Elsevier

Peptides

Volume 36, Issue 1, July 2012, Pages 142-150
Peptides

Review
The retinal renin–angiotensin system: Roles of angiotensin II and aldosterone

https://doi.org/10.1016/j.peptides.2012.04.008Get rights and content

Abstract

In the present review we examine the experimental and clinical evidence for the presence of a local renin–angiotensin system within the retina. Interest in a pathogenic role for the renin–angiotensin system in retinal disease originally stemmed from observations that components of the pathway were elevated in retina during the development of certain retinal pathologies. Since then, our knowledge about the contribution of the RAS to retinal disease has greatly expanded. We discuss the known functions of the renin–angiotensin system in retinopathy of prematurity and diabetic retinopathy. This includes the promotion of retinal neovascularization, inflammation, oxidative stress and neuronal and glial dysfunction. The contribution of specific components of the renin–angiotensin system is evaluated with a particular focus on angiotensin II and aldosterone and their cognate receptors. The therapeutic utility of inhibiting key components of the renin–angiotensin system is complex, but may hold promise for the prevention and improvement of vision threatening diseases.

Highlights

► Components of the renin–angiotensin system are expressed in both vascular and neuroglial cells. ► Angiotensin receptor and ACE blockade reduces vascular and neuroglial injury in retinopathy. ► A local aldosterone/mineralocorticoid system exists in the retina. ► Aldosterone may influence vascular pathology and glia and ganglion cell function in the retina.

Introduction

In the classical renin–angiotensin system (RAS), prorenin is activated to form renin in juxtaglomerular cells of the kidney (Fig. 1). Renin then binds to the liver-produced angiotensinogen to generate the decapeptide angiotensin I (Ang I; 1–10), which is then hydrolysed by angiotensin converting enzyme (ACE), which may be present in circulation or locally within tissues, to produce the oligopeptide angiotensin II (Ang II, 1–8). By binding to the type 1 angiotensin receptor (AT1R), Ang II is the predominant physiological regulator of blood pressure and is one of the major targets of pharmacological intervention for the treatment of hypertension [117], [120]. Ang II acting via the AT1R is also known to have important roles in the promotion of cellular pathology including apoptosis, hypertrophy, neovascularization, inflammation and fibrosis that may be dependent or independent of its effects on blood pressure [9], [67], [98], [110], [135]. Ang II also triggers the release of aldosterone from the adrenal cortex (Fig. 1). The binding of aldosterone to the mineralocorticoid receptor (MR) contributes to electrolyte and water balance in the body and can also influence heart, kidney and vascular pathology [38], [122]. Cross-talk between Ang II and aldosterone may also potentiate the signaling pathways that are activated by each agent [55], [87].

The RAS is now considered to be far more complex than this classical view of the pathway. Prorenin and renin may also bind to the (pro)renin receptor [(P)RR] [76]. Binding to the (P)RR can elicit angiotensin-independent actions via the activation of promyelocytic zinc finger, protein phosphatidylinositol-3 kinase and mitogen activated protein kinases to influence cellular proliferation and apoptosis [99], [100]. The antagonism of the AT1R within the cascade also requires consideration. Ang I can be cleaved to Ang 1–7, via the ACE homologue, ACE2, or neutral endopeptidase. Ang 1–7 may stimulate the Mas receptor to partially antagonize the effects of the AT1R by promoting vasodilation, the release of nitric oxide and phosphorylation of Akt [25], [118]. Similarly, the binding of Ang II to the type 1 angiotensin receptor (AT2R) has been reported to oppose the actions of the AT1R [54], [88].

The retina is a complex structure involving close anatomical connections between the microvasculature and various types of neurons and glia. The vision threatening pathology that develops in retinal diseases such as retinopathy of prematurity (ROP) and diabetic retinopathy involves changes to the microvasculature (e.g. apoptosis, neovascularization, leakage) as well as glial and neuronal dysfunction. Over the past 20 years, information has accumulated that clearly demonstrates the presence of a local RAS in the retina with components most abundantly expressed on retinal microvessels, glia (e.g. macroglial Müller cells) and neurons (ganglion cells). Components of the RAS have also been identified in other ocular structures such as the choroid and ciliary body. The cellular localization of RAS components in the eye is detailed in Table 1.

