Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Defining the binding interface of Amyloid Precursor Protein (APP) and Contactin3 (CNTN3) by site-directed mutagenesis

  • Xi Peng,

    Roles Conceptualization, Investigation, Writing – original draft, Writing – review & editing

    Affiliation Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America

  • John Williams,

    Roles Investigation

    Affiliations Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America

  • Philip M. Smallwood,

    Roles Investigation

    Affiliations Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America

  • Jeremy Nathans

    Roles Conceptualization, Funding acquisition, Investigation, Methodology, Project administration, Supervision, Writing – original draft, Writing – review & editing

    jnathans@jhmi.edu

    Affiliations Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America

Abstract

The Amyloid Precursor Protein (APP) and Contactin (CNTN) families of cell-surface proteins have been intensively studied in the context of neural development and neuropsychiatric diseases. Earlier studies demonstrated both genetic and biochemical interactions between the extracellular domains of APP and CNTN3, but their precise binding interfaces were not defined. In the present study, we have used binding assays between APP-alkaline phosphatase (AP) fusion proteins and CNTN-Fc fusion proteins, together with alanine substitution mutagenesis, to show that: (i) the second Fibronectin domain (Fn(2)) in CNTN3 mediates APP binding; (ii) the copper binding domain (CuBD) in APP mediates CNTN3 binding; and (iii) the most important amino acids for APP-CNTN3 binding reside on one face of CNTN3-Fn(2) and on one face of APP-CuBD. These experiments define the regions of direct contact that mediate the binding interaction between APP and CNTN3.

Introduction

Amyloid Precursor Protein (APP) is an evolutionarily conserved vertebrate protein that is expressed in the developing and adult nervous system, as well as in many non-neural tissues [1]. APP has a large extracellular domain (ECD), a single trans-membrane region, and a small cytoplasmic tail. The ECD of the major APP isoform (APP695) consists of the following domains (starting from the N-terminus): (i) the E1 domain, which consists of a growth factor-like domain (GFLD) and a copper-binding domain (CuBD), (ii) the extension domain (ED); (iii) the acidic domain (AcD); (iv) a central domain (E2); (v) the juxtamembrane region (JMR); (vi) the transmembrane domain; and (vii) the intracellular domain (AICD) (Fig 1A) [2]. Over the past 25 years, APP has been the object of intense interest because proteolytic cleavage in the juxtamembrane and transmembrane domains generates the beta-amyloid peptide that accumulates in patients with Alzheimer disease (AD) [3].

thumbnail
Fig 1. CNTN3 sequences required for binding to APP.

(A) Schematic of APP and CNTN3 full-length proteins, showing their domain structures. Red lines indicate the lipid bilayer. Abbreviations for APP: GFLD, growth factor-like domain; CuBD, copper binding domain; ED, extension domain; AcD, acidic domain; E2, central domain; JMR, juxtamembrane region; AICD, APP intracellular domain [2]. Abbreviations for CNTN3: Ig, immunoglobulin domain; Fn, fibronectin domain. (B) Image of a Protein-G coated 96-well tray showing the AP reaction product (blue). The indicated Fc-fusion proteins (“target”) were immobilized by Protein-G binding and then incubated with the indicated APP-AP fusion proteins (the full ECD or the E1 domain) or with AP alone (“probe”). For each CNTN protein the full ECD (including all Ig and Fn domains) was fused to Fc. The concentrations of the different Fc-fusions were adjusted to provide equivalent amounts, as determined by immunoblotting (S1 Fig). The control wells in these and other AP assays were coated with an irrelevant Fc fusion protein. (C) Domain deletions in the ECD of CNTN3-Fc show that Fn(1–2) contains the principal sites of APP-E1-AP binding. The parental fusion protein (WT) has the full CNTN3 ECD fused to Fc. The first Ig domain is adjacent to the signal peptide. (D) Sequence of CNTN3 Fn(2) showing the locations of alanine substitution mutations targeting groups of mostly polar surface amino acids (“first set”; upper sequence) or single amino acids (lower sequence). AS, alanine substitution. (E) 96-well binding assay showing APP-E1-AP binding to the CNTN3 Fn(2) alanine substitution mutations shown in (D) constructed in the context of CNTN3-Fn(1–2)-Fc. (F) Immunoblots of serum-free conditioned medium containing the indicated WT and mutant CNTN-Fc proteins, visualized with anti-human IgG. Mass in kDa is listed for the protein size standards. These are the same for the right-most four blots. Quantification of AP binding is shown in S1A and S1B Fig and is summarized in S1 Table.

