Chapter 9 the Down Syndrome Cell Adhesion Molecule

  • Upload
    silax

  • View
    223

  • Download
    0

Embed Size (px)

Citation preview

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    1/16

    Chapter 9

    The Down Syndrome Cell Adhesion Molecule

    Hitesh Kathuria and James C. Clemens

    Abstract The Down syndrome cell adhesion molecules (DSCAMs) are a struc-

    turally and functionally conserved family of cell surface receptors that playimportant roles in nervous system organization. These receptors are expressed

    on both axons and dendrites where they engage in isoform-specific binding

    interactions between DSCAM receptors on opposing cell surfaces. Massive

    alternative splicing of arthropod DSCAM transcripts greatly expands the

    complexity of the DSCAM family by endowing these organisms with the ability

    to produce tens of thousands of distinct receptor isoforms that undergo homo-

    philic binding. In addition to homophilic binding, DSCAM extracellular

    domains serve as receptors for other proteins such as the attractant netrin-1.

    These diverse interaction properties allow DSCAMs to control a variety ofnervous system patterning processes including axon path-finding and targeting,

    neurite branch segregation, self-recognition, and neurite tiling.

    Keywords DSCAM Ig domain Alternative splicing Axon guidance Neuron Synapse Drosophila

    9.1 Introduction

    In 1998, Julie Korenbergs group at the Cedars-Sinai Research Institute identi-

    fied a new member of the immunoglobulin superfamily of cell surface receptors

    (Yamakawa et al. 1998). Structurally, this protein was similar to previously

    identified proteins that function as cell adhesion molecules (CAMs), but it

    possesses a unique arrangement of domains within the extracellular region.

    The gene encoding this novel cell surface receptor is located on human chromo-

    some 21 in band 21q22. This is significant because an increased expression of

    J.C. Clemens (*)

    Department of Biochemistry, Purdue University, 175 S. University St., West Lafayette,IN 47907, USA

    e-mail: [email protected]

    M. Hortsch, H. Umemori (eds.),The Sticky Synapse,

    DOI 10.1007/978-0-387-92708-4_9, Springer ScienceBusiness Media, LLC 2009

    207

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    2/16

    genes in this chromosomal region correlates with the manifestation of Down

    syndrome (Ds) phenotypes, including mental retardation (Korenberg et al.

    1992, Delabar et al.1993, Korenberg et al.1994). Northern blot analysis and

    in situ hybridization studies revealed that this gene is broadly expressed within

    the nervous system (Yamakawa et al.1998). Based on these observations, theprotein was dubbed the Down syndrome cell adhesion molecule (DSCAM,

    pronounced [de-es-kam) (Yamakawa et al.1998).

    Since this initial report, the DSCAM family has grown with the discovery of

    paralogs and homologs in mammals, fish, birds, insects, sea urchins, crusta-

    ceans, and mollusks. In chordates DSCAM is generally written in all capital

    letters, while non-chordate Dscams are written with only the first letter capita-

    lized. DSCAM receptors are expressed throughout the nervous system during

    development and have been found to play widespread roles in patterning the

    nervous system, including axon targeting, neurite arborization, and branchsegregation (Zipursky et al. 2006, Schmucker 2007). An additional role for

    hyper-variable arthropod Dscams has been described in innate immunity, but

    this role is beyond the scope of this chapter and will therefore not be described

    (for reviews on this subject, see Watson et al.2005, Dong et al.2006).

    9.2 Identification of DSCAM Family Members

    As mentioned previously, the founding member of the DSCAM family was

    identified as a novel member of the immunoglobulin superfamily and as a

    putative cell adhesion molecule (Yamakawa et al.1998). Since the gene encod-

    ing this membrane receptor localizes to human chromosome band

    21q22.222.3, a region that is critical for the neurological phenotypes of Ds, it

    was tempting to speculate that duplication of the DSCAM gene might play a

    causative role in the production of these phenotypes. The finding that DSCAM

    is expressed largely in the developing nervous system is consistent with a

    putative role in Ds. However, a subsequent study (Ronan et al. 2007) further

    demarcated the Ds critical region on chromosome 21 that is responsible for the

    generation of Ds-related phenotypes and determined that the DSCAM gene

    resided outside of this region. This suggests that DSCAM is unlikely to be a

    causative agent in the development of Down syndrome.

    In an effort to further characterize DSCAM function, Yamakawa et al.

    (1998) isolated mouse homologs of DSCAM by using the human cDNA

    sequence as a probe to screen a mouse brain cDNA library. Tissue in situ

    hybridization of mouse DSCAM on mouse tissues revealed that DSCAM

    expression is largely localized to the central nervous system. The timing of its

    expression suggests that DSCAM may play a role during early nervous system

    development.

    The third DSCAM family member that was identified came from the class

    Insecta. Drosophila Dscam was first characterized as an unknown tyrosine

    208 H. Kathuria and J.C. Clemens

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    3/16

    phosphorylated protein that physically associates with the Dock (Dreadlocks)

    SH2 domain (Schmucker et al. 2000). Dock is an adapter protein that comprises

    three SH3 domains and one SH2 domain and is required for axon guidance in

    flies (Clemens et al. 1996, Garrity et al. 1996). Since dock mutants exhibit defects

    in axon guidance during the development of the visual system in an adult fly(Garrity et al.1996), as well as in the embryonic nervous system (Desai et al.

    1999), it was proposed that Dock serves as a vital link that connects targeting

    receptors to downstream regulators of the actin cytoskeleton to control neural

    patterning. This hypothesis was further supported by Hing et al. (1999) with the

    discovery that a known regulator of the actin cytoskeleton, Pak (p21-activated

    kinase), physically and genetically interacts with dock to control axon guidance.

