Structural Studies of the Alzheimer’s Amyloid Precursor Protein Copper-binding Domain Reveal How it Binds Copper Ions

https://doi.org/10.1016/j.jmb.2006.12.041Get rights and content

Abstract

Alzheimer's disease (AD) is the major cause of dementia. Amyloid β peptide (Aβ), generated by proteolytic cleavage of the amyloid precursor protein (APP), is central to AD pathogenesis. APP can function as a metalloprotein and modulate copper (Cu) transport, presumably via its extracellular Cu-binding domain (CuBD). Cu binding to the CuBD reduces Aβ levels, suggesting that a Cu mimetic may have therapeutic potential. We describe here the atomic structures of apo CuBD from three crystal forms and found they have identical Cu-binding sites despite the different crystal lattices. The structure of Cu2+-bound CuBD reveals that the metal ligands are His147, His151, Tyr168 and two water molecules, which are arranged in a square pyramidal geometry. The site resembles a Type 2 non-blue Cu center and is supported by electron paramagnetic resonance and extended X-ray absorption fine structure studies. A previous study suggested that Met170 might be a ligand but we suggest that this residue plays a critical role as an electron donor in CuBDs ability to reduce Cu ions. The structure of Cu+-bound CuBD is almost identical to the Cu2+-bound structure except for the loss of one of the water ligands. The geometry of the site is unfavorable for Cu+, thus providing a mechanism by which CuBD could readily transfer Cu ions to other proteins.

Introduction

Alzheimer's disease (AD) is the leading cause of dementia among the elderly and is characterized by the presence of amyloid plaques, extensive neuronal death and shrinkage of the brain. It is increasingly accepted that the neurotoxic Aβ peptide is responsible for compromising neuronal functions and triggering cell death.1 The peptide is derived from the cleavage of the amyloid precursor protein (APP)2 and is the main constituent of the amyloid plaques. APP belongs to a wider family of APP-like proteins (APLPs) that include APLP1 and APLP2.3 These proteins exhibit functional redundancy to some degree and can undergo cleavage, but only APP cleavage gives rise to the Aβ peptide. The cleavage occurs during trafficking and maturation through the protein secretory pathway by membrane-bound proteinases. Aβ arises through the sequential cleavage by the β-site APP cleaving enzyme (BACE)4., 5. at a site just outside the plasma membrane and the γ-secretase protein complex6 within the membrane (Figure 1).

APP is a Type-I transmembrane protein with a large extracellular portion, which can be structurally and functionally subdivided into several domains (Figure 1). At the N terminus is a cysteine-rich region consisting of the growth factor domain (GFD), which binds heparin and can stimulate neurite outgrowth,7., [8] and a copper-binding domain (CuBD) that binds Cu and zinc.9 The cysteine-rich region is followed by an acidic-rich region of sequence, a KPI (Kunitz-type protease inhibitor) domain and an OX2 domain.10., 11. The KPI and OX2 domains may be spliced out, to give rise to three (main) variants APP770 (with 770 amino acid residues), APP751 and APP695. APP695 is the most common isoform in the brain.12 Following these domains is a glycosylated domain referred to as E2 in the published structure by Wang and Ha.13 Next is a region predicted to adopt no regular secondary structure (unpublished results) and the transmembrane region. The C-terminal cytoplasmic tail may be involved in various cellular functions, such as transcription signaling,14 through interaction with a multitude of proteins.15 However, an endogenous ligand that triggers signaling through APP has yet to be identified. There is evidence that APP exists in a monomer to dimer equilibrium in the membrane.[16], 17. The binding of high molecular weight heparin also appears able to induce dimerization.18 Dimerization may cause an initiation of signaling events19 and play a role in modulating APP cleavage as the production of Aβ, in particular dimeric Aβ, is increased when APP is forcibly dimerized.16 What influences the monomer-dimer equilibrium is not clear, but certain pathological states such as heme deficiency20 or even Aβ itself21 may lead to greater APP oligomerization.

APP is a metalloprotein that contributes to Cu ion metabolism. APP binds to free Cu2+ ions and once bound, Cu2+ is reduced to Cu+ in vitro.22 This can lead to increased Cu-mediated neurotoxicity in cultured neurons,23 presumably through increased oxidative stress or the generation of reactive oxygen species.24 A role for APP in Cu efflux is supported by a series of experiments that demonstrate an increased intracellular accumulation of Cu in the brains of APP/APLP2 double knockout mice25 and neuronal cultures26 whilst transgenic over-expression of APP has the opposite effects in mice and cultured neurons.[26], 27. The involvement of CuBD in Cu efflux has been demonstrated in yeast expressing an APP ectodomain fragment. Mutating the CuBD significantly increases intracellular Cu levels compared to yeast expressing a wild-type fragment.28

