The role of omega-3 long-chain polyunsaturated fatty acids in health and disease of the retina

https://doi.org/10.1016/j.preteyeres.2004.06.002Get rights and content

Abstract

In this work we advance the hypothesis that omega-3 (ω-3) long-chain polyunsaturated fatty acids (LCPUFAs) exhibit cytoprotective and cytotherapeutic actions contributing to a number of anti-angiogenic and neuroprotective mechanisms within the retina. ω-3 LCPUFAs may modulate metabolic processes and attenuate effects of environmental exposures that activate molecules implicated in pathogenesis of vasoproliferative and neurodegenerative retinal diseases. These processes and exposures include ischemia, chronic light exposure, oxidative stress, inflammation, cellular signaling mechanisms, and aging. A number of bioactive molecules within the retina affect, and are effected by such conditions. These molecules operate within complex systems and include compounds classified as eicosanoids, angiogenic factors, matrix metalloproteinases, reactive oxygen species, cyclic nucleotides, neurotransmitters and neuromodulators, pro-inflammatory and immunoregulatory cytokines, and inflammatory phospholipids. We discuss the relationship of LCPUFAs with these bioactivators and bioactive compounds in the context of three blinding retinal diseases of public health significance that exhibit both vascular and neural pathology.

How is ω-3 LCPUFA status related to retinal structure and function? Docosahexaenoic acid (DHA), a major dietary ω-3 LCPUFA, is also a major structural lipid of retinal photoreceptor outer segment membranes. Biophysical and biochemical properties of DHA may affect photoreceptor membrane function by altering permeability, fluidity, thickness, and lipid phase properties. Tissue DHA status affects retinal cell signaling mechanisms involved in phototransduction. DHA may operate in signaling cascades to enhance activation of membrane-bound retinal proteins and may also be involved in rhodopsin regeneration. Tissue DHA insufficiency is associated with alterations in retinal function. Visual processing deficits have been ameliorated with DHA supplementation in some cases.

What evidence exists to suggest that LCPUFAs modulate factors and processes implicated in diseases of the vascular and neural retina? Tissue status of LCPUFAs is modifiable by and dependent upon dietary intake. Certain LCPUFAs are selectively accreted and efficiently conserved within the neural retina. On the most basic level, ω-3 LCPUFAs influence retinal cell gene expression, cellular differentiation, and cellular survival. DHA activates a number of nuclear hormone receptors that operate as transcription factors for molecules that modulate reduction-oxidation-sensitive and proinflammatory genes; these include the peroxisome proliferator-activated receptor-α (PPAR-α) and the retinoid X receptor. In the case of PPAR-α, this action is thought to prevent endothelial cell dysfunction and vascular remodeling through inhibition of: vascular smooth muscle cell proliferation, inducible nitric oxide synthase production, interleukin-1 induced cyclooxygenase (COX)-2 production, and thrombin-induced endothelin 1 production.

Research on model systems demonstrates that ω-3 LCPUFAs also have the capacity to affect production and activation of angiogenic growth factors, arachidonic acid (AA)-based vasoregulatory eicosanoids, and MMPs. Eicosapentaenoic acid (EPA), a substrate for DHA, is the parent fatty acid for a family of eicosanoids that have the potential to affect AA-derived eicosanoids implicated in abnormal retinal neovascularization, vascular permeability, and inflammation. EPA depresses vascular endothelial growth factor (VEGF)—specific tyrosine kinase receptor activation and expression. VEGF plays an essential role in induction of: endothelial cell migration and proliferation, microvascular permeability, endothelial cell release of metalloproteinases and interstitial collagenases, and endothelial cell tube formation. The mechanism of VEGF receptor down-regulation is believed to occur at the tyrosine kinase nuclear factor-kappa B (NFκB). NFκB is a nuclear transcription factor that up-regulates COX-2 expression, intracellular adhesion molecule, thrombin, and nitric oxide synthase. All four factors are associated with vascular instability. COX-2 drives conversion of AA to a number angiogenic and proinflammatory eicosanoids. Our general conclusion is that there is consistent evidence to suggest that ω-3 LCPUFAs may act in a protective role against ischemia-, light-, oxygen-, inflammatory-, and age-associated pathology of the vascular and neural retina.

