EM. Emerging roles for nuclear receptors in the pathogenesis of age-related macular degeneration. Cell Mol Life Sci (2014) 71(23):4617–4636.** Review of AMD pathology and nuclear receptors as potential regulators of disease development.
- 176.Sohrab MA, Smith RT, Fawzi AA. Imaging characteristics of dry age-related macular degeneration. Semin Ophthalmol (2011) 26(3):156–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Curcio CA, Millican CL. Basal linear deposit and large drusen are specific for early age-related maculopathy. Arch Ophthalmol (1999) 117(3):329–339. [DOI] [PubMed] [Google Scholar]
- 178.Green WR, Enger C. Age-related macular degeneration histopathologic studies. The 1992 lorenz e. Zimmerman lecture. Ophthalmology (1993) 100(10):1519–1535. [DOI] [PubMed] [Google Scholar]
- 179.Loffler KU, Lee WR. Basal linear deposit in the human macula. Graefes Arch Clin Exp Ophthalmol (1986) 224(6):493–501. [DOI] [PubMed] [Google Scholar]
- 180.Curcio CA, Medeiros NE, Millican CL. Photoreceptor loss in age-related macular degeneration. Invest Ophthalmol Vis Sci (1996) 37(7):1236–1249. [PubMed] [Google Scholar]
- 181.Ambati J, Fowler BJ. Mechanisms of age-related macular degeneration. Neuron (2012) 75(1):26–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Lim LS, Mitchell P, Seddon JM, et al. Age-related macular degeneration. Lancet (2012) 379(9827):1728–1738. [DOI] [PubMed] [Google Scholar]
- 183.Ng EW, Adamis AP. Targeting angiogenesis, the underlying disorder in neovascular age-related macular degeneration. Can J Ophthalmol (2005) 40(3):352–368. [DOI] [PubMed] [Google Scholar]
- 184.Johnson LV, Forest DL, Banna CD, et al. Cell culture model that mimics drusen formation and triggers complement activation associated with age-related macular degeneration. Proc Natl Acad Sci U S A (2011) 108(45):18277–18282.** First in vitro culture of ‘drusen’ using human fetal RPE cells.
- 185.Pilgrim MG, Lengyel I, Lanzirotti A, et al. Subretinal pigment epithelial deposition of drusen components including hydroxyapatite in a primary cell culture model. Invest Ophthalmol Vis Sci (2017) 58(2):708–719.** Detailed model of ‘drusen’ in culture using porcine RPE cells.
- 186.Galloway CA, Dalvi S, Hung SSC, et al. Drusen in patient-derived hipsc-rpe models of macular dystrophies. Proc Natl Acad Sci U S A (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Fletcher EL, Jobling AI, Greferath U, et al. Studying age-related macular degeneration using animal models. Optom Vis Sci (2014) 91(8):878–886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Pennesi ME, Neuringer M, Courtney RJ. Animal models of age related macular degeneration. Mol Aspects Med (2012) 33(4):487–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Grossniklaus HE, Kang SJ, Berglin L. Animal models of choroidal and retinal neovascularization. Prog Retin Eye Res (2010) 29(6):500–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Ramkumar HL, Zhang J, Chan CC. Retinal ultrastructure of murine models of dry age-related macular degeneration (amd). Prog Retin Eye Res (2010) 29(3):169–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Zeiss CJ. Animals as models of age-related macular degeneration: An imperfect measure of the truth. Vet Pathol (2010) 47(3):396–413. [DOI] [PubMed] [Google Scholar]
- 192.Edwards AO, Malek G. Molecular genetics of amd and current animal models. Angiogenesis (2007) 10(2):119–132. [DOI] [PubMed] [Google Scholar]
- 193.Marmorstein AD, Marmorstein LY. The challenge of modeling macular degeneration in mice. Trends Genet (2007) 23(5):225–231. [DOI] [PubMed] [Google Scholar]
- 194.Fritsche LG, Fariss RN, Stambolian D, et al. Age-related macular degeneration: Genetics and biology coming together. Annual review of genomics and human genetics (2014) 15(151–171.* Review of AMD with consideration of biology and genetics.