https://doi.org/10.1371/journal.pone.0219384.g001

The role(s) that APP plays in normal development and physiology are numerous and complex, and their molecular mechanisms are still largely unknown [1]. Gene knockout studies in mice have demonstrated substantially redundant roles for APP and its close homologues, APLP1 and APLP2 (APP-like proteins-1 and -2), in synaptic development, function, and plasticity [4,5], and in dendritic growth, branching, and spine maturation [69]. APP has been proposed to function as a cell-surface receptor [10], as a soluble ligand following proteolytic release of its ECD [11,12], and as the source of an intracellular signal via proteolytic release of its C-terminal tail [13,14].

Contactins (CNTNs) are glycosylphosphatidylinositol (GPI) anchored cell-surface proteins with a canonical structure of six amino-terminal Ig domains followed by four fibronectin III-like (Fn) domains (Fig 1A) [15]. There are six CNTN genes in mammals and, for some members of the CNTN family, additional sequence diversity is generated by differential splicing. CNTNs are highly enriched in the nervous system and they play important roles in axon guidance and axon-glial interactions, synapse formation and plasticity, and the organization of multi-protein complexes at the nodes of Ranvier [16,17]. Copy number variation encompassing CNTN genes and sequence variation in or near CNTN genes have been associated with a variety of neuro-developmental disorders, including autism, schizophrenia, Tourette syndrome, and mental retardation [1821].

A direct biochemical connection between APP and the Contactin (CNTN) family was discovered by Osterfield et al. [22], who showed (i) that among the six CNTNs, APP binds most strongly to CNTN3 and CNTN4, and (ii) that this binding interaction occurs between the N-terminal E1 domain of APP and the four membrane-proximal Fn domains of CNTN. More recently, a functional connection between APP and CNTN4 has been found in a study of retinal ganglion cell axon guidance in mice, in which (i) targeted mutation of App or Cntn4 produced a failure of axonal projections to the nucleus of the optic tract and (ii) Cntn4 over-expression produced a bias in axonal projections toward the nucleus of the optic tract that required the presence of APP [23].

These studies suggest that a more precise biochemical characterization of the APP-CNTN interaction would be of interest. In the present study, we have used site-directed mutagenesis to define the interfaces on APP and CNTN that mediate their mutual recognition.

Results

For in vitro binding assays, fusion proteins consisting of Contactin ECDs or their derivatives joined to the Fc domain of human IgG (“targets”) were immobilized in Protein-G coated microwells, and the microwells were then incubated with soluble fusion proteins consisting of the APP ECD or its derivatives joined to the catalytic domain of human placental alkaline phosphatase (AP) (“probes”). Bound probe was measured with a colorimetric substrate. Targets and probes were produced by transient transfection of HEK293T cells and collected as secreted proteins in serum-free conditioned medium (SFCM). The relative concentrations of targets and probes were estimated by (i) Coomassie Blue staining of protein-G-purified targets following SDS-PAGE and (ii) immunoblotting of SFCM using anti-human IgG (for targets) or anti-myc (for probes, which carry a myc-epitope tag). Examples of these immunoblots are shown in Figs 1F and 2D. To compare signals across multiple targets or probes, the volume of SFCM for each protein was adjusted with serum-free medium to equalize the target or probe concentrations across the set of samples. We note that both AP and Fc are homodimers [24], and therefore target-probe binding should exhibit enhanced avidity compared to binding between the corresponding monomeric target-probe pairs. This effect likely contributes to the high signal-to-noise ratio in the binding assays.

thumbnail
Fig 2. APP sequences required for binding to CNTN3.