Introduction

Long-chain polyunsaturated fatty acids (LCPUFAs) demonstrate anti-angiogenic, anti-vasoproliferative, and neuroprotective actions on factors and processes implicated in the pathogenesis of proliferative and degenerative retinal diseases. Many retinal diseases of public health significance manifest tissue and cellular dysfunction in the forms of abnormal angiogenesis, proliferative neovascularization, excessive vascular permeability, immunoregulatory dysfunction, alterations in physiologic reduction-oxidation (redox) balance, or neuronal/retinal pigment epithelial (RPE) cell degeneration. A number of bioactive molecules within the eye affect, and are effected by, such conditions. These molecules are activated in response to ischemia, light exposure, oxygen/energy metabolism and oxidative stress, apoptosis, cell signaling pathways, inflammation, and developmental processes associated with aging. They operate within complex systems and include eicosanoids, angiogenic factors, matrix metalloproteinases (MMPs), reactive oxygen species, cyclic nucleotides, neurotransmitters and neuromodulators, pro-inflammatory and immunoregulatory cytokines, and inflammatory phospholipids. Effects and actions of metabolic and environmental bioactivators and bioactive molecules include activation of phospholipase A2 (PLA2), cyclooxgenase (COX), and lipoxygenase (LOX). Activation of this enzyme system yields a pool of LCPUFAs and bioactive eicosanoids.

Omega-3 (ω-3) LCPUFAs demonstrate the capacity to modulate production, activation, and potency of bioactive molecules. In some cases these LCPUFAs operate as lipid–protein complexes via signaling cascades in nuclear and cytosolic compartments. In others, they affect substrate pools or availability of biosynthetic enzymes. They influence gene expression as ligands to a number of transcription factors and act as endocannabinoid autocoids. Docosahexaenoic acid (DHA, C22:6ω-3), a major dietary ω-3 LCPUFA, is also a major structural lipid in sensory and vascular retina. Metabolic and dietary DHA insufficiency is associated with alterations in visual system structure and function. DHA and its substrate, eicosapentaenoic acid (EPA, C20:5ω-3), influence eicosanoid metabolism by reducing ω-6 LCPUFA levels (mainly arachidonic acid (C20:4ω-6, AA)) and competing for enzymes (COX and LOX) used to produce AA-based angiogenic and proinflammatory series 2- and 4-eicosanoids.

In this work we present the body evidence implicating LCPUFAs as key modulators of processes influencing retinal health and disease. Section 2 contains a general overview of properties, functions, and actions of LCPUFAs; a more detailed treatment of the issue appears in Chow (2000). Section 3 contains an overview of LCPUFA metabolism, intake, transport, and accretion to the retina; additional information exists in Neuringer (1993), Salem et al. (2001), and Bazan et al. (1993). In Section 4 we consider actions of LCPUFAs on biochemical and biophysical processes that define properties of retinal membranes and signaling systems. Section 5 contains information on metabolic and environmental factors and processes that activate molecules driving retinal neovascularization and neural cell death. These bioactivating factors include ischemia, chronic light exposure, cellular redox balance, cell death, inflammation, neuroactive signaling molecules, and the aging process. In Section 6 we consider the role of LCPUFAs in the structure and function of the vascular retina. In Section 7 we consider the means by which ω-3 LCPUFAs may operate as protective factors in retinal diseases that manifest vascular and neural pathology; we present three examples: diabetic retinopathy (DR), age-related macular degeneration (AMD), and retinopathy of prematurity (ROP). These diseases were selected on the basis of life-span risk, the burden they exert on society, and the coexistence of vascular and neural degenerative pathologies. Our general conclusion is that there is consistent evidence to suggest that ω-3 LCPUFAs may act in a protective role against ischemia-, light-, oxygen-, inflammatory-, or age-associated retinal diseases. Section conclusions are displayed in Table 1.

Section snippets

DHA, EPA, and AA are LCPUFAs

Fatty acids are compounds synthesized through condensation of malonyl coenzyme A units by a fatty acid synthase complex. Two families of essential fatty acids (EFAs) exist in nature; ω-3 and ω-6. ω-3 and ω-6 LCPUFAs contain a carboxyl head group and an even numbered carbon chain (⩾18 carbons) with two-or-more methylene-interrupted double (unsaturated) bonds. EFAs and LCPUFAs are structurally classified by the number of carbons, double bonds, and proximity of the first double bond to the methyl

LCPUFAs are obtained through diet or biosynthesized from EFAs

Humans do not have capacity for de novo biosynthesis of EFAs (α-linolenic and linoleic acid, LA), due to a natural absence of Δ-15 and-12 desaturase enzymes. We are thus dependent on dietary sources of these compounds. LCPUFAs may be obtained directly through the diet or formed from 18-carbon EFAs. Enzymatic reactions yielding LCPUFAs do not satisfy the body's requirements.

After EFAs are obtained through the diet they are desaturated (by insertion of double bonds) and elongated (by addition of

Role of LCPUFAs in structure and function of sensory retina

The contents of this section describe the role of LCPUFAs in the structure and function of the sensory retina. Neuringer et al. (forthcoming publication in Progress in Retinal and Eye Research) review these issues in detail.

