Elsevier

Experimental Eye Research

Volume 85, Issue 4, October 2007, Pages 462-472
Experimental Eye Research

Pro-inflammatory cytokines increase reactive oxygen species through mitochondria and NADPH oxidase in cultured RPE cells

https://doi.org/10.1016/j.exer.2007.06.013Get rights and content

Abstract

Reactive oxygen species (ROS) generated during inflammation are believed to play critical roles in various ocular diseases. However, the underlying mechanisms remain poorly understood. We investigated if pro-inflammatory cytokines, tumor necrosis factor (TNF)-α, interleukin-1β (IL-1β), and interferon-γ (IFN-γ), induce ROS in human retinal pigment epithelial (RPE) cells. TNF-α, IL-1β and IFN-γ increased both intracellular and extracellular ROS production in a time- and dose-dependent manner. Thenoyltrifluoroacetone (TTFA), an inhibitor of mitochondrial respiratory chain, blocked TNF-α- and IFN-γ-, but not IL-1β-induced ROS, whereas other two mitochondrial respiratory chain inhibitors, rotenone and antimycin A, had no effect. NADPH oxidase inhibitor (diphenylene iodinium) abolished the ROS production induced by IL-1β or IFN-γ, but not by TNF-α, whereas 6-aminonicotinamide (6AN), an inhibitor of the hexose monophosphate shunt (HMS), had no significant effects on the ROS induced by all three cytokines. ROS scavengers, pyrrolidinedithiocarbamate (PDTC) and N-acetyl-cysteine (NAC), reduced the levels of ROS induced by TNF-α, IL-1β and IFN-γ (P < 0.05). Collectively, these results demonstrate that TNF-α, IL-1β and IFN-γ increase mitochondrial- and NADPH oxidase-generated ROS in human RPE cells.

Introduction

Reactive oxygen species (ROS) are ubiquitous, highly reactive, diffusible molecules, including superoxide anions, hydrogen peroxide, hydroxyl radical, and nitric oxide (Fridovich, 1997). Cells generate ROS intracellularly and may release them extracellularly (Karlsson and Dahlgren, 2002, Kopprasch et al., 2003). While intracellular ROS serve mainly for host defense against infectious agents, redox-sensitive signal transduction, and other cellular processes, the extracellular release of ROS may damage surrounding tissues, potentially promoting inflammatory processes (Duval et al., 2003, Kopprasch et al., 2003). ROS are involved in aging and many diseases such as atherosclerosis, angiogenesis, cancer, diabetes mellitus, neurological degeneration, and tumor invasion (Harris and Shi, 2003, Wu, 2004). ROS also promote oxidative damage in eye disorders including age-related macular degeneration (AMD), cataracts, and uveitis (Rao, 1990, Winkler et al., 1999, Beatty et al., 2000, Cai et al., 2000, Truscott, 2000).

The retinal pigment epithelium (RPE) separates the neural retina from its blood supply in the choroid. In this strategic position, the RPE helps maintain an appropriate environment for photoreceptor function by transporting fluid, ions, and metabolites into and out of the space surrounding the photoreceptor outer segments. The RPE is an important factor in the development of AMD; central vision decreases when RPE cells cease to function properly, causing photoreceptor degeneration or damage in the macula, the portion of the retina used for central vision (Green et al., 1985, Young, 1987, Hageman et al., 2001, Penfold et al., 2001). Several studies have indicated that RPE cells are capable of producing ROS under certain conditions (Dorey et al., 1989, Miceli et al., 1994, Tate et al., 1995a, Wu and Rao, 1999, Yoshida et al., 2003, Kannan et al., 2004, Kindzelskii et al., 2004). RPE cells may therefore be an important source of ROS in the eye. Indeed, focused light (Dorey et al., 1990), high oxygen tension in the macula (Alder and Cringle, 1985), and photoreceptor outer segment phagocytosis and degradation (Tate et al., 1995b, Higgins et al., 2003) all promote RPE oxidative stress (Tso, 1987, Gottsch et al., 1990, Cai et al., 1999, Wassell et al., 1999, Higgins et al., 2003, Kindzelskii et al., 2004). Godley et al. (2005) showed that ROS levels were strikingly higher in macular RPE compared to peripheral RPE. Usually, RPE oxidative stress is reduced by endogenous RPE antioxidants and antioxidant enzymes, such as superoxide dismutase (SOD), catalase and glutathione peroxidase (GPX) (Newsome et al., 1990, Tate et al., 1993, Tate et al., 1995a, Edge et al., 1997, Fukuzawa et al., 1998, Jarrett et al., 2006). With aging, however, these RPE defenses decrease (Tate et al., 1993, Liles et al., 1991, Samiec et al., 1998), permitting ROS to ultimately overwhelm RPE cell defenses, leading to apoptotic damage (Ballinger et al., 1999).

AMD is a degenerative eye disease, common in people over 65 years of age, in which low-grade inflammation is now recognized to play a role. The inflammatory response in AMD lesions is characterized by an infiltration of the blood–retina barrier including RPE layer, by macrophages and lymphocytes (Seregard et al., 1994, Reddy et al., 1995, Penfold et al., 2001, Grossniklaus et al., 2002). In AMD, reactive, migrating or proliferating RPE cells are found adjacent to newly formed vessels in the subretinal space of wet AMD lesions (Miller et al., 1986, Sakamoto et al., 1995). Activation of RPE, inflammatory and endothelial cells may result in the release of a plethora of inflammatory mediators that individually or in concert may induce pathological changes in the retina. Pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin-1β (IL-1β), and interferon-γ (IFN-γ) may be among the primary components responsible for the inflammatory response observed in the AMD, as TNF-α and IL-1β are secreted by macrophages and vescular endothelial cells while IFN-γ is secreted by lymphocytes (Oh et al., 1999). Furthermore, TNF-α, IL-1β, and IFN-γ are linked with pro-inflammatory RPE cell functions (Elner et al., 1990, Elner et al., 1991, Elner et al., 1992, Elner et al., 1997, Hollborn et al., 2001).

TNF-α, IL-1β, and IFN-γ are polypeptides that exert pleiotropic actions on multiple cell functions regulating gene expression, host defense reactions, and the immune response. TNF-α increases mitochondrial ROS production in tumor cells, endothelial cells, and hepatocytes (Schulze-Osthoff et al., 1993, Corda et al., 2001). IL-1β stimulates ROS production in various cell types (Mendes et al., 2003, Brigelius-Flohe et al., 2004, Hwang et al., 2004, Kaur et al., 2004). IFN-γ induces an immediate and marked augmentation of intracellular ROS in transformed lymphoblast cell lines (HSC536/N and PD149L) (Pearl-Yafe et al., 2003, Pearl-Yafe et al., 2004). IFN-γ also induces ROS and endoplasmic reticulum stress during IFN-γ-induced apoptosis of hepatocytes (Watanabe et al., 2003). However, the abilities of TNF-α, IL-1β, or IFN-γ to stimulate ROS production in RPE cells have not yet been reported.

In this study, therefore, we assessed: (1) whether human RPE cells produce ROS when stimulated by pro-inflammatory cytokines, TNF-α, IL-1β, or IFN-γ; and (2) if so, the cellular source of ROS production induced by these cytokines.

Section snippets

Materials

Recombinant human TNF-α, IL-1β, and IFN-γ were purchased from R&D Systems, Inc. (Minneapolis, MN) and PeproTech, Inc. (Rocky Hill, NJ). MitoTracker Red CMXRos and 5-(and 6)-chloromethyl-2′,7′-dichlorodihydrofluorescence diacetate, acetyl ester (CM-H2DCFDA) were purchased from Molecular Probes (Eugene, OR). Poly-d-lysine coated 96-well plates and Hoechst 33342 were purchased from Sigma-Aldrich (St. Louis, MO). All other reagents were purchased from Sigma-Aldrich or Gibco BRL Life Technologies

RPE ROS production is induced by TNF-α, IL-1β or IFN-γ

ROS play an important role in the pathogenesis of various forms of inflammatory ocular injury. Cells generate ROS intracellularly and may release them extracellularly (Karlsson and Dahlgren, 2002, Kopprasch et al., 2003). Therefore, we examined both intracellular and extracellular ROS production in response to cytokines (TNF-α, IL-1β and IFN-γ) in cultured human RPE cells.

As shown in Fig. 1A, TNF-α-induced RPE intracellular ROS levels in a dose-dependent manner and maximal stimulation was

Discussion

This study demonstrates that TNF-α, IL-1β and IFN-γ induce ROS production by human RPE cells in comparison to unstimulated RPE cells. TNF-α increases mitochondrial ROS production in RPE cells; IL-1β induces ROS production via NADPH oxidase; and IFN-γ induces ROS through both mitochondrial ROS and ROS via NADPH oxidase. Furthermore, ROS induced by each cytokine are readily abolished by two ROS scavengers, NAC and PDTC.

Although cytokine-induced intracellular ROS levels are low in RPE, trace

Acknowledgements

This research was supported by the National Institutes of Health grants EY09441 (V.M. Elner) and EY07003 (core). V.M. Elner is a recipient of Lew R. Wasserman Merit Award from Research to Prevent Blindness.

References (92)

  • L. Flohe et al.

    Redox regulation of NF-kappa B activation

    Free Radic. Biol. Med.

    (1997)
  • I. Fridovich

    Superoxide anion radical (O2radical dot), superoxide dismutases, and related matters

    J. Biol. Chem.

    (1997)
  • K. Fukami et al.

    AGEs activate mesangial TGF-beta-Smad signaling via an angiotensin II type I receptor interaction

    Kidney Int.

    (2004)
  • C. Garcia-Ruiz et al.

    Direct effect of ceramide on the mitochondrial electron transport chain leads to generation of reactive oxygen species. Role of mitochondrial glutathione

    J. Biol. Chem.

    (1997)
  • W.R. Green et al.

    Pathologic features of senile macular degeneration

    Ophthalmology

    (1985)
  • K.Z. Guyton et al.

    Activation of mitogen-activated protein kinase by H2O2. Role in cell survival following oxidant injury

    J. Biol. Chem.

    (1996)
  • G.S. Hageman et al.

    An integrated hypothesis that considers drusen as biomarkers of immune-mediated processes at the RPE-Bruch's membrane interface in aging and age related macular degeneration

    Prog. Retin. Eye Res.

    (2001)
  • G.K. Harris et al.

    Signaling by carcinogenic metals and metal-induced reactive oxygen species

    Mutat. Res.

    (2003)
  • A.C. Hermann et al.

    Development of a respiratory burst assay using zebrafish kidneys and embryos

    J. Immunol. Methods

    (2004)
  • G.J. Jaffe et al.

    Expression of interleukin-1 alpha, interleukin-1 beta, and an interleukin-1 receptor antagonist in human retinal pigment epithelial cells

    Exp. Eye Res.

    (1992)
  • Y.D. Jung et al.

    Role of P38 MAPK, AP-1, and NF-kappaB in interleukin-1beta-induced IL-8 expression in human vascular smooth muscle cells

    Cytokine

    (2002)
  • R. Kannan et al.

    Ceramide-induced apoptosis: role of catalase and hepatocyte growth factor

    Free Radic. Biol. Med.

    (2004)
  • A.L. Kindzelskii et al.

    Human, but not bovine, photoreceptor outer segments prime human retinal pigment epithelial cells for metabolic activation and massive oxidant release in response to lipopolysaccharide and interferon-gamma

    Exp. Eye Res.

    (2004)
  • M.V. Miceli et al.

    Evaluation of oxidative processes in human pigment epithelial cells associated with retinal outer segment phagocytosis

    Exp. Cell Res.

    (1994)
  • M. Pearl-Yafe et al.

    An oxidative mechanism of interferon induced priming of the Fas pathway in Fanconi anemia cells

    Biochem. Pharmacol.

