Megalin interacts with APP and the intracellular adapter protein FE65 in neurons

https://doi.org/10.1016/j.mcn.2010.07.005Get rights and content

Abstract

Increasing evidence has implicated megalin, a low-density lipoprotein receptor-related protein, in the pathogenesis of Alzheimer's disease (AD). In the brain, megalin is expressed in brain capillaries, ependymal cells and choroid plexus, where it participates in the clearance of brain amyloid β-peptide (Aβ) complex. Recently, megalin has also been detected in oligodendrocytes and astrocytes. In this study we demonstrate that megalin is widely distributed in neurons throughout the brain. Additionally, given that FE65 mediates the interaction between the low density lipoprotein receptor-related protein-1 and the amyloid precursor protein (APP) to modulate the rate of APP internalization from the cell surface, we hypothesize that megalin could also interact with APP in neurons. Our results confirm that megalin interacts with APP and FE65, suggesting that these three proteins form a tripartite complex. Moreover, our findings imply that megalin may participate in neurite branching. Taken together, these results indicate that megalin has an important role in Aβ-mediated neurotoxicity, and therefore may be involved in the neurodegenerative processes that occur in AD.

Introduction

Megalin, also known as low density lipoprotein-related protein-2 (LRP2) and glycoprotein 330, is the largest member of the low-density lipoprotein receptor (LDLR) family. Megalin expression was first detected in various absorptive epithelial cells throughout the body, such as the kidney and the intestinal brush border. In the central nervous system, megalin was initially described in brain capillaries, ependymal lining of the ventricular walls and choroid plexus (Chun et al., 1999, Kounnas et al., 1994, Zheng et al., 1994), and later in neural progenitors in embryonic mouse spinal cord (Wicher et al., 2005), and postnatal mouse spinal cord oligodendrocytes (Wicher et al., 2006). More recently, megalin has been described in cultured astrocytes (Bento-Abreu et al., 2008) and neurons (Ambjørn et al., 2008, Chung et al., 2008, Fleming et al., 2009).

Megalin binds and internalizes a number of ligands that represent a wide variety of molecules, including lipoproteins, hormones, vitamin-binding proteins, drugs, and immunorelated proteins (Hjälm et al., 1996, Moestrup & Verroust, 2001). Since many are involved in neuroprotection, they are considered potential new targets in AD research (Birn, 2006, Carro et al., 2005, Christensen & Birn, 2002, Dietrich et al., 2008, Moestrup & Verroust, 2001).

In the brain, megalin participates in endocytosis and transport of amyloid β-peptide (Aβ) complex across the blood–cerebrospinal fluid barrier at the choroid plexus (Carro et al., 2005, Zlokovic et al., 1996), and the blood-brain barrier (Deane et al., 2004, Hammad et al., 1997, Zlokovic et al., 1996). This process involves the interaction of megalin's cytoplasmic tail domain with several adaptor and scaffold proteins (Biemesderfer, 2006, May et al., 2003), including Dab2, MAGI-I, GIPC, ANKRA, MegBP, and ARH (Bonifacino & Traub, 2003, Jaeger & Pietrzik, 2008). The adaptor protein FE65 has been shown to mediate the interaction between another member of the LDLR superfamily, the LRP1, and the amyloid precursor protein (APP) (Cam & Bu, 2006, Pietrzik et al., 2004, Yoon et al., 2005).

Due to the growing evidence that megalin expression in the central nervous system is not restricted to tight-junction epithelia, but is also expressed in neurons, the objective of our study was to assess the expression of megalin in different subpopulations of neurons, explore the interaction between megalin, APP and FE65, and discuss its possible functional role in the central nervous system.

Section snippets

Megalin expression in neurons

WB of neuronal cells and choroid plexus proteins showed a megalin signal consistent with its reported size in primary hippocampal and cortical neuronal cell cultures (Fig. 1a). Single immunofluorescence using three different megalin antisera showed that in cultured neurons, megalin immunoreactivity appears as a punctate labeling throughout the neuronal cytoplasm, but mainly within cell bodies and proximal neurites (Fig. 1b). Double immunofluorescence experiments confirmed megalin expression in

Discussion

Three major observations can be drawn from our study: (1) that megalin is expressed in adult neurons, (2) that megalin interacts with FE65 and APP, suggesting the formation of a tripartite complex, and that (3) megalin may be involved in neurite branching.