The interaction between Cu ions and APP, via CuBD, can modulate Aβ production and the progression of AD. The treatment of CHO cells over-expressing APP with extracellular Cu2+ leads to reduced Aβ production and a shift of the cleavage equilibrium away from the BACE/γ-secretase pathway.29 The effects are abolished when the Cu binding residues within CuBD are mutated.30 The beneficial outcomes of Cu2+ binding to APP have been illustrated in two independent transgenic mouse studies. By supplementing drinking water with Cu2+ ions, the survival of APP23 mice (which overproduce Aβ) was improved along with increased bioavailable Cu2+ and reduced Aβ levels in the brain.31 Similar effects were achieved by crossing TgCRND8 mice (that overproduce Aβ) with mice that have raised brain Cu levels due to the expression of a mutant Cu transporter CuATPase7b. These mice had decreased amyloid plaque and plasma Aβ levels.32

To define the molecular interaction between the APP CuBD and Cu, we have determined the crystal structures of CuBD in apo and Cu-bound forms at near atomic resolution. We have also performed electron paramagnetic resonance (EPR) and extended X-ray absorption fine structure (EXAFS) experiments to probe the Cu binding site in solution. The structural studies reveal how Cu binds to APP and provide the basis for the design of novel therapeutics to combat AD.

Section snippets

Crystal structures of Apo CuBD

Although we had previously determined a structure of CuBD in apo form by NMR,9 we wished to obtain a higher resolution structure of the Cu binding site in the apo protein that we could compare to Cu-bound crystal structures of CuBD. We have obtained three different crystal forms, named Type A, B and C, consisting of 11 different views of the molecule, all diffracting to high resolution. Hence any crystal lattice effects on the Cu binding site could readily be discerned. All the Cu binding

Discussion

We have determined the structure of apo CuBD in three different crystal forms at high resolution with 11 different views of the molecule. The structure consists of an α-helix packed against a three-strand β-sheet. According to the classification by the CATH server,38 CuBD belongs to the class of mixed α/β proteins and displays an architecture of a two-layered sandwich. The topology of CuBD is classified as either defensin A-like proteins or α-β plaits. Proteins that exhibit the latter topology

Apo CuBD133-189 (Type A) structure

This construct consisted of residues 133 to 189 plus a glutamine and an alanine at the N terminus as a result of a cloning artifact. The expression, purification and crystallization of this construct have been described in detail elsewhere.55 In brief, the protein was expressed in Pichia pastoris, purified by gel filtration chromatography and concentrated to ∼10 mg ml−1. The protein was crystallized by the hanging drop method at room temperature in 0.1 M Hepes (pH 8.0), 28%–32% (w/v) PEG 10 000.

Acknowledgements

We thank Galina Polekhina for general advice on crystallography, and Harry Tong and other BioCARS staff for their help with data collection at APS. Use of APS and SSRL was supported by the U.S. Department of Energy, Basic Energy Sciences, Office of Energy Research. The SSRL Structural Molecular Biology Program is supported by the Department of Energy, Office of Biological and Environmental Research, and by the National Institutes of Health, National Center for Research Resources, Biomedical

References (64)

  • A.R. White et al.

    Copper levels are increased in the cerebral cortex and liver of APP and APLP2 knockout mice

    Brain Res.

    (1999)
  • C.J. Maynard et al.

    Overexpression of Alzheimer's disease amyloid-β opposes the age-dependent elevations of brain copper and iron

    J. Biol. Chem.

    (2002)
  • C. Treiber et al.

    Clioquinol mediates copper uptake and counteracts Cu efflux activities of the amyloid precursor protein of Alzheimer's disease

    J. Biol. Chem.

    (2004)
  • J. Peisach et al.

    Structural implications derived from the analysis of electron paramagnetic resonance spectra of natural and artificial copper proteins

    Arch. Biochem. Biophys.

    (1974)
  • M.E.P. Murphy et al.

    Structure of nitrite bound to copper-containing nitrite reductase from Alcaligenes faecalis

    J. Biol. Chem.

    (1997)
  • C.C. Curtain et al.

    Alzheimer's disease amyloid-beta binds copper and zinc to generate an allosterically ordered membrane-penetrating structure containing superoxide dismutase-like subunits

    J. Biol. Chem.

    (2001)
  • B. Angeletti et al.

    BACE1 cytoplasmic domain interacts with the copper chaperone for superoxide dismutase-1 and binds copper

    J. Biol. Chem.

    (2005)
  • Z. Otwinowski et al.

    Processing of X-ray diffraction data collected in oscillation mode

    In Methods Enzymol.

    (1997)
  • G.R. Hanson et al.

    XSophe-Sophe-XeprView. A computer simulation software suite (v.1.1.3) for the analysis of continuous wave EPR spectra

    J. Inorg. Biochem.

    (2004)
  • D.J. Selkoe

    Alzheimer's disease is a synaptic failure

    Science

    (2002)
  • J. Kang et al.