Metabolic and environmental bioactivators

A number of metabolic and environmental factors and processes serve as bioactivators of molecules associated with abnormal angiogenesis, proliferative neovascularization, excessive vascular permeability, immunoregulatory dysfunction, alterations in physiologic redox balance, and neuronal/RPE cell degeneration. Key factors and processes affecting the retina include ischemia, light exposure, oxidative stress, apoptosis, inflammation, neuroactive cell signaling molecules, and developmental

Role of LCPUFAs in structure and function of vascular retina

A rapidly amassing evidence base suggests ω-3 LCPUFAs exert anti-angiogenic and -vasculogenic properties through modulation of processes involved in intracellular signaling, activation of transcription factors, and production of inflammatory mediators. Since ω-3 LCPUFA tissue status is modifiable by and dependent on dietary intake from foods that are not commonly consumed in the Western diet, these nutrients may be reasonable choices for diet- or supplement-based interventions to prevent

Retinal diseases of public health significance

The factors and processes that activate PLA2, COX, LOX, and the bioactive molecules discussed above are associated with a number of retinal diseases of public health significance. These diseases usually manifest both vascular- and neural pathology. Here we discuss DR, AMD, and ROP.

Summary and future directions

The importance of LCPUFAs in the retina is indicated by the efficient conservation and use of these ‘easily oxidized’ lipids in areas that are both highly susceptible to oxidative stress and chonically exposed to conditions that facilitate production of reactive oxygen species (Gordon and Bazan, 1997). Conditions associated with activation of enzymes essential for liberating and moblizing LCPUFAs from tissue stores and then converting these comounds to eicosanoids are also associated with

Acknowledgements

We would like to acknowledge Drs. Norman Salem, Martha Neuringer, Burton Litman, and Joseph Hibbeln for generously discussing practical and theoretical aspects of their research and other applied research on LCPUFAs in human health and disease. I would like to thank Dr. Frank Sacks for first exposing me to work describing innovative mechanistic approaches for investigating the functions and actions of LCPUFAs in living systems (JPSG).

References (447)

  • J. Balsinde et al.

    Distinct roles in signal transduction for each of the phospholipase A(2) enzymes present in P388D(1) macrophages

    J. Biol. Chem.

    (1996)
  • A.J. Barber

    A new view of diabetic retinopathya neurodegenerative disease of the eye

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2003)
  • U. Barcelli et al.

    Enhancing effect of dietary supplementation with omega-3 fatty acids on plasma fibrinolysis in normal subjects

    Thromb. Res.

    (1985)
  • Y. Bayon et al.

    Effect of specific phospholipid molecular species incorporated in human platelet membranes on thromboxane A2/prostaglandin H2 receptors

    J. Lipid Res.

    (1995)
  • N.G. Bazan et al.

    Prostaglandins and other lipid mediators in Alzheimer's disease

    Prostaglandins Other Lipid Mediat.

    (2002)
  • S. Beatty et al.

    The role of oxidative stress in the pathogenesis of age-related macular degeneration

    Surv. Ophthalmol.

    (2000)
  • N. Bernoud et al.

    Astrocytes are mainly responsible for the polyunsaturated fatty acid enrichment in blood-brain barrier endothelial cells in vitro

    J. Lipid Res.

    (1998)
  • A. Bonanome et al.

    n-3 fatty acids do not enhance LDL susceptibility to oxidation in hypertriacylglycerolemic hemodialyzed subjects

    Am. J. Clin. Nutr.

    (1996)
  • M. Boulton et al.

    Retinal photodamage

    J. Photochem. Photobiol. B.

    (2001)
  • M.F. Brown

    Modulation of rhodopsin function by properties of the membrane bilayer

    Chem. Phys. Lipids

    (1994)
  • R.A. Bush et al.

    Light damage in the rat retinathe effect of dietary deprivation of N-3 fatty acids on acute structural alterations

    Exp. Eye Res.

    (1991)
  • G.E. Caughey et al.

    The effect on human tumor necrosis factor alpha and interleukin 1 beta production of diets enriched in n-3 fatty acids from vegetable oil or fish oil

    Am. J. Clin. Nutr.

    (1996)
  • B. Chandrasekar et al.

    Decreased pro-inflammatory cytokines and increased antioxidant enzyme gene expression by omega-3 lipids in murine lupus nephritis

    Biochem. Biophys. Res. Commun.

    (1994)
  • H. Chen et al.

    Differential incorporation of docosahexaenoic and arachidonic acids in frog retinal pigment epithelium

    J. Lipid Res.

    (1993)
  • L.Y. Chen et al.

    Reduction in human neutrophil superoxide anion generation by n-3 polyunsaturated fatty acidsrole of cyclooxygenase products and endothelium-derived relaxing factor

    Thromb. Res.

    (1994)
  • Y. Chen et al.