(A) Left, 96-well binding assay using the AP fusions with the full APP ECD or its amino-terminal domains as probe and CNTN3-Fn(1–3)-Fc as target. Right, schematic of APP ECD domain structure, showing APP-AP fusions. (B) Sequence of APP CuBD showing the locations of alanine substitution mutations targeting groups of mostly polar surface amino acids (“first set”; upper sequence) or single amino acids (lower sequence). (C) 96-well binding assay showing alanine substitution mutations in the CuBD of APP-CuBD-AP binding to CNTN3-Fn(1–2)-Fc. (D) Immunoblots of serum-free conditioned medium containing the indicated WT and mutant APP-AP proteins, visualized with anti-myc mAb. All AP fusion proteins contain a myc-tag between the APP segment and AP. Mass in kDa is listed for the protein size standards, which are the same for the three left-most blots. Quantification of AP binding is shown in S1C–S1E Fig, and is summarized in S1 Table. The concentrations of the different probes were adjusted to provide equivalent amounts of AP enzyme activity, as determined by colorimetric assay.

https://doi.org/10.1371/journal.pone.0219384.g002

Identifying amino acids in CNTN3 that are required for APP binding

In an initial experiment, we compared the binding of the full APP ECD and the APP E1 domain, expressed as AP fusions, to each of the six CNTN ECDs, without their GPI anchor motifs, expressed as Fc fusions (Fig 1B). In general agreement with Osterfield et al. [22], both APP probes bound with similar efficiencies, and the relative affinities for the different CNTNs was: CNTN3 ≃ CNTN4 > CNTN5 >> CNTN1, CNTN2, and CNTN6. The one discrepancy with Osterfeld et al. [22] is with CNTN5: Fig 1B shows weak binding whereas Osterfeld et al. [22] saw no binding. Based on this experiment, we focused on CNTN3 for in-depth structure-function studies and we used the APP-E1-AP probe to assess binding to the CNTN3 derivatives.

Deletion of the six N-terminal Ig domains of CNTN3 had no effect on APP-E1-AP binding, thereby localizing the binding interface to the four Fn domains, in agreement with Osterfield et al. [22] (Fig 1C). Deletion of different combinations of Fn domains (in the absence of the Ig domains) revealed an essential role for Fn(2), with Fn(1) providing a several-fold enhancement of the binding signal (Fig 1C). Fn(3) and Fn(4) were found to be dispensable for binding. The signal intensities in this and subsequent binding assays are summarized in the figures and in S1 Table, where -, +/-, +, ++, +++, and ++++ represent the range of intensities from undetectable (-) to maximal (++++). Based on this analysis, alanine substitution mutagenesis experiments were conducted with a target Fc fusion protein containing only the first two Fn domains of CNTN3 (CNTN3-Fn(1–2)-Fc).

To define the amino acids within CNTN3 Fn(2) that mediate APP binding, we designed two rounds of alanine substitution mutagenesis guided by the crystal structure of Fn(2) PDB ID 5I99 [25]. In the first round of mutagenesis, 22 clusters of 1–4 amino acids each were substituted with alanine to narrow down the region(s) of interest. These amino acids were chosen based on their surface location in the Fn(2) crystal structure. Among the 22 mutant Fn(1–2)-Fc proteins, ten were expressed with yields roughly comparable to wild type (WT) Fn(1–2)-Fc (Fig 1F). These ten proteins are assumed to be correctly folded, and they are labeled alanine substitution (AS)1-10 in Fig 1D, upper sequences. AS2, AS6, AS7, AS8, and AS9 exhibited greatly reduced binding to APP-E1-AP, suggesting a role for one or more amino acid side chains in these clusters (Fig 1E, left panel and S1A Fig).

In the second round of mutagenesis, each of the 13 amino acids that were mutated in CNTN3-Fn(1–2)-Fc AS2, AS6, AS7, AS8, and AS9 was individually mutated to alanine (Fig 1D, lower sequences). Twelve of the 13 individual alanine substitution mutants were secreted with yields roughly comparable to WT (Fig 1F). E716A was produced with a poor yield and was not further studied. Among the 12 single alanine mutants, V752A showed undetectable binding, and R714A, R760A, and R764A showed greatly reduced binding (Fig 1E, right panel and S1B Fig), indicating an important role for each of these four amino acids in APP binding. It is interesting that three of these four amino acids are arginine.

Identifying amino acids in APP that are required for CNTN3 binding

We applied a similar strategy to localize the CNTN3 binding site within APP E1. We first tested the binding properties of the two subdomains of E1 –GFLD and CuBD–and observed that CuBD alone conferred binding to CNTN3 with essentially no contribution from GFLD (Fig 2A). APP CuBD contains a Type 2 non-blue copper center in which His147, His151, and Tyr168 bind the central copper ion [26].