- 195.Gerth C The role of the erg in the diagnosis and treatment of age-related macular degeneration. Documenta ophthalmologica Advances in ophthalmology (2009) 118(1):63–68. [DOI] [PubMed] [Google Scholar]
- 196.Cai J, Nelson KC, Wu M, et al. Oxidative damage and protection of the rpe. Prog Retin Eye Res (2000) 19(2):205–221. [DOI] [PubMed] [Google Scholar]
- 197.Wenzel A, Grimm C, Samardzija M, et al. Molecular mechanisms of light-induced photoreceptor apoptosis and neuroprotection for retinal degeneration. Prog Retin Eye Res (2005) 24(2):275–306. [DOI] [PubMed] [Google Scholar]
- 198.Cruickshanks KJ, Hamman RF, Klein R, et al. The prevalence of age-related maculopathy by geographic region and ethnicity. The colorado-wisconsin study of age-related maculopathy. Arch Ophthalmol (1997) 115(2):242–250. [DOI] [PubMed] [Google Scholar]
- 199.Vingerling JR, Hofman A, Grobbee DE, et al. Age-related macular degeneration and smoking. The rotterdam study. Arch Ophthalmol (1996) 114(10):1193–1196. [DOI] [PubMed] [Google Scholar]
- 200.Behndig A, Svensson B, Marklund SL, et al. Superoxide dismutase isoenzymes in the human eye. Invest Ophthalmol Vis Sci (1998) 39(3):471–475. [PubMed] [Google Scholar]
- 201.Imamura Y, Noda S, Hashizume K, et al. Drusen, choroidal neovascularization, and retinal pigment epithelium dysfunction in sod1-deficient mice: A model of age-related macular degeneration. Proc Natl Acad Sci U S A (2006) 103(30):11282–11287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Crabb JW, Miyagi M, Gu X,et al. Drusen proteome analysis: An approach to the etiology of age-related macular degeneration. Proc Natl Acad Sci U S A (2002) 99(23):14682–14687.* The first study to systematically investigate the composition of drusen.
- 203.Hashizume K, Hirasawa M, Imamura Y, et al. Retinal dysfunction and progressive retinal cell death in sod1-deficient mice. Am J Pathol (2008) 172(5):1325–1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Kimura K, Isashiki Y, Sonoda S, et al. Genetic association of manganese superoxide dismutase with exudative age-related macular degeneration. Am J Ophthalmol (2000) 130(6):769–773. [DOI] [PubMed] [Google Scholar]
- 205.Li Y, Huang TT, Carlson EJ, et al. Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat Genet (1995) 11(4):376–381. [DOI] [PubMed] [Google Scholar]
- 206.Justilien V, Pang JJ, Renganathan K, et al. Sod2 knockdown mouse model of early amd. Invest Ophthalmol Vis Sci (2007) 48(10):4407–4420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Cousins SW, Espinosa-Heidmann DG, Alexandridou A, et al. The role of aging, high fat diet and blue light exposure in an experimental mouse model for basal laminar deposit formation. Exp Eye Res (2002) 75(5):543–553. [DOI] [PubMed] [Google Scholar]
- 208.Espinosa-Heidmann DG, Suner IJ, Catanuto P, et al. Cigarette smoke-related oxidants and the development of sub-rpe deposits in an experimental animal model of dry amd. Invest Ophthalmol Vis Sci (2006) 47(2):729–737. [DOI] [PubMed] [Google Scholar]
- 209.Fujihara M, Nagai N, Sussan TE, et al. Chronic cigarette smoke causes oxidative damage and apoptosis to retinal pigmented epithelial cells in mice. PLoS One (2008) 3(9):e3119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Wang AL, Neufeld AH. Smoking mice: A potential model for studying accumulation of drusen-like material on bruch’s membrane. Vision research (2010) 50(7):638–642. [DOI] [PubMed] [Google Scholar]