    Docosahexaenoic acid modulates the interactions of the interphotoreceptor retinoid-binding protein with 11-cis-retinal

    J. Biol. Chem.

    (1996)
  • E. Cho et al.

    Prospective study of dietary fat and the risk of age-related macular degeneration

    Am. J. Clin. Nutr.

    (2001)
  • H.G. Choe et al.

    Unique molecular species composition of glycerolipids of frog rod outer segments

    Exp. Eye Res.

    (1990)
  • M.T. Clandinin et al.

    Relationship between fatty acid accretion, membrane composition, and biologic functions

    J. Pediatr.

    (1994)
  • E.S. Collie-Duguid et al.

    Inhibitory effect of fish oil N-3 polyunsaturated fatty acids on the expression of endothelial cell adhesion molecules

    Biochem. Biophys. Res. Commun.

    (1996)
  • J.A. Conquer et al.

    Supplementation with an algae source of docosahexaenoic acid increases (n-3) fatty acid status and alters selected risk factors for heart disease in vegetarian subjects

    J. Nutr.

    (1996)
  • A.L. Cooper et al.

    Effect of dietary fish oil supplemenation on fever and cytokine production in human volunteers

    Clin. Nutr.

    (1993)
  • F.J. Daemen

    Vertebrate rod outer segment membranes

    Biochim. Biophys. Acta

    (1973)
  • J.B. D’Ambola et al.

    Effect of dietary (n-3) and (n-6) fatty acids on in vivo pulmonary bacterial clearance by neonatal rabbits

    J. Nutr.

    (1991)
  • A. Das et al.

    Retinal and choroidal angiogenesispathophysiology and strategies for inhibition

    Prog. Retina Eye Res.

    (2003)
  • S. Davies et al.

    Photocytotoxicity of lipofuscin in human retinal pigment epithelial cells

    Free Radic. Biol. Med.

    (2001)
  • R. De Caterina et al.

    Fatty acid modulation of endothelial activation

    Am. J. Clin. Nutr.

    (2000)
  • R. De Caterina et al.

    Antiarrhythmic effects of omega-3 fatty acidsfrom epidemiology to bedside

    Am. Heart J.

    (2003)
  • J.P. de la Cruz et al.

    Effect of WEB 2086-BS, an antagonist of platelet-activating factor receptors, on retinal vascularity in diabetic rats

    Eur. J. Pharmacol.

    (1998)
  • M.C. de Wilde et al.

    The effect of n-3 polyunsaturated fatty acid-rich diets on cognitive and cerebrovascular parameters in chronic cerebral hypoperfusion

    Brain Res.

    (2002)
  • P. Delerive et al.

    Hypoxia-reoxygenation and polyunsaturated fatty acids modulate adrenergic functions in cultured cardiomyocytes

    J. Mol. Cell. Cardiol.

    (1999)
  • S. Delion et al.

    Age-related changes in phospholipid fatty acid composition and monoaminergic neurotransmission in the hippocampus of rats fed a balanced or an n-3 polyunsaturated fatty acid-deficient diet

    J. Lipid Res.

    (1997)
  • I. Delton-Vandenbroucke et al.

    Polyunsaturated fatty acid metabolism in retinal and cerebral microvascular endothelial cells

    J. Lipid Res.

    (1997)
  • L. Demaison et al.

    Influence of the phospholipid n-6/n-3 polyunsaturated fatty acid ratio on the mitochondrial oxidative metabolism before and after myocardial ischemia

    Biochim. Biophys. Acta

    (1994)
  • T. Abribat et al.

    Decreased serum insulin-like growth factor-I in burn patientsrelationship with serum insulin-like growth factor binding protein-3 proteolysis and the influence of lipid composition in nutritional support

    Crit. Care Med.

    (2000)
  • G.D. Aguirre et al.

    Diets enriched in docosahexaenoic acid fail to correct progressive rod-cone degeneration (prcd) phenotype

    Invest. Ophthalmol. Vis. Sci.

    (1997)
  • L.P. Aiello

    Clinical implications of vascular growth factors in proliferative retinopathies

    Curr. Opin. Ophthalmol.

    (1997)
  • M. Akbar et al.

    Protective effects of docosahexaenoic acid in staurosporine-induced apoptosisinvolvement of phosphatidylinositol-3 kinase pathway

    J. Neurochem.

    (2002)
  • S. Akiba et al.

    Involvement of lipoxygenase pathway in docosapentaenoic acid-induced inhibition of platelet aggregation

    Biol. Pharm. Bull.

    (2000)
  • N.J. Alexander et al.

    Dietary fat modulation of in vitro lymphocyte function

    Ann. Nutr. Metab.

    (1988)
  • Cited by (666)

    View all citing articles on Scopus
    View full text