To further localize the amino acids within APP CuBD responsible for CNTN3 binding, nine clusters of 1–5 alanine substitution mutations were constructed in nearby surface residues in the APP-CuBD-AP fusion protein, based on the CuBD and E1 crystal structures PDB ID 2FKL and PDB ID 3KTN [26, 27] (Fig 2B AS 1–9, upper sequences). All nine variants were secreted with yields roughly comparable to WT (Fig 2D). In the binding assays with WT CNTN3-Fn(1–2)-Fc, APP-CuBD-AP mutants AS2 and AS8 showed no binding, AS1 and AS3 showed strongly reduced binding, and AS4 showed modestly reduced binding (Fig 2C, left panel and S1C Fig). Interestingly, mutating His147 (in AS5), which is one of the copper-liganding residues, did not affect binding.

In the second round of mutagenesis, each of the 12 amino acids that were mutated in APP-CuBD-AP AS1, AS2, AS3, AS4, and AS8 was individually mutated to alanine (Fig 2B, lower sequences). All 12 mutants were expressed with yields roughly comparable to WT (Fig 2D), and, among these, H137A and E183A showed undetectable binding, and K132A, Q138A, E139A, R140A, M141A, K178A, and R180A showed a several fold reduction in binding (Fig 2C, right two panels and S1D and S1E Fig).

Spatial locations of amino acids that are essential for APP-CNTN3 binding

The locations of the alanine substitution mutations were mapped onto ribbon diagrams of CNTN3 Fn(2) and APP CuBD, color-coded by their effect on binding (Fig 3). The four CNTN3 Fn(2) substitutions that most severely disrupt binding reside on one face of the Fn(2) domain (Fig 3A). Similarly, the two APP CuBD substitutions that most severely disrupt binding reside on adjacent strands of a beta-sheet on one face of the CuBD (Fig 3B). Five of the six APP CuBD substitutions with several-fold lower binding efficiency (Fig 3B, color-coded blue) reside adjacent to these two critical positions, and the sixth resides further away on the same face of the CuBD. As the mutations with the greatest effect on CNTN3 Fn(2) and APP CuBD binding reside on one face of each of these domains, the most parsimonious explanation for the mutagenesis data is that these two faces interact.

thumbnail
Fig 3. Amino acids that are critical for APP-CNTN3 binding displayed on the 3-dimensional structures of CNTN3-Fn(2) and APP-CuBD.

(A) Locations of alanine substitutions in CNTN3-Fn(2) PDB ID 5I99 [25], color coded by their effect on APP-E1-AP binding. (B) Locations of alanine substitutions in APP CuBD PDB ID 2FKL [26], color coded by their effect on CNTN-Fn(1–2)-Fc binding. The cylinders and arrows show the 90 degree rotations that relate the images in adjacent panels. Only data from CNTN-Fn(1–2)-Fc or APP-CuBD-AP fusion proteins that were secreted well are shown. In the leftmost panels, side chains are labeled for those amino acid that are most critical for binding, i.e., those for which alanine substitution severely impairs binding (categories: +/- (magenta) and–(red)).

https://doi.org/10.1371/journal.pone.0219384.g003

Discussion

In this study, we have confirmed and extended the findings of Osterfeld et al. [22], by showing that: (i) binding of the full ECD or the E1 domain of APP occurs with affinities CNTN3 ≃ CNTN4 > CNTN5 >> CNTN1, CNTN2, and CNTN6; (ii) in CNTN3, APP binds to Fn(2); (iii) in APP, CNTN3 binds to the CuBD; and (iv) the most important amino acids for binding reside on one face of CNTN3-Fn(2) and on one face of APP-CuBD. The first of these observations is consistent with the relative sequence similarity among members of the Contactin family: CNTN1-2 and CNTN3-6 form two main groups of related sequences, and, within the second group, CNTN3 and CNTN4 are the most closely related pair [15].

Multiple interacting protein partners have been identified for both the APP/APLP and CNTN families, and it seems likely that both protein families function via a network of cis- and trans-acting partners. Additionally, both families generate cell surface and secreted protein isoforms, implying that they can act both locally or at longer range. As noted in the Introduction, only one study has explored functional interactions between mammalian CNTN and APP in vivo [23]. In addition to the retinal ganglion cell axon guidance defects found in Cntn4 knockout mice [23], glomeruli in the olfactory exhibit reduced specificity of innervation by olfactory receptor neurons [28]. By analogy with the experiments of Osterhout et al. [23], it would be of interest to determine whether App or Aplp deletion or over-expression also affects the olfactory bulb phenotype. A similar approach could also be used to look for App or Aplp affects on the phenotypes associated with targeted mutation in Cntn5, which include defects in the auditory system [29], the development of axo-axonic synapses in the spinal cord [30], and dendritic patterning among retinal ganglion cells [31].