    These findings suggest that manipulating Pak signaling is one mechanism that

    insect Dscam family members employ to control connectivity within the ner-

    vous system. Additionally, a subsequent human DSCAM study found thatDSCAM directly binds to Pak and stimulates Pak phosphorylation and activity

    (Li and Guan 2004). This suggests that DSCAM-mediated control of Pak

    activity appears to be a general property of all DSCAM receptors.

    Following the identification of DSCAM genes in human, mouse, and fruit

    flies, DSCAM homologs have been identified primarily by sequence database

    comparisons in a number of other species. DSCAMs appear to be present in

    most animals that have a nervous system. The notable exception is theCaenor-

    habditis elegans genome, which lacks a DSCAM gene product that exhibits clear

    sequence conservation. Despite the growing number of DSCAM reports inother species, most of what we know about DSCAM function comes from

    Drosophila. Therefore, much of this chapter will center onDrosophila Dscam

    function with some additional examples of vertebrate DSCAM functions.

    9.3 General Domain Structure

    DSCAMs are type I cell surface transmembrane receptors that belong to the

    immunoglobulin (Ig) superfamily (Yamakawa et al. 1998). Members of the

    DSCAM protein family are made up of approximately 2,000 amino acids

    with an average molecular weight of 221 kDa. The amino acid sequence and

    domain structure of the extracellular region is conserved and encompasses ten

    Ig domains and six fibronectin type III modules (Fig. 9.1). Nine of the Ig

    domains are tandemly arrayed in the membrane distal (N-terminal) region of

    the extracellular domain. The six fibronectin type III modules are tandemly

    arrayed in the membrane proximal region with the tenth Ig domain located

    between fibronectin modules four and five. This unique arrangement of Ig and

    fibronectin domains is the hallmark that distinguishes DSCAM receptors from

    other Ig superfamily members.

    The extracellular domain is connected to the cytoplasmic region of DSCAM

    by a single membrane-spanning domain. In the case of human DSCAM, it has

    9 The Down Syndrome Cell Adhesion Molecule 209

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    4/16

    been shown that the transmembrane domain is necessary and sufficient for

    association of the DSCAM receptor with the netrin-binding receptor deleted in

    colorectal carcinomas (DCC) (Ly et al.2008). It is unclear whether this is a

    general property of all DSCAM receptors or whether this is a property that is

    specific for human DSCAM.

    The cytodomains of DSCAMs range in size from approximately 300 to 400amino acids and contain no previously characterized catalytic domains or

    substantial sequence motifs. DSCAM family members within the same phylum

    tend to have cytoplasmic domain sequences that are fairly well conserved.

    However, comparison of cytoplasmic DSCAM sequences across the phyla

    (Arthropoda, Chordata, Platyhelminthes, Echinodermata, and Mollusca) reveals

    little sequence conservation. In general, DSCAM cytodomains contain multiple

    tyrosines, which are thought to serve as binding sites for SH2 domain-containing

    proteins such as Dock, as well as a C-terminal putative PDZ domain-binding

    site. PDZ (named after the first three letters of proteins containing this domain:PSD95, DlgA, and ZO-1) domain-containing proteins frequently bind to the C-

    terminal sequences of transmembrane receptors and serve as scaffolds that hold

    together signaling complexes (Ponting et al. 1997). No proteins have been

    reported to interact with this site in DSCAMs and it is currently unknown if

    this region functionally interacts with PDZ domain-containing proteins.

    9.4 DSCAM Molecular Diversity

    DSCAM transcripts usually undergo alternative splicing. Therefore (most or

    all),DSCAMgenes typically express multiple protein isoforms. The extent and

    complexity ofDSCAMgene transcript alternative splicing varies greatly among

    DSCAM Domain Structure

    signal peptide Ig domainFibronectin III

    domain

    Transmembrane

    domain

    Ectodomain Cytodomain

    Fig. 9.1 DSCAM domain structure. The general domain structure of DSCAM receptor family

    members is shown. Symbols representing subdomains within DSCAMs are labeled below the

    receptor structure. The DSCAM ectodomain is separated from the cytodomain by a single

    transmembrane domain segment. The majority of DSCAM receptor sequences are located in

    the ectodomain, which is composed of an N-terminal signal peptide, ten Ig domains and sixfibronectin type III domains in the order shown. The C-terminal cytodomain contains no

    catalytic domains or substantial sequence motifs

    210 H. Kathuria and J.C. Clemens

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    5/16

    species. Non-arthropodDSCAM transcripts undergo little if any alternative

    splicing (Barlow et al. 2002). In contrast, unprecedented alternative splicing

    of some of the arthropodDscam transcripts leads to the production of tens

    of thousands of different Dscam protein isoforms (Schmucker et al. 2000,

    Graveley et al. 2004, Watson et al. 2005, Brites et al. 2008). Thus far, in allarthropods examined (fruit fly, mosquito, honey bee, beetle and Daphnia) a

    Dscam gene has been detected that exhibits hyper-variable alternative splicing.

    However, Dscam paralogs that do not exhibit hyper-variable splicing have also

    been described in these organisms (Funada et al.2007, Millard et al.2007).

    TheDrosophila Dscamgene serves as an example to illustrate hyper-variable

    Dscamsplicing and the impact this splicing has on Dscam receptor molecular

    diversity. TheDrosophila melanogaster genome contains four Dscam genes. The

    first that was identified is called Dscam and the subsequently identified paralogs

    are known asDscam2,Dscam3, andDscam4(Millard et al.2007). Of these fourgenes, only the (original)Dscam gene product is hyper-variable through the

    process of alternative splicing.Dscam2 has been shown to encode two splice

    variants, whileDscam3 and Dscam4 are predicted to encode a single protein

    product (Millard et al.2007).