Megalin is a LDLR involved in endocytosis and transport of Aβ complex across the blood barrier at the choroid plexus (Carro et al., 2005, Zlokovic et al., 1996) and brain endothelial cells (Deane et al., 2004, Hammad et al., 1997, Zlokovic et

Conclusions

In conclusion, the results described herein demonstrate that megalin is expressed in adult neurons, that it interacts with APP and FE65, and that neurite branching may be associated with a reduced expression of megalin. Our data argue in favor of an important role of neuronal megalin in Aβ-mediated neurotoxicity. We propose that one of the mechanisms that participate in the neurodegenerative processes observed in AD involves a dysfunction in neuronal megalin.

Primary antibodies

Mouse monoclonal anti-β-III tubulin (1:1000; Millipore); mouse monoclonal anti- Glial fibrillary acidic protein (GFAP; 1:1000; Sigma-Aldrich); goat polyclonal anti-megalin (1:500; Santa Cruz Biotechnology, Inc.); rabbit polyclonal anti-megalin (1:500; kind gift from Dr. Michele Marino, Department of Endocrinology, University of Pisa, Italy); mouse polyclonal anti-megalin (1:500; Abcam); rabbit polyclonal anti-APP (1:1000; Sigma-Aldrich); goat polyclonal anti-FE65 (1:250; Santa Cruz

Disclosure statement

There are no actual or potential conflicts of interest with other people or organizations.

Funding was provided by grants from ‘Fondo de Investigación Sanitaria (FIS)’ (CP04/00179, PI060155), ‘Fundación Investigación Médica Mutua Madrileña’ (2006.125), ‘CIBERNED’, and ‘Ministerio de Educación y Ciencia’ (BFU2006-07430/BFI).

Acknowledgments

This work was supported by grants from Fondo de Investigación Sanitaria (FIS) (CP04/00179, PI060155), Fundación Investigación Médica Mutua Madrileña (2006.125; 2008.93), CIBERNED, and Ministerio de Educación y Ciencia (BFU2006-07430/BFI). We thank Begoña Santiago for her expert help in FRET measurements. We also thank Dr. Carlos Avendaño for his helpful suggestions and Ms. Marta Callejo Móstoles for her valuable help.

References (52)

  • Q. Liu et al.

    Amyloid precursor protein regulates brain apolipoprotein E and cholesterol metabolism through lipoprotein receptor LRP1

    Neuron

    (2007)
  • S. Pons et al.

    Insulin-like growth factor-I stimulates dephosphorylation of ikappa B through the serine phosphatase calcineurin (protein phosphatase 2B)

    J. Biol. Chem.

    (2000)
  • S.L. Sabo et al.

    Regulation of beta-amyloid secretion by FE65, an amyloid protein precursor-binding protein

    J. Biol. Chem.

    (1999)
  • R.E. Tanzi et al.

    Twenty years of the Alzheimer's disease amyloid hypothesis: a genetic perspective

    Cell

    (2005)
  • E. Waldron et al.

    LRP1 modulates APP trafficking along early compartments of the secretory pathway

    Neurobiol. Dis.

    (2008)
  • D.T. Walsh et al.

    Amyloid-beta peptide is toxic to neurons in vivo via indirect mechanisms

    Neurobiol. Dis.

    (2002)
  • I.S. Yoon et al.

    Sequences from the low density lipoprotein receptor-related protein (LRP) cytoplasmic domain enhance amyloid beta protein production via the beta-secretase pathway without altering amyloid precursor protein/LRP nuclear signaling

    J. Biol. Chem.

    (2005)
  • M. Ambjørn et al.

    Metallothionein and a peptide modeled after metallothionein, EmtinB, induce neuronal differentiation and survival through binding to receptors of the low-density lipoprotein receptor family

    J. Neurochem.

    (2008)
  • A. Bento-Abreu et al.