Invertebrate genomes also code for APP and CNTN family members. Conveniently, the two best studied invertebrate model organisms, C. elegans and D. melanogaster, each have only a single APP homologue (APL-1 in C. elegans [32] and APPL in D. melanogaster [33]) and a single CNTN homologue (RIG-6 in C. elegans [34] and CONT in D. melanogaster [35]). At present, there are no reports of APP-CNTN interactions in C. elegans or D. melanogaster. However, in the developing nervous system of the hawkmoth Manduca sexta, interactions between APPL (the M. sexta homologue of APP), which is expressed by migratory neurons, and MsContactin (the M. sexta Contactin homologue), which is expressed on adjacent glia, are important for guiding neuronal migration and outgrowth [36]. The M. sexta experiments suggest that there may be an ancient evolutionary connection between these two families of proteins. It would be of interest to determine whether the M. sexta APPL and MsContactin proteins exhibit the same mode of binding as the mammalian APP and CNTN3 proteins.

The multitude of protein partners for both APP/APLP and CNTN family members complicate the interpretation of genetic interaction experiments based on gene deletion or over-expression because such gross genetic perturbations can produce indirect effects that are mediated by protein partners other than the ones under consideration. The present experiments lay the groundwork for a more incisive analysis of CNTN-APP interactions in vivo by defining a minimal set of mutations that alter their mutual binding without affecting protein structure or stability. Thus, introducing a minimal set of mutations into either of these two genes by CRISPR/Cas9 editing should produce an allele in which the APP-CNTN interaction is selectively eliminated, while preserving most or all of the binding interactions with other partners.

Materials and methods

Constructs

Fc and AP fusions were cloned into the pRK5 vector for expression in HEK/293T cells [37]. For the Fc-fusion proteins, the open reading frame consisted of (i) the native signal peptide or the mouse Frizzled8 signal peptide, (ii) the full ECD or the subdomain of interest, (iii) a 22 amino acid linker corresponding to the region immediately distal to the cysteine-rich region of mouse Frizzled8, and (iv) the Fc region of human IgG1 starting from the hinge [38]. For the AP-fusion proteins, the open reading frame consisted of: (i) the native signal peptide or the mouse Frizzled8 signal peptide, (ii) the full ECD or the subdomain of interest, (iii) a glycine/serine linker followed by a myc epitope tag, and (iv) the coding sequence of human placental alkaline phosphatase (PLVAP) lacking the GPI-anchoring sequences [39].

CNTN and APP inserts

Full-length CNTN2, CNTN4.1, CNTN5, and APP ECDs were amplified from a mixture of prenatal mouse embryo and neonatal mouse brain cDNA by nested PCR. Mouse CNTN1, CNTN3, and CNTN6 ECD-Fc constructs were a generous gift of Dr. Davide Comoletti. CNTN3 and APP domain deletions and alanine substitutions were generated by PCR using the cloned ECD sequences as templates.

Cell culture and transfection

HEK/293T cells (ATCC CRL-11268) were transiently transfected with polyethylenimine (PEI). For small-scale transfections, HEK293T cells were grown in 12-well plates. 24 hours after transfection, wells were washed twice with 1 ml serum-free medium (SFM), and then cultured for an additional 24 hours in 0.5 ml SFM. The serum-free conditioned medium (SFCM) was centrifuged at 5000xg for 5 minutes, and 20 ul of the supernatant was used for immunoblotting. Plasmids that produced ~1 ug/ml protein in the SFCM were used for large-scale transfection. For large-scale transfections, HEK/293T cells were plated in 10 cm dishes and processed as described above, except that (i) washes were performed three times with 5 mls SFM, (ii) 5 mls SFCM was generated per plate, and (iii) SFCM production was continued with a fresh 5 ml of SFM for an additional day, if more protein was needed. SFCM was filtered through a 0.2 um filter, and stored at 4°C with sodium azide added to 0.01%.