    The Drosophila Dscam gene contains 115 exons (Fig.9.2). Twenty of these

    are considered as constant exons and are present in allDscamtranscripts, while

    95 are variable and their inclusion in the Dscam mRNA is controlled by

    alternative splicing (Schmucker et al.2000). Each mature Dscam mRNA com-

    prises 24 exons: 20 constant exons and 4 variable exons (Fig.9.2). The variableexons are exons 4, 6, 9, and 17, which are arrayed within theDscam gene in

    linear clusters. Exon 4 has 12 alternatives, exon 6 has 48 alternatives, exon 9 has

    33 alternatives, and exon 17 has 2 alternatives (Fig.9.2). Alternative exons are

    included in the Dscam mRNA in a mutually exclusive fashion such that each

    mRNA will contain exactly one of each alternative exon 4, 6, 9, and 17. As a

    result, all Dscam mRNAs encode Dscam protein isoforms that share the same

    overall domain structure, but differ in the amino acid sequence at four distinct

    regions. The first half of Ig domain 2 is encoded by alternative exon 4, the first

    half of Ig domain 3 is encoded by alternative exon 6, Ig domain 7 is encoded byalternative exon 9, and the transmembrane domain is encoded by alternative

    exon 17.

    This extraordinary example of alternative splicing lendsDrosophila Dscama

    unique advantage over other cell adhesion genes in that, by itself, theDscam

    gene encodes a large family of molecularly diverse cell surface receptors. Math-

    ematically, a total of 19,008 (12 48 33) different extracellular domains fused

    to one of two different transmembrane domains (resulting in 38,016 isoforms)

    can be produced by the flyDscam gene (Schmucker et al.2000). A standard

    nomenclature has been adopted by Dscam researchers to designate the usage of

    alternative exons. The particular variant within an alternative exon cluster is

    represented by a code consisting of two numbers separated by a decimal point.

    For example, the second variant within the alternative exon 4 array is repre-

    sented as 4.2 in this scheme. To refer to a particular Dscam splice form that

    9 The Down Syndrome Cell Adhesion Molecule 211

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    6/16

    contains alternative exons 4.2, 6.20, 9.27, and 17.1 the first numbers of the code

    are dropped and the alternative exon variant numbers are listed sequentially

    and separated by decimal points: 2.20.27.1.

    Dscam mRNA expression studies reveal that all alternative exons appear to

    be used with the exception of alternative exons 6.11 and 9.33, which have not

    been detected in any Dscam cDNA (Neves et al.2004). Splicing of the alter-

    native exons appears to be independent of each other such that random combi-

    nations of exons 4, 6, and 9 are produced. Individual neurons are predicted to

    contain 1450 copies of the Dscam mRNA, and it is known that this population

    consists of a mixture of different splice forms (Neves et al.2004).

    In general, splicing of the alternative exons appears to be random; however,

    developmental and tissue-specific alternative exon trends have been observed.

    Within the alternative exon 4 cluster, exon 4.2 is rarely used in Drosophila

    embryos but its usage greatly increases in later developmental stages (Celotto

    and Graveley2001, Neves et al.2004). Splicing of alternative exon 6 appears to

    be random. The greatest splicing biases occur during alternative exon 9 selec-

    tion. Alternative exons 9.6, 9.9, 9.13, 9.30, and 9.31 are highly favored in the

    embryonic stage, whereas during later stages of development a more even

    distribution of exon 9 variants are used (Neves et al.2004). Exon 9 also appears

    Exon 4

    12 Alternatives

    Exon 6

    48 Alternatives

    Exon 9

    33 Alternatives

    Exon 17

    2 Alternatives

    Gene

    mRNA

    4 6 9 17

    Protein

    2 3 7

    Drosophila Dscam

    Fig. 9.2 Drosophila Dscam gene, transcript, and protein structure. The Drosophila Dscam gene,

    transcript, and protein are shown. Constant exons are represented as black vertical barsin the

    gene structure and dark gray segments in the mRNA structure. Variable exons are represented

    as shortergray barsin the gene structure and aslight graysegments in the mRNA structure.

    The constant exons are present in all Dscam mRNAs while only one of each variable domain

    exon is selected for inclusion in the Dscam mRNA. All Dscam protein isoforms share the

    same overall domain structure as shown, but differ in sequence in the gray-coloredregions

    comprising the N-terminal halves of Ig domains 2 and 3, all of Ig domain 7, and the

    transmembrane domain

    212 H. Kathuria and J.C. Clemens

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    7/16

    to have a tissue-specific bias. For example, photoreceptor cells selectively use

    the same five variants that are prevalent in the embryo, while other neurons use

    a broader repertoire (Neves et al. 2004). At present the significance of the

    developmental and tissue-specific splicing preferences is not known.

    9.5 Homophilic Interactions

    The extracellular domain of a DSCAM receptor expressed on one cell surface

    can physically interact with the extracellular domain of a DSCAM receptor

    located on a neighboring cell membrane (Agarwala et al.2000). These interac-

    tions, which span two cellular membranes, are referred to astransinteractions

    and only occur between two identical DSCAM receptor proteins (homophilic).For example, there are two DSCAM paralogs in chicken: DSCAM and

    DSCAML (Yamagata and Sanes 2008). Non-neuronal cultured cells expressing

    DSCAM will aggregate due to the trans homophilic interactions of DSCAM

    ectodomains. Similarly, cells expressing DSCAML will also form aggregates

    through DSCAML ectodomaintrans homophilic interactions. However, cells

    expressing DSCAM do not form heterophilic interaction with cells expressing

    DSCAML (Yamagata and Sanes2008).

    This homophilic-specific binding property appears to be a conserved attri-

    bute shared by all DSCAM family members. Perhaps even more remarkablethan the sheer number of Drosophila Dscam ectodomains (19,008) is the

    way these isoforms interact with each other. Dscam isoforms interact intrans

    (Fig. 9.3), but generally only do so when they contain identical sequences

    (alternative exons) at all three variable Ig domains (Wojtowicz et al. 2004,

    2007). If they differ in one or more variable Ig domains (2, 3, and 7), no

    interaction is detectable. In some rare cases heterophilic interactions have

    been detected, but in each of these instances two of the variable domains are

    identical while the non-identical third domains are highly related to each other

    (Wojtowicz et al.2007).

    Because homophilic interaction relies on sequence identity at Ig domains 2,

    3, and 7, it was proposed that these domains are major sites of interaction

    between Dscam monomers (Wojtowicz et al. 2004). Recent crystallographic

    and mutagenic studies support this hypothesis (Meijers et al.2007, Wojtowicz

    et al.2007, Sawaya et al.2008). These structural studies have determined that

    specific sequences within the variable regions of Ig domains 2, 3, and 7 partici-

    pate in binding interactions between identical Dscam splice forms. The indivi-

    dual variable domains of one monomer bind to the corresponding variable

    domain of the identical second monomer in an antiparallel configuration such

    that Ig 2 binds Ig domain 2, Ig 3 binds Ig 3, and Ig 7 binds Ig 7 (Fig. 9.3).

    Overall, the structure adopted by the first seven Ig domains of each interacting

    Dscam monomer resembles an S-shape in which Ig domains 2, 3, and 7 are

    roughly in a linear arrangement. The S-shape conformation is only observed

    9 The Down Syndrome Cell Adhesion Molecule 213

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    8/16

    when Dscam isoforms are participating in homophilic binding. When

    Dscam is monomeric, the distal Ig domains 14 exist in a horseshoe con-

    formation (the top half of the S), while Ig domains 57 are in a flexible

    extended conformation (Meijers et al.2007; Sawaya et al.2008). This horse-

    shoe conformation has been observed in other Ig superfamily members such

    as insect hemolin (Su et al. 1998) and the neural CAM axonin-1 (Freigang

    et al. 2000).

    Fig. 9.3 Dscam isoform-specific homophilic interaction. A schematic representation of theDscam receptor is shown. (AC) Ig domains are represented as rounded rectangles and

    fibronectin domains as ellipses. Ig domains 19 are numbered. A representation of a lipid

    bilayer separates the cytodomain from the ectodomain. Ig domains 2, 3, and 7 contain

    antiparallel-binding determinants represented by white and black geometric shapes. These

    binding determinants interact such thatblackdeterminants bind towhitedeterminants if and

    only if they are the same shape. (A) Identical Dscam monomers: Ig domains 14 pack into a

    horseshoe-shaped conformation while the other extracellular domains do not participate in

    higher order structures. (B)Identical Dscam isoforms engaged in trans homophilic interaction:

    Theblackdeterminants in Ig domains 2, 3, and 7 of each receptor bind to the respective white

    determinants present in the other receptor in an antiparallel manner. This results in the

    formation of an ordered S-shaped conformation involving Ig domains 17 of each mono-mer and stable receptor binding. (C)Non-identical Dscam isoforms: The two Dscam isoforms

    shown have identical Ig domains 2 and 7 but differ in Ig domain 3 as indicated by differently

    shaped binding determinants (crosses versus circles). This results in an inability to form astable interaction between these different isoforms

    214 H. Kathuria and J.C. Clemens

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    9/16

    The most well-documented biological response of Dscam homophilic inter-

    action is repulsion of neurites expressing identical (interacting) forms. The

    molecular mechanism of homophilic repulsion is unknown, but it is hypothe-

    sized to function in a stepwise manner. Initially, the identical ectodomains

    interact to create an adhesive complex. This in turn activates the Dscamcytodomain to initiate a signaling cascade that ultimately modifies the cytoske-

    leton to direct movement away from the sites of interaction (Hughes et al.2007,

    Matthews et al.2007, Soba et al.2007). Indeed, mutation of the cytodomain

    leads to a stable adhesive interaction instead of repulsion (Hughes et al.2007,

    Matthews et al.2007, Soba et al.2007). A second signal must also be generated

    to disrupt adhesion between the trans interacting ectodomains. It is thought

    that this occurs by one or more of the following mechanisms: a conformational

    shift, receptor internalization, or a proteolytic event.

    In Drosophila, Dscam-mediated homophilic repulsion has been demon-strated to control neurite branch segregation (Zhan et al. 2004) and self-

    avoidance (Hughes et al. 2007, Matthews et al. 2007, Soba et al. 2007),

    while Dscam2-mediated homophilic repulsion contributes to neuronal tiling

    (Millard et al. 2007). In mice, DSCAM-mediated homophilic repulsion is

    involved in aspects of neuronal self-avoidance and tiling (Fuerst et al.2008).

    9.6 Branch Segregation and Self-Avoidance

    Hyper-variable arthropod Dscam genes provide a molecular mechanism for

    neurons to distinguish self from non-self. In the case of theDrosophila Dscam

    gene, neurons select a nearly random population of 1450 Dscam isoforms

    from a pool of roughly 38,000 possibilities. Because of this, neighboring neu-

    rons express a unique collection of Dscam isoforms and are consequently

    unlikely to contain any isoforms in common. Therefore, the Dscam isoforms

    expressed by an individual neuron serves as a molecular signature that can be

    used to differentiate itself from all other neurons (Hattori et al.2007).