In 1996 we reported prorenin and renin to be expressed in Müller cells [10], a macroglial cell that expands almost the entire width of the retina, and closely interacts with the inner retinal microvasculature. Müller cells are viewed to contribute to the neovascularization of ROP and diabetic retinopathy by providing a source of the potent angiogenic and permeability factor, vascular endothelial growth factor (VEGF) [85], [126]. Müller cells also contribute to the formation of epiretinal membranes that can develop in severe diabetic retinopathy [15]. Other components of the RAS including ACE, ACE2, Ang 1–7, Ang II and AT1R, have also been localized to retinal Müller cells in a variety of species [28], [51], [102], [119], [129]. Other retinal glia such as astrocytes and microglia have been reported to express RAS components (Table 1). With respect to neurons, almost all components of the RAS have been localized to ganglion cells, which are located at the retinal surface and are involved in ROP via their increased production of VEGF in response to retinal hypoxia [45]. Ganglion cells may degenerate in ROP and diabetic retinopathy to contribute to an overall decline in retinal function [50]. Some RAS components are also expressed in other retinal neurons such as amacrine cells, bipolar cells and photoreceptors (Table 1). Endothelial cells and pericytes of the retinal microvasculature also contain RAS components. Until recently, studies of the retinal RAS have largely focused on the cellular location of Ang II and earlier parts of the RAS pathway. Emerging evidence indicates that a local aldosterone system may exist in the retina, with our report of aldosterone synthase mRNA in whole retina, glia and ganglion cells [24], [133], and the MR located on vascular cells (endothelial cells and pericytes), ganglion cells, glia and retinal pigment epithelium [24], [133]. The presence of 11β-hydroxysteroid dehydrogenase 2 in certain retinal cell types suggests that aldosterone rather than cortisol may influence MR's actions in retina [24], [144].

The impetus for the study of the role of the RAS in the retina was arguably due to observations that components of the pathway were elevated in situations of retinal pathology. For instance, prorenin, renin, ACE and Ang II are increased in the plasma and eyes of individuals with diabetic retinopathy [21], [48], [102], [140] and ROP [139], and in experimental models of these diseases [67], [68]. To date, most studies evaluating a causal role for the RAS in retinopathy have focused on the microvasculature, given that retinal vascular pathology is a major contributor to vision loss. There is evidence that Ang II and aldosterone influence both endothelial cells and pericytes in the retinal microvasculature. In cultured retinal endothelial cells, Ang II modulates both survival and proliferation. For instance, Ang II induced retinal endothelial cell apoptosis [66] and reduced the expression of pigment epithelium derived growth factor mRNA, an anti-angiogenic factor in the retina [133]. Both Ang II and aldosterone have mitogenic effects on retinal endothelial cells [79], [80], [133], and Ang II has been reported to enhance VEGF-stimulated endothelial cell proliferation, which involves angiopoeitin2, Tie2 and protein kinase C [79], [80]. Furthermore, the administration of aldosterone resulted in the exacerbation of pathological neovascularization in experimental ROP [133], and retinal swelling and activation of Müller cells in rats [144].

The direct effects of Ang II on the retinal microvasculature have perhaps best been studied in retinal pericytes. Pericytes are considered to be the vascular smooth muscle cell counterpart for microvessels, and thereby are implicated in the regulation of capillary tone [136]. Pericytes may have additional functions including the maintenance of microvascular homeostasis [136]. For example, in diabetes their demise is considered to be an early sign of diabetic retinopathy. There is convincing evidence that Ang II directly influences retinal pericytes, by uncoupling them from the microvasculature, via the activation on non-specific cation and calcium-activated chloride channels [49], [143]. Tissue culture studies have revealed that via the AT1R, Ang II can stimulate pericyte migration which involves both transforming growth factor-β and platelet derived growth factor (PDGF) [70], [71]. Additionally, Ang II can also influence pericyte survival, by augmenting pericyte apoptosis, which may involve the advanced glycation end-product pathway [66], [137]. Overall, it is clear that Ang II influences the retinal microvasculature, and emerging evidence suggests similar effects of aldosterone. The effects of Ang II, prorenin and aldosterone and their cognate receptors on non-vascular cells in the retina, however, require further elucidation.

Section snippets

Pathogenesis of retinopathy of prematurity

ROP is a vasoproliferative disorder of the developing retina that mainly occurs in some preterm newborns [6]. ROP is characterized by changes to the immature vasculature of the developing eye, and can be mild with no visual defects to severe with retinal neovascularization and subsequent retinal detachment and blindness. A major factor in the development of ROP is exposure to changes in the concentration of inspired oxygen. Briefly, when premature infants are exposed to high levels of inspired

Pathogenesis of diabetic retinopathy

The incidence of diabetic retinopathy is frightening with a recent study highlighting that the worldwide health burden includes 93 million people having diabetic retinopathy, and 17 million the proliferative and blinding form of the disease [138]. Almost all individuals with type I diabetes will develop retinopathy over a 15–20 year period, and approximately 20–30% will advance to the blinding form of diabetic retinopathy [32]. Greater than 60% of individuals with type 2 diabetes will have

Cellular mechanisms involved in the effects of the retinal RAS

The cellular mechanisms by which the RAS exerts its effects in the retina are still under investigation. It is likely that reactive oxygen species (ROS) are important given Ang II and aldosterone's ability to increase ROS production [34], [101], and the contribution of ROS to cellular damage in both ROP and diabetic retinopathy [37], [63]. A major source of ROS is nicotinamide adenine dinucleotide phosphate (NADPH, NOX). Six homologs of the cytochrome subunit of the phagocyte NOX

Conclusion

Over the past 20 years, a wealth of information has firmly established the existence of a retinal RAS that is widely distributed in both vascular and neuroglial cells. It is clear that Ang II contributes to the development of ROP and diabetic retinopathy with documented evidence of its influence on neovascularization, vascular leakage, inflammation, neuroglial dysfunction, oxidative stress and growth factor production. To be more clearly defined are the effects of prorenin and aldosterone on

Disclosure statement

The authors have nothing to disclose.

Role of the funding source

A National Health and Medical Research Council of Australia (NHMRC) Senior Research Fellowship supports JW-B.

References (144)

  • A. Mataftsi et al.

    Mediators involved in retinopathy of prematurity and emerging therapeutic targets

    Early Hum Dev

    (2011)
  • C.J. Moravski et al.

    The renin–angiotensin system influences ocular endothelial cell proliferation in diabetes: transgenic and interventional studies

    Am J Pathol

    (2003)
  • J.A. Nadal et al.

    Angiotensin II and retinal pericytes migration

    Biochem Biophys Res Commun

    (1999)
  • H. Nakamura et al.

    Pharmacological and pharmacokinetic study of olmesartan medoxomil in animal diabetic retinopathy models

    Eur J Pharmacol

    (2005)
  • Y. Okada et al.

    Increased expression of angiotensin-converting enzyme in retinas of diabetic rats

    Jpn J Ophthalmol

    (2001)
  • A. Patel et al.

    Effects of a fixed combination of perindopril and indapamide on macrovascular and microvascular outcomes in patients with type 2 diabetes mellitus (the ADVANCE trial): a randomised controlled trial

    Lancet

    (2007)
  • F. Praddaude et al.

    Angiotensin II-induced hypertension regulates AT1 receptor subtypes and extracellular matrix turnover in mouse retinal pigment epithelium

    Exp Eye Res

    (2009)
  • Y. Rautureau et al.

    Cross-talk between aldosterone and angiotensin signaling in vascular smooth muscle cells

    Steroids

    (2011)
  • P. Rong et al.

    Renin processing and secretion in adrenal and retina of transgenic (mREN-2)27 rats

    Kidney Int

    (1994)
  • S. Sarlos et al.

    Retinal angiogenesis is mediated by an interaction between the angiotensin type 2 receptor, VEGF, and angiopoietin

    Am J Pathol

    (2003)
  • S. Satofuka et al.

    (Pro)renin receptor promotes choroidal neovascularization by activating its signal transduction and tissue renin–angiotensin system

    Am J Pathol

    (2008)
  • The Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Research Group

    Retinopathy and nephropathy in patients with type 1 diabetes four years after a trial of intensive therapy

    N Engl J Med

    (2000)
  • United Kingdom Prospective Diabetes Study Group

    Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38

    Br Med J

    (1998)
  • M. Al-Shabrawey et al.

    Role of NADPH oxidase in retinal vascular inflammation

    Invest Ophthalmol Vis Sci

    (2008)
  • N. Ashton et al.