    The presursor of Alzheimer's disease amyloid A4 protein resembles a cell-surface receptor

    Nature

    (1987)
  • S. Sinha et al.

    Purification and cloning of amyloid precursor protein β-secretase from human brain

    Nature

    (1999)
  • R. Yan et al.

    Membrane-anchored aspartyl protease with Alzheimer's disease β-secretase activity

    Nature

    (1999)
  • J. Rossjohn et al.

    Crystal structure of the N-terminal, growth factor-like domain of Alzheimer amyloid precursor protein

    Nature Struct. Biol.

    (1999)
  • R.E. Tanzi et al.

    Protease inhibitor domain encoded by an amyloid protein precursor mRNA associated with Alzheimer's disease

    Nature

    (1988)
  • C. Haass et al.

    Processing of beta-amyloid precursor protein in microglia and astrocytes favors an internal localization over constitutive secretion

    J. Neurosci.

    (1991)
  • X. Cao et al.

    A transcriptively active complex of APP with Fe65 and histone acetyltransferase Tip60

    Science

    (2001)
  • B. De Strooper et al.

    Proteolytic processing and cell biological functions of the amyloid precursor protein

    J. Cell Sci.

    (2000)
  • P. Soba et al.

    Homo- and heterodimerization of APP family members promotes intercellular adhesion

    EMBO J.

    (2005)
  • C.M. Sondag et al.

    Amyloid precursor protein mediates proinflammatory activation of monocytic lineage cells

    J. Biol. Chem.

    (2004)
  • H. Atama et al.

    Heme deficiency may be a factor in the mitochondrial and neuronal decay of aging

    Proc. Natl Acad. Sci. USA

    (2002)
  • D.C. Lu et al.

    Amyloid β protein toxicity mediated by the formation of amyloid-β protein precursor complexes

    Annals Neurol.

    (2003)
  • Cited by (86)

    • The role of d-block metal ions in neurodegenerative diseases

      2023, Comprehensive Inorganic Chemistry III, Third Edition
    • Members of the vertebrate contactin and amyloid precursor protein families interact through a conserved interface

      2022, Journal of Biological Chemistry
      Citation Excerpt :

      The FN1–FN3 region of chicken CNTN4 is characterized by a sharp bend between the FN2 and FN3 domains that appears to be a hallmark of vertebrate CNTNs (34). The binding site involves the second FN repeat of CNTN4 and the copper-binding domain of APP, though it does not overlap with residues involved in copper binding (Figs. 4A and S2) (37, 38). The complex interface occludes only ∼600 Å2 of surface area, but with a high shape complementarity coefficient of 0.78, which is consistent with values calculated for trypsin–trypsin-inhibitor structures (39).

    • Copper imbalance in Alzheimer's disease: Convergence of the chemistry and the clinic

      2019, Coordination Chemistry Reviews
      Citation Excerpt :

      Examples of established copper sites in relevant proteins are shown in Fig. 2A–2F. Most notably are the two copper sites in the extracellular E2 domain of APP (Fig. 2A and 2B), one having a 4-histidine coordination sphere similar to that in SOD1 (Fig. 2E), and the other E2 copper binding in a trigonal coordination geometry to one histidine, a carboxylate group of a glutamate residue, and a solvent water, with a potential coordinating methionine near-by [139]. Another copper site in APP is the so-called copper binding domain (CuBD) [144]; this copper binds two histidines and a tyrosine and two solvent molecules in a distorted trigonal bipyramidal geometry (Fig. 2D) [141]. CCO, which is the terminal protein in the electron transport chain and thus required for energy production, contains both a CuA and CuB center.

    • Cytochrome b561, copper, β-cleaved amyloid precursor protein and niemann-pick C1 protein are involved in ascorbate-induced release and membrane penetration of heparan sulfate from endosomal S-nitrosylated glypican-1

      2017, Experimental Cell Research
      Citation Excerpt :

      In the copper-binding domain of sAPPβ, Cu(II) is spontaneously reduced to Cu(I) [16,36]. Apparently, Cu(I) ions can then be transferred to other proteins [36], such as Gpc-1-SNO. As cells approach growth quiescence, formation of HS-anMan ceases.

    View all citing articles on Scopus
    1

    Present address: G. K.-W. Kong, Abt. Molekulare Strukturbiologie, Max-Planck-Institut für Biochemie, Am Klopferspitz 18, D-82152 Martinsried, Germany; J. J. Adams, Australian Synchrotron Project, 800 Blackburn Road, Clayton, Victoria 3168, Australia; W. J. McKinstry, Pfizer Animal Health, Parkville, Victoria 3052, Australia.

    Co-senior authors.

    View full text