Binding assays

For estimating Fc fusion protein concentrations, 1 ml of SFCM was captured on Protein-G sepharose beads (Thermo-Fisher), and analyzed by SDS-PAGE and Coomassie Blue staining. Protein concentrations were compared to a bovine serum albumin (BSA) dilution series. Relative protein concentrations for WT and mutant Fc- and AP-fusion proteins were determined using the LI-COR fluorescent immunoblotting system (LI-COR Biosciences), and the volumes of SFCM adjusted accordingly to equalize the concentrations across WT and mutant versions in the binding assays. For binding assay, the wells of a Protein-G coated 96-well plate (Thermo-Fisher Scientific/Pierce) were washed twice with PBS, 100 μl of volume-adjusted conditioned medium containing each Fc-fusion proteins was added per well, and then the plate was rotated horizontally for 1.5–2 hours at room temperature. The medium was removed and the wells washed 5 times in PBS with 0.5% Triton X-100, 0.1% bovine serum albumin (PBSTA). 100 μl of volume-adjusted conditioned medium containing the AP-fusion protein was added per well, and the plate was rotated for 1.5–2 hours at room temperature, and then washed 5 times with PBSTA. Five minutes before use, a 1:1 mixture of the BluePhos Microwell Substrate (Kirkegaard and Perry Laboratories) was prepared and 100 μl was added to each well. Absorbance at 620 nm was recorded at 5 minutes intervals at room temperature using a SpectraMax M3 Microplate Reader (Molecular Devices) for 60–90 minutes. Plates were photographed after the incubation was complete.

Supporting information

S1 Fig.

Quantification of the binding reactions between APP-E1-AP and alanine substitution mutants of CNTN3-Fn(1–2)-Fc (A and B) and between alanine substitution mutants of APP-CuBD-AP and CNTN3-Fn(1–2)-Fc (C-E). The AP reaction utilized the Blu-Phos Microwell substrate and was monitored at 5-minute intervals over 60–90 minutes at room temperature with a 96-well plate reader.

https://doi.org/10.1371/journal.pone.0219384.s001

(EPS)

S1 Table. Binding properties of CNTN3 and APP.

The CNTN3 binding data refer to Fig 1 and S1A and S1B Fig. The APP binding data refer to Fig 2 and S1C–S1E Fig.

https://doi.org/10.1371/journal.pone.0219384.s002

(DOCX)

Acknowledgments

We thank Amir Rattner for performing RT-PCR on mouse embryo cDNA, Davide Comoletti for gifts of CNTN cDNA clones, Solomon Snyder for the use of his plate reader, James Berger and Chris Cho for assistance with visualizing and analyzing protein structures, and Chris Cho and Amir Rattner for advice and discussions.