- 211.Hu P, Herrmann R, Bednar A, et al. Aryl hydrocarbon receptor deficiency causes dysregulated cellular matrix metabolism and age-related macular degeneration-like pathology. Proc Natl Acad Sci U S A (2013) 110(43):E4069–4078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Kim SY, Yang HJ, Chang YS, et al. Deletion of aryl hydrocarbon receptor ahr in mice leads to subretinal accumulation of microglia and rpe atrophy. Invest Ophthalmol Vis Sci (2014) 55(9):6031–6040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Choudhary M, Kazmin D, Hu P, et al. Aryl hydrocarbon receptor knock-out exacerbates choroidal neovascularization via multiple pathogenic pathways. J Pathol (2015) 235(1):101–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Klein R, Klein BE, Tomany SC, et al. The association of cardiovascular disease with the long-term incidence of age-related maculopathy: The beaver dam eye study. Ophthalmology (2003) 110(6):1273–1280. [DOI] [PubMed] [Google Scholar]
- 215.van Leeuwen R, Ikram MK, Vingerling JR, et al. Blood pressure, atherosclerosis, and the incidence of age-related maculopathy: The rotterdam study. Invest Ophthalmol Vis Sci (2003) 44(9):3771–3777. [DOI] [PubMed] [Google Scholar]
- 216.Curcio CA, Millican CL, Bailey T, et al. Accumulation of cholesterol with age in human bruch’s membrane. Invest Ophthalmol Vis Sci (2001) 42(1):265–274. [PubMed] [Google Scholar]
- 217.Pauleikhoff D, Harper CA, Marshall J, et al. Aging changes in bruch’s membrane. A histochemical and morphologic study. Ophthalmology (1990) 97(2):171–178. [PubMed] [Google Scholar]
- 218.Sheraidah G, Steinmetz R, Maguire J, et al. Correlation between lipids extracted from bruch’s membrane and age. Ophthalmology (1993) 100(1):47–51. [DOI] [PubMed] [Google Scholar]
- 219.Li CM, Chung BH, Presley JB, et al. Lipoprotein-like particles and cholesteryl esters in human bruch’s membrane: Initial characterization. Invest Ophthalmol Vis Sci (2005) 46(7):2576–2586. [DOI] [PubMed] [Google Scholar]
- 220.Mares-Perlman JA, Brady WE, Klein R, et al. Dietary fat and age-related maculopathy. Arch Ophthalmol (1995) 113(6):743–748. [DOI] [PubMed] [Google Scholar]
- 221.Seddon JM, Cote J, Rosner B. Progression of age-related macular degeneration: Association with dietary fat, transunsaturated fat, nuts, and fish intake. Arch Ophthalmol (2003) 121(12):1728–1737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Klaver CC, Kliffen M, van Duijn CM, et al. Genetic association of apolipoprotein e with age-related macular degeneration. Am J Hum Genet (1998) 63(1):200–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Simonelli F, Margaglione M, Testa F, et al. Apolipoprotein e polymorphisms in age-related macular degeneration in an italian population. Ophthalmic Res (2001) 33(6):325–328. [DOI] [PubMed] [Google Scholar]
- 224.Cousins SW, Marin-Castano ME, Espinosa-Heidmann DG, et al. Female gender, estrogen loss, and sub-rpe deposit formation in aged mice. Invest Ophthalmol Vis Sci (2003) 44(3):1221–1229. [DOI] [PubMed] [Google Scholar]
- 225.Plump AS, Smith JD, Hayek T, et al. Severe hypercholesterolemia and atherosclerosis in apolipoprotein e-deficient mice created by homologous recombination in es cells. Cell (1992) 71(2):343–353. [DOI] [PubMed] [Google Scholar]
- 226.Dithmar S, Curcio CA, Le NA, et al. Ultrastructural changes in bruch’s membrane of apolipoprotein e-deficient mice. Invest Ophthalmol Vis Sci (2000) 41(8):2035–2042. [PubMed] [Google Scholar]
- 227.Malek G, Johnson LV, Mace BE, et al. Apolipoprotein e allele-dependent pathogenesis: A model for age-related retinal degeneration. Proc Natl Acad Sci U S A (2005) 102(33):11900–11905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Malek G, Mace B, Saloupis P, et al. Initial observations of key features of age-related macular degeneration in apoe targeted replacement mice. Adv Exp Med Biol (2006) 572(109–117. [DOI] [PubMed] [Google Scholar]
- 229.Sullivan PM, Mezdour H, Aratani Y, et al. Targeted replacement of the mouse apolipoprotein e gene with the common human apoe3 allele enhances diet-induced hypercholesterolemia and atherosclerosis. J Biol Chem (1997) 272(29):17972–17980. [DOI] [PubMed] [Google Scholar]
