Cell Signalling and Gene Expression Mediated by RET

  • Upload
    ifti007

  • View
    215

  • Download
    0

Embed Size (px)

Citation preview

  • 7/29/2019 Cell Signalling and Gene Expression Mediated by RET

    1/7

    MINISYMPOSIUM

    Cell signalling and gene expression mediated by RET

    tyrosine kinase

    K . K U R O K A W A1 , K . K A W A I1 , M . H A S H I M O T O1 , Y . I T O 2 & M . T A K A H A S H I1

    From the 1Department of Pathology, and the 2Equipment Center for Research and Education, Nagoya University Graduate School of Medicine,

    Showa-ku, Nagoya, Japan

    Abstract. Kurokawa K, Kawai K, Hashimoto M, ItoY, Takahashi M (Nagoya University Graduate

    School of Medicine, Showa-ku, Nagoya, Japan).

    Cell signalling and gene expression mediated by

    RET tyrosine kinase (Minisymposium). J Intern Med

    2003; 253: 627633.

    Germline mutations of the RETproto-oncogene cause

    multiple endocrine neoplasia (MEN) 2A or 2B by

    different mechanisms. As is the case for other receptor

    tyrosine kinases, mutant RET recruits a variety of

    signalling molecules via phosphorylated tyrosine

    residues present in the kinase domain and carboxy-

    terminal tail. As we previously reported, the signalingvia phosphorylated tyrosine 1062 plays a crucial role

    in the transforming activities of both RET-MEN2A

    and RET-MEN2B mutant protein. Interestingly, this

    single tyrosine residue represents a binding site for

    several signalling molecules including SHC, Enigma,

    SNT/FRS2, DOK and IRS1 and is responsible for ac-

    tivation of the RAS/ERK, PI3-K/AKT, JNK,

    p38MAPK and ERK5 signalling pathways. Amongstthese, the PI3-K/AKT and JNK pathways appeared to

    be more strongly activated in the cells expressing

    RET-MEN2B than in the cells expressing RET-

    MEN2A, suggesting the possibility that these path-

    ways may be involved in the disease phenotype. In

    addition, RET is alternatively spliced to produce three

    isoforms and the splicing site is present just down-

    stream of tyrosine 1062. These isoforms play different

    roles for the tumour development associated with

    MEN 2 or the development of the kidney and the

    enteric nervous system. Moreover, using differential

    display analysis, we identified several genes whoseexpression is highly induced by RET-MEN2B mutant

    proteins. The differential gene expression by RET-

    MEN2A and RET-MEN2B may also be important for

    the development of their phenotypes.

    Keywords: DOK1, multiple endocrine neoplasia type

    2, RET isoforms, SHC, SNT/FRS2, stanniocalcin 1.

    Introduction

    The RETproto-oncogene encodes a receptor tyrosine

    kinase [1, 2] that is essential for the development of

    the enteric nervous system and the kidney [3]. The

    glial cell line-derived neurotrophic factor (GDNF)

    family ligands (GFLs) including GDNF, neurturin,

    persephin and artemin exert their physiological

    functions via activation of RET tyrosine kinase.

    However, GFLs do not bind to the extracellulardomain of RET directly but require glycosylphos-

    phatidylinositol-linked cell surface proteins called

    GFRas for complex formation with RET. GFRa family

    members (GFRa14) show preferential binding to a

    particular GFL and plays specific roles in vivo

    through the preferred ligandreceptor complex

    formation [4]. Gene ablation studies revealed that

    the GDNF/GFRa1/RET complex formation is crucial

    Journal of Internal Medicine 2003; 253: 627633

    2003 Blackwell Publishing Ltd 627

  • 7/29/2019 Cell Signalling and Gene Expression Mediated by RET

    2/7

    for the development of the kidney and the enteric

    nervous system [5].

    RET mutations are responsible for the develop-

    ment of several human diseases including papillary

    thyroid carcinoma, multiple endocrine neoplasia

    (MEN) 2A and 2B, familial medullary thyroid carci-noma (FMTC) and Hirschsprungs disease (HSCR)

    [610]. More than 10 rearranged forms ofREThave

    been cloned from sporadic and radiation-associated

    papillary thyroid carcinomas [11, 12], whereas

    germline point mutations of RET were identified in

    MEN2A, MEN2B and FMTC. MEN2A or FMTC

    mutations were mostly found in one of six cysteine

    residues in the RET extracellular cysteine-rich do-

    main [13]. These cysteine mutations induce disul-

    phide-linked RET dimerization, leading to its ligand-

    independent activation [14, 15]. On the contrary,

    the MEN2B mutations were detected at codon 918(methionine to threonine) in most cases or at codon

    883 (alanine to phenylalanine) in fewer than 4%

    cases [13], which are present in the catalytic core of

    the RET tyrosine kinase domain. These mutations

    probably induce conformational change of the kinase

    domain, resulting in activation without dimerization

    [1517]. However, we do not fully understand the

    mechanisms by which the different phenotypes

    between MEN2A and MEN2B are caused.

    Hirschsprungs disease is a congenital malforma-

    tion associated with aganglionosis of the gastroin-

    testinal tract. RET mutations are responsible forabout 50% of familial and 1020% of sporadic cases

    of HSCR and a variety of missense, non-sense and

    frame shift mutations have been identified along

    with its whole coding sequence [1822]. