Amyloid-beta mediates the receptor of advanced glycation end product-induced pro-inflammatory response via toll-like receptor 4 signaling pathway in retinal ganglion cell line RGC-5

https://doi.org/10.1016/j.biocel.2015.03.002Get rights and content

Abstract

Patients with diabetes mellitus have an increased risk of developing Alzheimer's disease. Amyloid-β, a product of amyloid precursor protein, is associated with neuro-inflammation in patients with Alzheimer's diseases. The correlation between amyloid-beta and advanced glycation end products, which accumulate in tissue of diabetic patients, is not clear. The aims of this study were to determine the effect of advanced glycation end product on the expression of amyloid precursor protein/amyloid-beta and associated pro-inflammatory responses in retinal ganglion cell line RGC-5. Treatment with advanced glycation end product produced upregulation of amyloid precursor protein and increased secretion of amyloid-β(1–40). Additionally, amyloid-β(1–40) induced toll-like receptor 4-dependent phosphorylation of tyrosine in myeloid differentiation primary response gene (88). We found that N-[N-(3,5-Difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester, a γ-secretase inhibitor, reduced the secretion of amyloid-β(1–40) and inhibited the advanced glycation end product-induced activation of myeloid differentiation primary response gene (88). Amyloid-β(1–40) induced the activation of NF-κB and the expression of TNFα mRNA. Knockdown of toll-like receptor 4 inhibited the amyloid-β(1–40)-induced phosphorylation of p65 in NF-κB. Additionally, the nuclear translocation of p65 and transcriptions of TNFα were inhibited by siRNA knockdown of receptor of advanced glycation end product or toll-like receptor 4. The advanced glycation end product-induced secretion of VEGF-A was also reduced by knockdown of toll-like receptor 4. Taken together, our data suggested that amyloid-β(1–40) mediates the interaction between receptor of advanced glycation end product and toll-like receptor 4. Inhibition of the toll-like receptor 4 is an effective method for suppressing the amyloid-β(1–40)-induced pro-inflammatory responses in RGC-5 cells.

Introduction

The presence of amyloid-beta (amyloid-β or Aβ) plaques formed by Aβ aggregation and neurofibrillary tangles containing the microtubule-stabilizing protein tau is a characteristic feature of Alzheimer's disease (AD) (Huang and Jiang, 2009, Sisodia and Price, 1995). The correlation between AD and diabetes mellitus (DM) has gained increasing attention. Epidemiological studies show that diabetic patients have an increased risk of developing AD (Maher and Schubert, 2009, Ott et al., 1996, Ott et al., 1999). The two are likely linked because impaired insulin signaling in Type 2 DM disrupts both the processing of amyloid precursor protein (APP) and the clearing of Aβ (Messier and Teutenberg, 2005).

The receptor for advanced glycation end products (RAGE), which mediates oxidative stress and vascular inflammation in DM (Nogueira-Machado and Chaves, 2008, Schmidt et al., 1999), is a putative receptor for Aβ peptide and plays a role in AD (Arancio et al., 2004, Deane et al., 2003, Donahue et al., 2006, Yan et al., 1996). RAGE is also involved in the translocation of Aβ from the extracellular to intracellular space and is correlated with neuronal cytotoxicity in the central nervous system (Takuma et al., 2009). Therefore, cerebrovascular alteration, which is a common pathological change in both AD and DM, could be the mechanism linking the two diseases (Takeda et al., 2011).

RAGE and toll-like receptors (TLRs) share common ligands and show crosstalk in signaling pathways that mediate the host immune response (Ibrahim et al., 2013). The TLR family is the best characterized of the pattern recognition receptor families, which detect conserved molecular patterns invariant among entire classes of pathogens and thereby trigger an innate immune response (Kawai and Akira, 2010). TLRs are activated in patients with DM (Dasu et al., 2010). Nutrients or other ligands can trigger TLRs to activate the inflammasome in cells, which releases cytokines, or to induce insulin resistance (Reynolds et al., 2012). Evidence from in vitro and in vivo studies suggests that TLR signaling pathway, including TLR2, 4 or 9 may be involved in clearance of Aβ-deposits in the brain and, therefore, that the TLR family could be a therapeutic target for AD (Tahara et al., 2006). Evidence for TLR4 signaling has been demonstrated in the mouse model of AD, in which it was associated with an increase expression in inflammatory cytokines (Jin et al., 2008).