- 230.Malek G, Li CM, Guidry C, et al. Apolipoprotein b in cholesterol-containing drusen and basal deposits of human eyes with age-related maculopathy. Am J Pathol (2003) 162(2):413–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Espinosa-Heidmann DG, Sall J, Hernandez EP, et al. Basal laminar deposit formation in apo b100 transgenic mice: Complex interactions between dietary fat, blue light, and vitamin e. Invest Ophthalmol Vis Sci (2004) 45(1):260–266. [DOI] [PubMed] [Google Scholar]
- 232.Fujihara M, Bartels E, Nielsen LB, et al. A human apob100 transgenic mouse expresses human apob100 in the rpe and develops features of early amd. Exp Eye Res (2009) 88(6):1115–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Sallo FB, Bereczki E, Csont T, et al. Bruch’s membrane changes in transgenic mice overexpressing the human biglycan and apolipoprotein b-100 genes. Exp Eye Res (2009) 89(2):178–186. [DOI] [PubMed] [Google Scholar]
- 234.Choudhary M, Ding JD, Qi X, et al. Pparbeta/delta selectively regulates phenotypic features of age-related macular degeneration. Aging (Albany NY) (2016) 8(9):1952–1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Priya RR, Chew EY, Swaroop A. Genetic studies of age-related macular degeneration: Lessons, challenges, and opportunities for disease management. Ophthalmology (2012) 119(12):2526–2536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Hageman GS, Anderson DH, Johnson LV, et al. A common haplotype in the complement regulatory gene factor h (hf1/cfh) predisposes individuals to age-related macular degeneration. Proc Natl Acad Sci U S A (2005) 102(20):7227–7232.* First identification of association between CFH and AMD.
- 237.Edwards AO, Ritter R, , Abel KJ, et al. Complement factor h polymorphism and age-related macular degeneration. Science (2005) 308(5720):421–424.* First identification of association between CFH and AMD.
- 238.Klein RJ, Zeiss C, Chew EY, et al. Complement factor h polymorphism in age-related macular degeneration. Science (2005) 308(5720):385–389.* First identification of association between CFH and AMD.
- 239.Haines JL, Hauser MA, Schmidt S, et al. Complement factor h variant increases the risk of age-related macular degeneration. Science (2005) 308(5720):419–421.* First identification of association between CFH and AMD.
- 240.Mullins RF, Russell SR, Anderson DH, et al. Drusen associated with aging and age-related macular degeneration contain proteins common to extracellular deposits associated with atherosclerosis, elastosis, amyloidosis, and dense deposit disease. FASEB J (2000) 14(7):835–846. [PubMed] [Google Scholar]
- 241.Lee AY, Raya AK, Kymes SM, et al. Pharmacogenetics of complement factor h (y402h) and treatment of exudative age-related macular degeneration with ranibizumab. Br J Ophthalmol (2009) 93(5):610–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Chen H, Yu KD, Xu GZ. Association between variant y402h in age-related macular degeneration (amd) susceptibility gene cfh and treatment response of amd: A meta-analysis. PLoS One (2012) 7(8):e42464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Coffey PJ, Gias C, McDermott CJ, et al. Complement factor h deficiency in aged mice causes retinal abnormalities and visual dysfunction. Proc Natl Acad Sci U S A (2007) 104(42):16651–16656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Williams JA, Greenwood J, Moss SE. Retinal changes precede visual dysfunction in the complement factor h knockout mouse. PLoS One (2013) 8(7):e68616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Ufret-Vincenty RL, Aredo B, Liu X, et al. Transgenic mice expressing variants of complement factor h develop amd-like retinal findings. Invest Ophthalmol Vis Sci (2010) 51(11):5878–5887. [DOI] [PubMed] [Google Scholar]