    Megalin is a receptor for albumin in astrocytes and is required for the synthesis of the neurotrophic factor oleic acid

    J. Neurochem.

    (2008)
  • H. Birn

    The kidney in vitamin B12 and folate homeostasis: characterization of receptors for tubular uptake of vitamins and carrier proteins

    Am. J. Physiol. Renal Physiol.

    (2006)
  • J.S. Bonifacino et al.

    Signals for sorting of transmembrane proteins to endosomes and lysosomes

    Annu. Rev. Biochem.

    (2003)
  • J.A. Cam et al.

    Modulation of beta-amyloid precursor protein trafficking and processing by the low density lipoprotein receptor family

    Mol. Neurodegener.

    (2006)
  • X. Cao et al.

    A transcriptionally [correction of transcriptively] active complex of APP with Fe65 and histone acetyltransferase Tip60

    Science

    (2001)
  • E. Carro et al.

    Circulating insulin-like growth factor I mediates effects of exercise on the brain

    J. Neurosci.

    (2000)
  • E. Carro et al.

    Serum insulin-like growth factor I regulates brain amyloid-beta levels

    Nat. Med.

    (2002)
  • E. Carro et al.

    Choroid plexus megalin is involved in neuroprotection by serum insulin-like growth factor I

    J. Neurosci.

    (2005)
  • Cited by (55)

    • Evaluation of chronic lead effects in the blood brain barrier system by DCE-CT

      2020, Journal of Trace Elements in Medicine and Biology
      Citation Excerpt :

      In AD, disrupted BBB may cause the dysfunction of Aβ transport from brain to the peripheral circulation [13]. Additionally, choroid plexus may also be involved in AD etiology [14–16]. Studies have found that choroid plexus participates in brain amyloidosis and Aβ transport [14,15,17,18].

    • Expression Profiles of Metallothionein I/II and Megalin in Cuprizone Model of De- and Remyelination

      2018, Neuroscience
      Citation Excerpt :

      This would be also a plausible explanation for the selective transport of some substrates across the BBB and blood–cerebrospinal fluid barrier in cuprizone intoxication, since generally it does not induce the breakdown of the blood–brain barrier (BBB) (Hiremath et al., 2008; Skripuletz et al., 2011; Praet et al., 2014). The finding might be of particular interest, because the endogenous processes and therapies that increase megalin expression at the choroid plexus may control also the accumulation of brain Aβ and enhance the repair in the damaged brain, since LDL-R family are involved in trafficking and processing of the amyloid precursor protein (APP) and the uptake of apolipoprotein J/clusterin, a binding protein for the Aβ peptide (Zlokovic et al., 1996; Alvira-Botero and Carro, 2010; Alvira-Botero et al., 2010). In this context our data support the current knowledge pointing to the therapeutic potential of MT-I/II and peptides modeled from MT in AD and MS, as well as in copper and zinc-dyshomeostasis, mitochondrial dysfunctions, aging and other neurodegenerative disorders (Coyle et al., 2002; Mocchegiani et al., 2007; Malavolta et al., 2008; Chung et al., 2008a, 2010; Pedersen et al., 2009; Manso et al., 2011; West et al., 2011; Sharma and Ebadi, 2013; Szewczyk, 2013).

    • Linking deregulation of non-coding RNA to the core pathophysiology of Alzheimer's disease: An integrative review

      2017, Progress in Neurobiology
      Citation Excerpt :

      It should also be pointed out that the integrity of the BBB itself is disrupted by Aβ42, in part by its interference with endothelial tight junction complexes containing crucial proteins like Claudin-1/5 and Occludin (Reinhold and Rittner, 2017; Toyama et al., 2016). The low-density lipoprotein related receptors (LRP) 1 (and 2) − the latter also known as Megalin − are major mechanisms for ejecting Aβ42 from the brain, in addition to their roles in Aβ42 internalisation and autophagic-lysosomal degradation by microglial cells (Section 6.9) (Alvira-Botero et al., 2010; Auderset et al., 2016; Ramanathan et al., 2015). LRP 1 and 2 possess single transmembrane domains and, intriguingly, are cleaved at the cell surface by several APP-processing enzymes.

    View all citing articles on Scopus
    1

    These authors have contributed equally to this work.

    View full text