References

  1. 1. Müller UC, Deller T, Korte M. Not just amyloid: physiological functions of the amyloid precursor protein family. Nat Rev Neurosci. 2017 May;18(5):281–298. pmid:28360418
  2. 2. Coburger I, Hoefgen S, Than ME. The structural biology of the amyloid precursor protein APP—a complex puzzle reveals its multi-domain architecture. Biol Chem. 2014 May;395(5):485–98. pmid:24516000
  3. 3. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer's disease at 25 years. EMBO Mol Med. 2016 Jun 1;8(6):595–608. pmid:27025652
  4. 4. Wang P, Yang G, Mosier DR, Chang P, Zaidi T, Gong YD, et al. Defective neuromuscular synapses in mice lacking amyloid precursor protein (APP) and APP-Like protein 2. J Neurosci. 2005 Feb 2;25(5):1219–25. pmid:15689559
  5. 5. Wang Z, Wang B, Yang L, Guo Q, Aithmitti N, Songyang Z, et al. Presynaptic and postsynaptic interaction of the amyloid precursor protein promotes peripheral and central synaptogenesis. J Neurosci. 2009 Sep 2;29(35):10788–801. pmid:19726636
  6. 6. Lee KJ, Moussa CE, Lee Y, Sung Y, Howell BW, Turner RS, et al. Beta amyloid-independent role of amyloid precursor protein in generation and maintenance of dendritic spines. Neuroscience. 2010 Aug 11;169(1):344–56. pmid:20451588
  7. 7. Tyan SH, Shih AY, Walsh JJ, Maruyama H, Sarsoza F, Ku L, et al. Amyloid precursor protein (APP) regulates synaptic structure and function. Mol Cell Neurosci. 2012 Aug;51(1–2):43–52. pmid:22884903
  8. 8. Weyer SW, Klevanski M, Delekate A, Voikar V, Aydin D, Hick M, et al. APP and APLP2 are essential at PNS and CNS synapses for transmission, spatial learning and LTP. EMBO J. 2011 Jun 1;30(11):2266–80. pmid:21522131
  9. 9. Hick M, Herrmann U, Weyer SW, Mallm JP, Tschäpe JA, Borgers M, et al. Acute function of secreted amyloid precursor protein fragment APPsα in synaptic plasticity. Acta Neuropathol. 2015 Jan;129(1):21–37. pmid:25432317
  10. 10. Kang J, Lemaire HG, Unterbeck A, Salbaum JM, Masters CL, Grzeschik KH, et al. The precursor of Alzheimer's disease amyloid A4 protein resembles a cell-surface receptor. Nature. 1987 Feb 19–25;325(6106):733–6. pmid:2881207
  11. 11. Caillé I, Allinquant B, Dupont E, Bouillot C, Langer A, Müller U, et al. Soluble form of amyloid precursor protein regulates proliferation of progenitors in the adult subventricular zone. Development. 2004 May;131(9):2173–81. pmid:15073156
  12. 12. Nikolaev A, McLaughlin T, O'Leary DD, Tessier-Lavigne M. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature. 2009 Feb 19;457(7232):981–9. pmid:19225519
  13. 13. A transcriptionally active complex of APP with Fe65 and histone acetyltransferase Tip60. Cao X1, Südhof TC. Science. 2001 Jul 6;293(5527):115–20. pmid:11441186
  14. 14. Cao X, Südhof TC. Dissection of amyloid-beta precursor protein-dependent transcriptional transactivation. J Biol Chem. 2004 Jun 4;279(23):24601–11. pmid:15044485
  15. 15. Zuko A, Bouyain S, van der Zwaag B, Burbach JP. Contactins: structural aspects in relation to developmental functions in brain disease. Adv Protein Chem Struct Biol. 2011;84:143–80. pmid:21846565
  16. 16. Gennarini G, Bizzoca A, Picocci S, Puzzo D, Corsi P, Furley AJW. The role of Gpi-anchored axonal glycoproteins in neural development and neurological disorders. Mol Cell Neurosci. 2017 Jun;81:49–63. pmid:27871938
  17. 17. Chatterjee M, Schild D, Teunissen CE. Contactins in the central nervous system: role in health and disease. Neural Regen Res. 2019 Feb;14(2):206–216. pmid:30530999
  18. 18. Fernandez T, Morgan T, Davis N, Klin A, Morris A, Farhi A, et al. Disruption of contactin 4 (CNTN4) results in developmental delay and other features of 3p deletion syndrome. Am J Hum Genet. 2004 Jun;74(6):1286–93 pmid:15106122
  19. 19. Roohi J, Montagna C, Tegay DH, Palmer LE, DeVincent C, Pomeroy, et al. Disruption of contactin 4 in three subjects with autism spectrum disorder. J Med Genet. 2009 Mar;46(3):176–82. pmid:18349135
  20. 20. Huang AY, Yu D, Davis LK, Sul JH, Tsetsos F, Ramensky V, et al. Rare Copy Number Variants in NRXN1 and CNTN6 Increase Risk for Tourette Syndrome. Neuron. 2017 Jun 21;94(6):1101–1111. pmid:28641109