    (2003)
  • M. Pearl-Yafe et al.

    The p38 pathway partially mediates caspase-3 activation induced by reactive oxygen species in Fanconi anemia C cells

    Biochem. Pharmacol.

    (2004)
  • P.L. Penfold et al.

    Immunological and aetiological aspects of macular degeneration

    Prog. Retin. Eye Res.

    (2001)
  • V.M. Reddy et al.

    Distribution of growth factors in subfoveal neovascular membranes in age-related macular degeneration and presumed ocular histoplasmosis syndrome

    Am. J. Ophthalmol.

    (1995)
  • P.S. Samiec et al.

    Glutathione in human plasma: decline in association with aging, age-related macular degeneration, and diabetes

    Free Radic. Biol. Med.

    (1998)
  • K. Schulze-Osthoff et al.

    Cytotoxic activity of tumor necrosis factor is mediated by early damage of mitochondrial functions. Evidence for the involvement of mitochondrial radical generation

    J. Biol. Chem.

    (1992)
  • G. Shanker et al.

    Free radical formation in cerebral cortical astrocytes in culture induced by methylmercury

    Brain Res. Mol. Brain Res.

    (2004)
  • J. Wassell et al.

    The photoreactivity of the retinal age pigment lipofuscin

    J. Biol. Chem.

    (1999)
  • S.I. Yamagishi et al.

    Leptin induces mitochondrial superoxide production and monocyte chemoattractant protein-1 expression in aortic endothelial cells by increasing fatty acid oxidation via protein kinase A

    J. Biol. Chem.

    (2001)
  • A. Yoshida et al.

    Thrombin regulates chemokine induction during human retinal pigment epithelial cell/monocyte interaction

    Am. J. Pathol.

    (2001)
  • A. Yoshida et al.

    Activated monocytes induce human retinal pigment epithelial cell apoptosis through caspase-3 activation

    Lab. Invest.

    (2003)
  • R.W. Young

    Pathophysiology of age-related macular degeneration

    Surv. Ophthalmol.

    (1987)
  • V.A. Alder et al.

    The effect of the retinal circulation on vitreal oxygen tension

    Curr. Eye Res.

    (1985)
  • M. Baggiolini et al.

    Interleukin-8 and related chemotactic Cytokines-CXC and CC chemokines

    Adv. Immunol.

    (1994)
  • Z.M. Bian et al.

    Differential involvement of phosphoinositide 3-kinase/Akt in human RPE MCP-1 and IL-8 expression

    Invest. Ophthalmol. Vis. Sci.

    (2004)
  • J. Cai et al.

    Oxidant-induced apoptosis in cultured human retinal pigment epithelial cells

    Invest. Ophthalmol. Vis. Sci.

    (1999)
  • S. Corda et al.

    Rapid reactive oxygen species production by mitochondria in endothelial cells exposed to tumor necrosis factor-alpha is mediated by ceramide

    Am. J. Respir. Cell Mol. Biol.

    (2001)
  • S.S. Deshpande et al.

    Rac1 inhibits TNF- alpha-induced endothelial cell apoptosis: dual regulation by reactive oxygen species

    FASEB J

    (2000)
  • C.K. Dorey et al.

    Superoxide production by porcine retinal pigment epithelium in vitro

    Invest. Ophthalmol. Vis. Sci.

    (1989)
  • C.K. Dorey et al.

    Growth of cultured RPE and endothelial cells is inhibited by blue light but not green or red light

    Curr. Eye Res.

    (1990)
  • V.M. Elner et al.

    Neutrophil chemotactic factor (IL-8) gene expression by cytokine-treated retinal pigment epithelial cells

    Am. J. Pathol.

    (1990)
  • S.G. Elner et al.

    Monocyte chemotactic protein gene expression by cytokine-treated human retinal pigment epithelial cells

    Lab. Invest.

    (1991)
  • Cited by (326)

    • Application of amniotic membrane in osteoarthritis management

      2024, Journal of Cartilage and Joint Preservation
    View all citing articles on Scopus
    View full text