In the eye, Aβ deposition disrupts retinal structure and may contribute to the visual deficits and retinal inflammation observed in the mouse model of AD (Ning et al., 2008, Perez et al., 2009). Aβ is also a known component of drusen, and it plays a proinflammatory role in the retinal pigment epithelium and neuroretina (Liu et al., 2013). However, the role played by APP and Aβ in retinal cells during DM is currently unclear. Therefore, we conducted an in vitro study using RGC-5 cells, which are identical to the retinal photoreceptor cell line 661W (Krishnamoorthy et al., 2013), to represent a neuronal cell in the retina. We used RGC-5 cells to investigate the effect of advanced glycation end products (AGEs), components that are associated with hyperglycemia, hyperlipidemia, enhanced oxidative stress and obesity (Gaens et al., 2013) on APP expression. Additionally, we studied the role of RAGE and TLR signaling in mediating Aβ-induced innate immune responses.

Section snippets

RGC-5 cell line culture and AGE preparation

We used the RGC-5 cell line (ATCC, Manassas, VA) to study the association between AGE and the upregulation of APP. RGC-5 cells were cultured in Dulbecco's modified Eagle's medium (DMEM), containing low glucose, 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin, at 37 °C and in a humidified atmosphere with 5% CO2. Unless otherwise stated, 2.5 × 106 cells were plated in 60-mm dishes (430166; Corning, Tewksbury, MA). After sub-confluent cell growth, the medium was replaced by DMEM with 0.5%

AGE upregulates APP expression in RGC-5 cells

Compared to RGC-5 cells treated with 200 μg/mL BSA, cells treated with 200 μg/mL AGE-BSA exhibited higher levels of APP mRNA between 4 h and 24 h relative to PBS-treated control, and this difference was significant at 8 and 12 h (Fig. 1A). Western blotting of whole-cell lysates from RGC-5 cells showed that BSA 200 μg/mL BSA had no effect on APP protein expression, while, 200 μg/mL AGE-BSA significantly increased levels of APP at 8 and 12 h (Fig. 1B). During a 24 h treatment, AGE-BSA increased levels of

Discussion

In this study, we demonstrated that AGE induces the upregulation of APP and the secretion of Aβ1–40 in the RGC-5 cell line. Additionally, we showed that the TLR4–MyD88 signaling pathway plays an important role in the NF-κB-dependent expression of TNFα and the secretion of VEGF-A (see summary in Fig. 8).

It is possible that the effect of AGE on cells could be amplified by autocrine signaling via the upregulation of molecules that also interact with RAGE, such as Aβ. In our study, upregulation of

Conclusions

Here, we found that AGE induced the upregulation of APP and the release of Aβ1–40, which was associated with the activation of the TLR4/MyD88 signaling pathway. We suggested that TLR4–MyD88 interacts with RAGE to regulate the activation of NF-κB and the downstream expression of genes such as TNFα. Aβ1–40 could also mediate the AGE-induced upregulation of VEGF-A via the TLR4 signaling pathway. Our results provide new insights into the potential role that Aβ and/or TLR4 inhibition could play in

Acknowledgments

This research was supported by National Science Council Taiwan grant NSC-98-2314-B-182-007-MY3 and Chang Gung Medical Research Program grant CMRPG-8B1471. The ELISA reader were kindly provided by Dr. Li-Tung Huang of Free Radical and Children Medicine Research Laboratory, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine.

References (48)

  • M.R. Dasu et al.

    Increased toll-like receptor (TLR) activation and TLR ligands in recently diagnosed type 2 diabetic subjects

    Diabetes Care

    (2010)
  • R. Deane et al.

    RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain

    Nat Med

    (2003)
  • R. Deane et al.

    A multimodal RAGE-specific inhibitor reduces amyloid beta-mediated brain disorder in a mouse model of Alzheimer disease

    J Clin Invest

    (2012)
  • V. Dinet et al.