    This signature is the molecular basis of self-avoidance inDrosophilaand is

    used to help pattern the nervous system. Since only identical Dscam ectodo-

    mains interact, neighboring neurons do not repel each other because they do

    not express identical isoforms (Fig.9.4A). Therefore, individual neurons can

    bundle into nerves (fasciculate) and grow along common pathways. If neurons

    that normally fasciculate are experimentally forced to express a Dscam isoform

    in common using a transgene, the neuron bundle will defasciculate and the

    individual neurons will move away from each other (Fig. 9.4B) (Schmucker

    et al.2000, Zhan et al.2004).

    In the case of branched neurons, neurite branches display isoforms in com-

    mon since each branch originates from the same cell body. Homophilic inter-

    actions between identical Dscam isoforms on each sister branch leads to the

    initiation of repulsion and the dispersion of sister branches. This mechanism

    9 The Down Syndrome Cell Adhesion Molecule 215

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    10/16

    ensures that neurites emanating from the same cell body segregate properly to

    different targets instead of converging on a single target (Fig.9.4C) (Wang et al.

    2002, Zhan et al.2004). It also allows dendritic branches of a neuron to fully

    disperse for the uniform exploration of a spatial field (Fig.9.4D) (Zhu et al.

    2006, Hughes et al.2007, Matthews et al.2007, Soba et al.2007).

    Dscam mutant neurons display defects in segregation of sister branches to

    their proper target fields. This can be illustrated within the mushroom bodies

    (MB), which are multi-lobed structures in the Drosophila brain that are

    involved in olfactory learning and memory. Axons extend from the MB cell

    bodies down a common pathway called the peduncle. At the distal end of the

    peduncle the MB axons branch. One branch targets to the dorsal lobe, while

    the second branch targets to the medial lobe. In Dscam mutant MB axons, the

    branches form but frequently target to the same lobe (Wang et al.2002, Zhan

    et al.2004).

    A

    +1+1

    +1

    B

    T1

    T2

    C

    D

    Fig. 9.4 Dscam-mediated repulsion: self-recognition versus tiling. (AC) Neurons are repre-

    sented schematically. Axons (lines) extending from neuron cell bodies (ovals) end in growthcones, which are complex motile structures that elaborate numerous filopodia. (A) Each

    neuron expresses and displays different Dscam populations on their plasma membranes as

    indicated by different shading patterns. Since these neurons do not express common Dscam

    isoforms, they do not recognize each other as self. Their neurites do not repel each other and

    are able to fasciculate and grow along common pathways. ( B) In addition to their normal

    unique combination of Dscam isoforms expressed from their endogenous Dscamgenes (as in

    panel A), each neuron also expresses a Dscam isoform in common due to the presence of a

    transgene. Interaction between these common isoforms is interpreted as recognition of self

    and leads to neurite repulsion, which prevents them from growth along a common pathway.

    (C) In the case of axons that branch, each branch will contain the same Dscam isoforms

    allowing for self-recognition. This results in repulsion between the two growth cones and helpsensure that each branch seeks out different targets (T1 and T2) rather than both converging on

    the nearest target (T1). (D) The dendritic fields from four hypothetical neurons are shown.

    Self-recognition mediated by hyper-variable Dscam causes neurites originating from the same

    cell body to spread out and effectively cover a large spatial area. Tiling interactions (gray

    double-headed arrows) mediated by non-diverse Dscam receptors such as Dscam2 prevent

    overgrowth of the dendrites from one cell body into the area occupied by the dendrites coming

    from a different cell body

    216 H. Kathuria and J.C. Clemens

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    11/16

    Dscam mutant neurons display defects in patterning dendritic arborizations.

    Sensory neurons in theDrosophilalarva body wall elaborate complex dendritic

    arborizations that spread out to evenly cover the sensory field. The highly

    branched dendrites of these neurons, known as dendritic arborization (da)

    neurons, never cross over each other in the plane of the body wall, elegantlyillustrating the principle of self-avoidance (Fig.9.4D).Dscam mutant da neu-

    rons still elaborate dendritic fields that display complex branch patterns; how-

    ever, these branches cross over one another and fail to evenly disperse within the

    body wall (Hughes et al.2007, Matthews et al.2007, Soba et al.2007).

    Self-avoidance is a general property that likely functions in all species to

    properly disperse neurite branches. In arthropods hyper-variable Dscam genes

    have arisen to provide one mechanism to recognize and move away from self.

    SinceDrosophila Dscam is expressed in most, if not all neurons during devel-

    opment, the single Dscam gene may be sufficient to confer self-avoidanceproperties to all neurites. Outside the arthropod lineage, a similar DSCAM

    diversity has not been detected; therefore, it is unlikely that DSCAMs are the

    only molecules responsible for this process. For example, DSCAM mutant mice

    display defects in self-avoidance of neurite arbors in two sub-populations of

    retina amacrine cells that normally express DSCAM protein (Fuerst et al.

    2008). However, other retina amacrine cell populations exhibited normal neur-

    ite patterning in these mice. Therefore self-avoidance in non-arthropods may

    require the cell-type-specific expression of different repulsion receptor genes.

    Alternatively, a more general self-avoidance mechanism may have evolved inthese organisms, which is yet to be elucidated.

    9.7 Tiling

    Tiling within the nervous system is the process of completely and evenly filling a

    spatial region with multiple synaptic domains of a particular neural class.

    Similar to tiles in a floor, the synaptic domains of each neuron are contained

    within a distinct region that approaches, but does not overlap with adjacent

    domains. The result of this is a uniform distribution of neural processes that are

    restricted to each individual domain. One possible mechanism to constrain

    neural processes to a particular domain (tile) is to utilize a repulsive receptor

    system. Receptors expressed on the processes of adjacent tiling neurons could

    prevent overlap of neurite fields by inducing repulsion upon receptor interac-

    tion (Fig.9.4D).

    Hyper-variable Drosophila Dscam is not a good candidate to perform this

    role. While Dscam diversity is well suited for self-avoidance, since individual

    neurons make different isoforms, Dscam is incapable of signaling repulsion

    between processes from different neurons. Non-variable Dscam receptors, on

    the other hand, which are expressed in a cell-type-specific manner, have been

    demonstrated to function in neuron tiling. InDrosophila, Dscam2gene function

    9 The Down Syndrome Cell Adhesion Molecule 217

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    12/16

    is required for tiling the L1 subclass of lamina neurons within the visual system

    (Millard et al.2007).Dscam2mutant L1 neurons target the correct layer in the

    medulla, but extend processes laterally and invade neighboring columns (tiles).

    Presumably this is due to the lack of homophilic interaction-based repulsion

    between L1 neuron axons. A similar role for DSCAM-mediated tiling has beenuncovered in mice (Fuerst et al.2008). DSCAM is required in two sub-popula-

    tions of retinal amacrine cells for proper dispersion of these neurons in a spatial

    field. In the absence of DSCAM, these neurons migrate together and inappro-

    priately fasciculate.

    In both the preceding examples, DSCAM family members serve as homo-

    typic repulsion receptors, whose function is to preserve the spatial patterning

    (tiling) between members of the same neuron type. Since tiling is not self-

    avoidance, but rather avoidance of members within a particular class, it

    would follow that multiple tiling receptors must exist. This is because in somebiological environments, different neuron classes will tile independently of each

    other within a shared spatial region (Hughes et al.2007, Matthews et al.2007,

    Soba et al. 2007). For example, da class III dendrites exhibit tiling in the

    Drosophila body wall. Da class IV dendrites also exhibit tiling in the same

    environment. Therefore, since class III dendrites do not repel those of class

    IV, the receptors that prevent class III dendrites from overlapping must be

    different from those that prevent class IV dendrites from overlapping. It is

    known that Dscam and Dscam2 are not responsible for da class IV tiling. An

    attractive possibility is that Dscam3 or 4 might play a role in tiling theseneurons.

    9.8 Non-repulsive DSCAM Functions

    There is a growing body of evidence indicating that the DSCAM family of

    receptors can function in roles other than signaling homotypic repulsion. Loss-

    of-function studies inDrosophilasuggest that Dscam is involved in aspects of

    axon target selection in both the olfactory system (Hummel et al.2003) and

    mechanosensory neurons (Chen et al.2006). The phenotypes observed in each

    of these studies are inconsistent with merely a loss of Dscam-mediated homo-

    philic repulsion and hint at a possible instructive requirement of Dscam splice

    forms for target selection (Chen et al.2006). For example, it might be the case

    that Dscam receptors signal attraction or even adhesion in response to either

    different ligands (non-Dscam) or changes in Dscam receptor molecular

    contexts.

    Recent studies in chordates have uncovered new functions for DSCAMs.

    Yamagata and Sanes (2008) isolated chick Dscam and DscamL, orthologs of

    human DSCAM and DSCAML1, respectively, and have determined that these

    receptors function in a manner similar to sidekick receptors to pattern laminar

    arborizations in the chick retina (see Chapter 10 for a description of sidekick

    218 H. Kathuria and J.C. Clemens

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    13/16

    receptors). Like other Dscams, the ectodomains of these receptors were found

    to participate in homophilic, but not heterophilic, interactions. Instead of

    inducing repulsion between neurons, chick Dscam or DscamL homophilic

    interactions result in trans-synaptic adhesion between neurons at synaptic

    junctions within the inner plexiform layer (IPL) of the chick retina. Dscamprotein is predominantly localized to the S5 sublamina of the IPL, while

    DscamL receptors localize to sublamina S1, S2, and S4. Depletion of Dscam

    expression disrupts the laminar patterning of S5 by causing the processes of

    affected neurons to extend beyond the S5 boundary. The processes of non-

    Dscam-expressing neurons in other sublamina were not affected by Dscam

    depletion. Moreover, ectopic expression of Dscam rerouted neuronal processes

    to the Dscam-positive layer S5. Similarly, ectopic expression of DscamL

    rerouted neurites to layers other than S3, a layer that is DscamL negative.

    Taken together, these data suggest that Dscam signaling is not limited tohomotypic repulsion, but can also promote adhesion, and hint at putative

    roles for Dscams in synaptic specification or maintenance (Yamagata and

    Sanes2008).

    9.9 Non-DSCAM Interactions

    During vertebrate spinal cord development commissural axons are attracted tothe ventral midline due to expression of netrin-1 by the floor plate cells. Deleted

    in colorectal cancer (DCC) is a netrin-1 cell surface receptor expressed on

    commissural axons that function as a key mediator of the attractive and out-

    growth promoting properties of netrin-1 (Keino-Masu et al. 1996). While

    impairment of DCC function blocks netrin-1-stimulated outgrowth of commis-

    sural axon explants, it does not completely block turning of these axons toward

    a netrin-1 source (Keino-Masu et al. 1996). These results suggest that an

    additional netrin receptor functions with DCC to induce axon turning toward

    the netrin-1 gradient.