    Effect of oxygen on developing retinal vessels with particular reference to the problem of retrolental fibroplasia

    Br J Ophthalmol

    (1954)
  • A.J. Barber et al.

    Neural apoptosis in the retina during experimental and human diabetes. Early onset and effect of insulin

    J Clin Invest

    (1998)
  • K. Bedard et al.

    The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology

    Physiol Rev

    (2007)
  • A. Benigni et al.

    Angiotensin II revisited: new roles in inflammation, immunology and aging

    EMBO Mol Med

    (2010)
  • J.L. Berka et al.

    Renin-containing Muller cells of the retina display endocrine features

    Invest Ophthalmol Vis Sci

    (1995)
  • A.S. Bomback et al.

    The incidence and implications of aldosterone breakthrough

    Nat Clin Pract Nephrol

    (2007)
  • C.R. Brandt et al.

    Renin mRNA is synthesized locally in rat ocular tissues

    Curr Eye Res

    (1994)
  • G.H. Bresnick et al.

    Electroretinographic oscillatory potentials predict progression of diabetic retinopathy. Preliminary report

    Arch Ophthalmol

    (1984)
  • G.H. Bresnick et al.

    Predicting progression to severe proliferative diabetic retinopathy

    Arch Ophthalmol

    (1987)
  • A. Bringmann et al.

    Involvement of Muller glial cells in epiretinal membrane formation

    Graefes Arch Clin Exp Ophthalmol

    (2009)
  • B.V. Bui et al.

    ACE inhibition salvages the visual loss caused by diabetes

    Diabetologia

    (2003)
  • P. Chen et al.

    Role of NADPH oxidase and ANG II in diabetes-induced retinal leukostasis

    Am J Physiol Regul Integr Comp Physiol

    (2007)
  • A.H. Danser et al.

    Angiotensin levels in the eye

    Invest Ophthalmol Vis Sci

    (1994)
  • A.H. Danser et al.

    Renin, prorenin, and immunoreactive renin in vitreous fluid from eyes with and without diabetic retinopathy

    J Clin Endocrinol Metab

    (1989)
  • J. Deinum et al.

    Identification and quantification of renin and prorenin in the bovine eye

    Endocrinology

    (1990)
  • D. Deliyanti et al.

    Neovascularization is attenuated with aldosterone synthase inhibition in rats with retinopathy

    Hypertension

    (2012)
  • M. Dilauro et al.

    Angiotensin-(1–7) and its effects in the kidney

    ScientificWorldJournal

    (2009)
  • L.E. Downie et al.

    Angiotensin type-1 receptor inhibition is neuroprotective to amacrine cells in a rat model of retinopathy of prematurity

    J Comp Neurol

    (2010)
  • L.E. Downie et al.

    AT1 receptor inhibition prevents astrocyte degeneration and restores vascular growth in oxygen-induced retinopathy

    Glia

    (2008)
  • R.O. Estacio et al.

    Effect of blood pressure control on diabetic microvascular complications in patients with hypertension and type 2 diabetes

    Diabetes Care

    (2000)
  • D.L. Feldman et al.

    Effects of aliskiren on blood pressure, albuminuria, and (pro)renin receptor expression in diabetic TG(mRen-2)27 rats

    Hypertension

    (2008)
  • D.S. Fong et al.

    Retinopathy in diabetes

    Diabetes Care

    (2004)
  • M. Fukumoto et al.

    Involvement of angiotensin II-dependent vascular endothelial growth factor gene expression via NADPH oxidase in the retina in a type 2 diabetic rat model

    Curr Eye Res

    (2008)
  • M.J. Gastinger et al.

    Loss of cholinergic and dopaminergic amacrine cells in streptozotocin-diabetic rat and Ins2Akita-diabetic mouse retinas

    Invest Ophthalmol Vis Sci

    (2006)
  • L. Geng et al.

    Angiotensin converting enzyme (ACE) activity in porcine ocular tissue: effects of diet and ACE inhibitors

    J Ocul Pharmacol Ther

    (2003)
  • F. Giacco et al.

    Oxidative stress and diabetic complications

    Circ Res

    (2010)
  • Cited by (77)

    • Nanoparticles as drug delivery agents for managing diabetic retinopathy

      2023, Applications of Multifunctional Nanomaterials
    View all citing articles on Scopus

    This is part of reviews on local renin–angiotensin systems edited by Walmor C. De Mello.

    View full text