    Distinct effects of inflammation on gliosis, osmohomeostasis, and vascular integrity during amyloid beta-induced retinal degeneration

    Aging Cell

    (2012)
  • J.E. Donahue et al.

    RAGE, LRP-1, and amyloid-beta protein in Alzheimer's disease

    Acta Neuropathol

    (2006)
  • K.H. Gaens et al.

    Advanced glycation endproducts and its receptor for advanced glycation endproducts in obesity

    Curr Opin Lipidol

    (2013)
  • H.C. Huang et al.

    Accumulated amyloid-beta peptide and hyperphosphorylated tau protein: relationship and links in Alzheimer's disease

    J Alzheimers Dis

    (2009)
  • J.J. Jin et al.

    Toll-like receptor 4-dependent upregulation of cytokines in a transgenic mouse model of Alzheimer's disease

    J Neuroinflammation

    (2008)
  • T. Kawai et al.

    The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors

    Nat Immunol

    (2010)
  • R.R. Krishnamoorthy et al.

    A forensic path to RGC-5 cell line identification: lessons learned

    Invest Ophthalmol Vis Sci

    (2013)
  • J.J. Lee et al.

    High-mobility group box 1 protein is implicated in advanced glycation end products-induced vascular endothelial growth factor A production in the rat retinal ganglion cell line RGC-5

    Mol Vis

    (2012)
  • J.H. Li et al.

    Advanced glycation end products activate Smad signaling via TGF-beta-dependent and independent mechanisms: implications for diabetic renal and vascular disease

    FASEB J

    (2004)
  • R.T. Liu et al.

    Inflammatory mediators induced by amyloid-beta in the retina and RPE in vivo: implications for inflammasome activation in age-related macular degeneration

    Invest Ophthalmol Vis Sci

    (2013)
  • P.A. Maher et al.

    Metabolic links between diabetes and Alzheimer's disease

    Expert Rev Neurother

    (2009)
  • Cited by (18)

    • Intracellular amyloid-β disrupts tight junctions of the retinal pigment epithelium via NF-κB activation

      2020, Neurobiology of Aging
      Citation Excerpt :

      NF-κB inactivation through IKK inhibition prevented the downregulation of ZO-1 and occludin in RPE cells. One of the mechanisms underlying NF-κB–mediated tight-junction disruption is the upregulation of tumor necrosis factor-α (Lee et al., 2015; Sun et al., 2017), which modulates the property of blood-neural barriers (Kniesel and Wolburg, 2000). Another possible and more direct mechanism is the suppression of gene expression of tight-junction proteins by NF-κB (Aveleira et al., 2010).

    • Roles of Toll-Like Receptor 4 for Cellular Pathogenesis in Primary Open-Angle Glaucoma: A potential therapeutic strategy

      2019, Journal of Microbiology, Immunology and Infection
      Citation Excerpt :

      Moreover, the activation inhibitor NF-κB, which is down-stream signaling TLR4 activation via MyD88 dependent pathway, is able to protect ganglion cell layer during HMGB1 treatments.52 In addition, knockdown TLR4 by using siRNA suppress amyloid-β induced pro-inflammatory response via NF-κB activation in RGC.53 Whereas, some evidences showed the Octreotide protects retinal ischemic by activation of NF- κB.54

    • Toll-like receptors 4, 5, 6 and 7 are constitutively expressed in non-human primate retinal neurons

      2018, Journal of Neuroimmunology
      Citation Excerpt :

      The main function of ganglion cells is to receive synaptic inputs from bipolar and amacrine cells and transfer the signals to the lateral geniculate nucleus (de Souza et al., 2016). Previous studies have discovered a link between TLR4 activation, initiation of inflammatory pathways, and subsequent RGC degeneration (Kilic et al., 2008; Lee et al., 2015; Morzaev et al., 2015; Qi et al., 2014). In addition, an increase in TLR4 expression, along with a decrease in RGC viability, was noted after exposure to a high glucose environment (Zhao et al., 2016).

    • Receptor for advanced glycation end-products: Biological significance and imaging applications

      2024, Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology
    View all citing articles on Scopus
    View full text