    In addition to homophilic interactions, the extracellular domain of rat

    DSCAM has been shown to engage in heterophilic interactions with netrin-1

    (Ly et al.2008). A protein truncation analysis revealed that the netrin-1-binding

    site is located within a region containing DSCAM Ig domains 79. Addition-

    ally, DSCAM and DCC form a receptor complex in the absence of netrin-1.

    This complex requires the transmembrane domain of DSCAM and it dissoci-

    ates upon netrin-1 stimulation (Ly et al. 2008). Like knockdown of DCC,

    knockdown of DSCAM by siRNA does not completely block the turning

    response of rat commissural axons to netrin-1. However, simultaneous knock-

    down of both DSCAM and DCC results in a complete blockage of netrin-1-

    induced axon turning (Ly et al.2008). Finally, ectopic expression of DSCAM in

    Xenopusspinal neurons is sufficient to confer a turning response to netrin-1 in

    the absence of DCC activity (Ly et al. 2008). These studies demonstrate that

    9 The Down Syndrome Cell Adhesion Molecule 219

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    14/16

    vertebrate DSCAM has additional heterophilic binding partners, which enable

    it to function as a netrin-1 receptor and mediate axon turning, much in the same

    way as DCC functions. A recent study in Drosophila has generated similar

    findings and suggests that netrin binding might be general property of all

    DSCAMs (Andrews et al. ).

    9.10 Concluding Remarks

    It has been a decade since the discovery of the founding DSCAM family

    member. DSCAM genes have been identified in virtually all organisms that

    have a nervous system. During this time our understanding of DSCAM bio-

    chemical properties and biological functions has grown, but is far from com-

    plete. Thus far, all DSCAMs have been reported to engage in isoform-specifichomophilic interactions. The biological consequences of these interactions

    appear to vary between homotypic repulsion and adhesion. This may simply

    reflect different species-specific roles that have evolved for DSCAMs since what

    we know about DSCAM function is being pieced together from experiments in

    a wide variety of organisms. An alternative and attractive hypothesis is that the

    signaling output from homotypic interactions is dependent on the signaling

    contexts in which these interactions occur. For example, it might be the case

    that axonaxon- or dendritedendrite-based DSCAM homotypic interactions

    lead to repulsion to segregate neurite branches, disperse arborizations, and tilereceptive fields. On the other hand, axondendrite-based DSCAM homotypic

    interactions may be instructive for the selection and stabilization of synaptic

    partners as in the chicken IPL. It may even be the case that the hyper-variable

    arthropod Dscams play an instructive role in connection specificity as well,

    rather than simply serving as a means to distinguish self from non-self. Recent

    studies have added a new wrinkle to the DSCAM story with the discovery that

    DSCAMs also functions as netrin receptors. Finally the involvement of

    DSCAM in human Down syndrome and other nervous system structural dis-

    orders awaits critical assessment.

    References

    Agarwala KL, Nakamura S, Tsutsumi Y et al. (2000) Down syndrome cell adhesion molecule

    DSCAM mediates homophilic intercellular adhesion. Brain Res Mol Brain Res

    79:118126

    Andrews GL, Tanglao S, Farmer WT, (2008) Dscam guides embryonic axons by Netrin-

    dependent and -independent functions. Development 135:38393848

    Barlow GM, Micales B, Chen XN et al. (2002) Mammalian DSCAMs: roles in the develop-ment of the spinal cord, cortex, and cerebellum? Biochem Biophys Res Commun

    293:881891Brites D, McTaggart S, Morris K et al. (2008) The Dscam homologue of the crustacean

    Daphnia is diversified by alternative splicing like in insects. Mol Biol Evol 25:14291439

    220 H. Kathuria and J.C. Clemens

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    15/16

    Celotto AM and Graveley BR (2001) Alternative splicing of the Drosophila Dscam pre-

    mRNA is both temporally and spatially regulated. Genetics 159:599608

    Chen BE, Kondo M, Garnier A et al. (2006) The molecular diversity of Dscam is functionally

    required for neuronal wiring specificity in Drosophila. Cell 125:607620Clemens JC, Ursuliak Z, Clemens KK et al. (1996) A Drosophila protein-tyrosine phospha-

    tase associates with an adapter protein required for axonal guidance. J Biol Chem

    271:1700217005Delabar JM, Theophile D, Rahmani Z et al. (1993) Molecular mapping of twenty-four

    features of Down syndrome on chromosome 21. Eur J Hum Genet 1:114124

    Desai CJ, Garrity PA, Keshishian H et al. (1999) The Drosophila SH2SH3 adapter protein

    Dock is expressed in embryonic axons and facilitates synapse formation by the RP3

    motoneuron. Development 126:15271535

    Dong Y, Taylor HE and Dimopoulos G (2006) AgDscam, a hypervariable immunoglobulin

    domain-containing receptor of the Anopheles gambiae innate immune system. PLoS Biol

    4:e229

    Freigang J, Proba K, Leder L et al. (2000) The crystal structure of the ligand binding module of

    axonin-1/TAG-1 suggests a zipper mechanism for neural cell adhesion. Cell 101:425433