- 246.Toomey CB, Kelly U, Saban DR, et al. Regulation of age-related macular degeneration-like pathology by complement factor h. Proc Natl Acad Sci U S A (2015) 112(23):E3040–3049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Ding JD, Kelly U, Landowski M, et al. Expression of human complement factor h prevents age-related macular degeneration-like retina damage and kidney abnormalities in aged cfh knockout mice. Am J Pathol (2015) 185(1):29–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Lyzogubov VV, Tytarenko RG, Jha P, et al. Role of ocular complement factor h in a murine model of choroidal neovascularization. Am J Pathol (2010) 177(4):1870–1880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Rohrer B, Long Q, Coughlin B, et al. A targeted inhibitor of the alternative complement pathway reduces angiogenesis in a mouse model of age-related macular degeneration. Invest Ophthalmol Vis Sci (2009) 50(7):3056–3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Guymer RH, Tao LW, Goh JK, et al. Identification of urinary biomarkers for age-related macular degeneration. Invest Ophthalmol Vis Sci (2011) 52(7):4639–4644. [DOI] [PubMed] [Google Scholar]
- 251.Combadiere C, Feumi C, Raoul W, et al. Cx3cr1-dependent subretinal microglia cell accumulation is associated with cardinal features of age-related macular degeneration. J Clin Invest (2007) 117(10):2920–2928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Tuo J, Bojanowski CM, Zhou M, et al. Murine ccl2/cx3cr1 deficiency results in retinal lesions mimicking human age-related macular degeneration. Invest Ophthalmol Vis Sci (2007) 48(8):3827–3836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Vessey KA, Greferath U, Jobling AI, et al. Ccl2/cx3cr1 knockout mice have inner retinal dysfunction but are not an accelerated model of amd. Invest Ophthalmol Vis Sci (2012) 53(12):7833–7846. [DOI] [PubMed] [Google Scholar]
- 254.Staton CA, Reed MW, Brown NJ. A critical analysis of current in vitro and in vivo angiogenesis assays. Int J Exp Pathol (2009) 90(3):195–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Tao Y, Jiang P, Wei Y, et al. Alpha-lipoic acid treatment improves vision-related quality of life in patients with dry age-related macular degeneration. The Tohoku journal of experimental medicine (2016) 240(3):209–214. [DOI] [PubMed] [Google Scholar]
- 256.Ramkumar HL, Tuo J, Shen DF, et al. Nutrient supplementation with n3 polyunsaturated fatty acids, lutein, and zeaxanthin decrease a2e accumulation and vegf expression in the retinas of ccl2/cx3cr1-deficient mice on crb1rd8 background. The Journal of nutrition (2013) 143(7):1129–1135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Shen D, Cao X, Zhao L, et al. Naloxone ameliorates retinal lesions in ccl2/cx3cr1 double-deficient mice via modulation of microglia. Invest Ophthalmol Vis Sci (2011) 52(6):2897–2904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Kim SJ, Lee HJ, Yun JH, et al. Intravitreal tsg-6 suppresses laser-induced choroidal neovascularization by inhibiting ccr2+ monocyte recruitment. Scientific reports (2015) 5(11872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Ren X, Li J, Xu X, et al. Ibi302, a promising candidate for amd treatment, targeting both the vegf and complement system with high binding affinity in vitro and effective targeting of the ocular tissue in healthy rhesus monkeys. Exp Eye Res (2016) 145(352–358. [DOI] [PubMed] [Google Scholar]
- 260.Jo DH, Kim JH, Yang W, et al. Anti-complement component 5 antibody targeting mg4 domain inhibits choroidal neovascularization. Oncotarget (2017) 8(28):45506–45516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Loyet KM, Good J, Davancaze T, et al. Complement inhibition in cynomolgus monkeys by anti-factor d antigen-binding fragment for the treatment of an advanced form of dry age-related macular degeneration. The Journal of pharmacology and experimental therapeutics (2014) 351(3):527–537. [DOI] [PubMed] [Google Scholar]