  21. 21. Oguro-Ando A, Zuko A, Kleijer KTE, Burbach JPH. A current view on contactin-4, -5, and -6: Implications in neurodevelopmental disorders. Mol Cell Neurosci. 2017 Jun;81:72–83. pmid:28064060
  22. 22. Osterfield M, Egelund R, Young LM, Flanagan JG. Interaction of amyloid precursor protein with contactins and NgCAM in the retinotectal system. Development. 2008 Mar;135(6):1189–99. pmid:18272596
  23. 23. Osterhout JA, Stafford BK, Nguyen PL, Yoshihara Y, Huberman AD. Contactin-4 mediates axon-target specificity and functional development of the accessory optic system. Neuron. 2015 May 20;86(4):985–999. pmid:25959733
  24. 24. Le Du MH, Stigbrand T, Taussig MJ, Menez A, Stura EA. Crystal structure of alkaline phosphatase from human placenta at 1.8 A resolution. Implication for a substrate specificity. J Biol Chem. 2001 Mar 23;276(12):9158–65. pmid:11124260
  25. 25. Nikolaienko RM, Hammel M, Dubreuil V, Zalmai R, Hall DR, Mehzabeen N, et al. Structural Basis for Interactions Between Contactin Family Members and Protein-tyrosine Phosphatase Receptor Type G in Neural Tissues. J Biol Chem. 2016 Oct 7;291(41):21335–21349. pmid:27539848
  26. 26. Kong GK, Adams JJ, Harris HH, Boas JF, Curtain CC, Galatis D, et al. Structural studies of the Alzheimer's amyloid precursor protein copper-binding domain reveal how it binds copper ions. J Mol Biol. 2007 Mar 16;367(1):148–61. pmid:17239395
  27. 27. Dahms SO, Hoefgen S, Roeser D, Schlott B, Gührs KH, Than ME. Structure and biochemical analysis of the heparin-induced E1 dimer of the amyloid precursor protein. Proc Natl Acad Sci U S A. 2010 Mar 23;107(12):5381–6. pmid:20212142
  28. 28. Kaneko-Goto T, Yoshihara S, Miyazaki H, Yoshihara Y. BIG-2 mediates olfactory axon convergence to target glomeruli. Neuron. 2008 Mar 27;57(6):834–46. pmid:18367085
  29. 29. Li H, Takeda Y, Niki H, Ogawa J, Kobayashi S, Kai N, et al. Aberrant responses to acoustic stimuli in mice deficient for neural recognition molecule NB-2. Eur J Neurosci. 2003 Mar;17(5):929–36. pmid:12653969
  30. 30. Ashrafi S, Betley JN, Comer JD, Brenner-Morton S, Bar V, Shimoda Y, et al. Neuronal Ig/Caspr recognition promotes the formation of axoaxonic synapses in mouse spinal cord. Neuron. 2014 Jan 8;81(1):120–9. pmid:24411736
  31. 31. Peng YR, Tran NM, Krishnaswamy A, Kostadinov D, Martersteck EM, Sanes JR. Satb1 Regulates Contactin 5 to Pattern Dendrites of a Mammalian Retinal Ganglion Cell. Neuron. 2017 Aug 16;95(4):869–883. pmid:28781169
  32. 32. Hornsten A, Lieberthal J, Fadia S, Malins R, Ha L, Xu X, et al. APL-1, a Caenorhabditis elegans protein related to the human beta-amyloid precursor protein, is essential for viability. Proc Natl Acad Sci U S A. 2007 Feb 6;104(6):1971–6. pmid:17267616
  33. 33. Cassar M, Kretzschmar D. Analysis of Amyloid Precursor Protein Function in Drosophila melanogaster. Front Mol Neurosci. 2016 Jul 26;9:61. pmid:27507933
  34. 34. Katidou M, Tavernarakis N, Karagogeos D. The contactin RIG-6 mediates neuronal and non-neuronal cell migration in Caenorhabditis elegans. Dev Biol. 2013 Jan 1;373(1):184–95. pmid:23123963
  35. 35. Faivre-Sarrailh C, Banerjee S, Li J, Hortsch M, Laval M, Bhat MA. Drosophila contactin, a homolog of vertebrate contactin, is required for septate junction organization and paracellular barrier function. Development. 2004 Oct;131(20):4931–42. pmid:15459097
  36. 36. Ramaker JM, Swanson TL, Copenhaver PF. Manduca Contactin Regulates Amyloid Precursor Protein-Dependent Neuronal Migration. J Neurosci. 2016 Aug 17;36(33):8757–75. pmid:27535920
  37. 37. Gorman CM. Mammalian cell expression. Curr Opin Biotechnol. 1990 Oct;1(1):36–47. pmid:1367916
  38. 38. Aruffo A, Stamenkovic I, Melnick M, Underhill CB, Seed B. CD44 is the principal cell surface receptor for hyaluronate. Cell. 1990 Jun 29;61(7):1303–13. pmid:1694723
  39. 39. Smallwood PM, Williams J, Xu Q, Leahy DJ, Nathans J. Mutational analysis of Norrin-Frizzled4 recognition. J Biol Chem. 2007 Feb 9;282(6):4057–68. pmid:17158104