    Fuerst PG, Koizumi A, Masland RH et al. (2008) Neurite arborization and mosaic spacing in

    the mouse retina require DSCAM. Nature 451:470474

    Funada M, Hara H, Sasagawa H et al. (2007) A honey bee Dscam family member, AbsCAM,

    is a brain-specific cell adhesion molecule with the neurite outgrowth activity which

    influences neuronal wiring during development. Eur J Neurosci 25:168180

    Garrity PA, Rao Y, Salecker I et al. (1996) Drosophila photoreceptor axon guidance and

    targeting requires the dreadlocks SH2/SH3 adapter protein. Cell 85:639650

    Graveley BR, Kaur A, Gunning D et al. (2004) The organization and evolution of the dipteran

    and hymenopteran Down syndrome cell adhesion molecule (Dscam) genes. RNA

    10:14991506Hattori D, Demir E, Kim HW et al. (2007) Dscam diversity is essential for neuronal wiring

    and self-recognition. Nature 449:223227Hing H, Xiao J, Harden N et al. (1999) Pak functions downstream of Dock to regulate

    photoreceptor axon guidance in Drosophila. Cell 97:853863

    Hughes ME, Bortnick R, Tsubouchi A et al. (2007) Homophilic Dscam interactions control

    complex dendrite morphogenesis. Neuron 54:417427

    Hummel T, Vasconcelos ML, Clemens JC et al. (2003) Axonal targeting of olfactory receptor

    neurons in Drosophila is controlled by Dscam. Neuron 37:221231

    Keino-Masu K, Masu M, Hinck L et al. (1996) Deleted in Colorectal Cancer (DCC) encodes a

    netrin receptor. Cell 87:175185

    Korenberg JR, Bradley C and Disteche CM (1992) Down syndrome: molecular mapping ofthe congenital heart disease and duodenal stenosis. Am J Hum Genet 50:294302

    Korenberg JR, Chen XN, Schipper R et al. (1994) Down syndrome phenotypes: the con-

    sequences of chromosomal imbalance. Proc Natl Acad Sci U S A 91:49975001

    Li W and Guan KL (2004) The Down syndrome cell adhesion molecule (DSCAM) interacts

    with and activates Pak. J Biol Chem 279:3282432831

    Ly A, Nikolaev A, Suresh G et al. (2008) DSCAM is a netrin receptor that collaborates with

    DCC in mediating turning responses to netrin-1. Cell 133:12411254

    Matthews BJ, Kim ME, Flanagan JJ et al. (2007) Dendrite self-avoidance is controlled by

    Dscam. Cell 129:593604

    Meijers R, Pu ttmann-Holgado R, Skiniotis G et al. (2007) Structural basis of Dscam isoform

    specificity. Nature 449:487491Millard SS, Flanagan JJ, Pappu KS et al. (2007) Dscam2 mediates axonal tiling in the

    Drosophila visual system. Nature 447:720724

    Neves G, Zucker J, Daly M et al. (2004) Stochastic yet biased expression of multiple Dscam

    splice variants by individual cells. Nat Genet 36:240246

    9 The Down Syndrome Cell Adhesion Molecule 221

  • 8/13/2019 Chapter 9 the Down Syndrome Cell Adhesion Molecule

    16/16

    Ponting CP, Phillips C, Davies KE et al. (1997) PDZ domains: targeting signalling molecules

    to sub-membranous sites. Bioessays 19:469479

    Ronan, A, Fagan, K, Christie, L (2007) Familial 4.3 Mb duplication of 21q22 sheds new light

    on the Down syndrome critical region. J Med Genet 44:448451

    Sawaya MR, Wojtowicz WM, Andre I et al. (2008) A double S shape provides the structural

    basis for the extraordinary binding specificity of Dscam isoforms. Cell 134:10071018Schmucker D (2007) Molecular diversity of Dscam: recognition of molecular identity in

    neuronal wiring. Nat Rev Neurosci 8:915920Schmucker D, Clemens JC, Shu H et al. (2000) Drosophila Dscam is an axon guidance

    receptor exhibiting extraordinary molecular diversity. Cell 101:671684

    Soba P, Zhu S, Emoto K et al. (2007) Drosophila sensory neurons require Dscam for dendritic

    self-avoidance and proper dendritic field organization. Neuron 54:403416Su XD, Gastinel LN, Vaughn DE et al. (1998) Crystal structure of hemolin: a horseshoe shape

    with implications for homophilic adhesion. Science (New York, NY) 281:991995

    Wang J, Zugates CT, Liang IH et al. (2002) Drosophila Dscam is required for divergent

    segregation of sister branches and suppresses ectopic bifurcation of axons. Neuron

    33:559571Watson FL, Pu ttmann-Holgado R, Thomas F et al. (2005) Extensive diversity of Ig-super-

    family proteins in the immune system of insects. Science (New York, NY) 309:18741878

    Wojtowicz WM, Flanagan JJ, Millard SS et al. (2004) Alternative splicing of Drosophila

    Dscam generates axon guidance receptors that exhibit isoform-specific homophilic bind-

    ing. Cell 118:619633

    Wojtowicz WM, Wu W, Andre I et al. (2007) A vast repertoire of Dscam binding specificitiesarises from modular interactions of variable Ig domains. Cell 130:11341145

    Yamagata M and Sanes JR (2008) Dscam and Sidekick proteins direct lamina-specific

    synaptic connections in vertebrate retina. Nature 451:465469

    Yamakawa K, Huot YK, Haendelt MA et al. (1998) DSCAM: a novel member of the

    immunoglobulin superfamily maps in a Down syndrome region and is involved in thedevelopment of the nervous system. Hum Mol Genet 7:227237

    Zhan XL, Clemens JC, Neves G et al. (2004) Analysis of Dscam diversity in regulating axon

    guidance in Drosophila mushroom bodies. Neuron 43:673686

    Zhu H, Hummel T, Clemens JC et al. (2006) Dendritic patterning by Dscam and synaptic

    partner matching in the Drosophila antennal lobe. Nat Neurosci 9:349355

    Zipursky SL, Wojtowicz WM and Hattori D (2006) Got diversity? Wiring the fly brain with

    Dscam. Trends Biochem Sci 31:581588

    222 H. Kathuria and J.C. Clemens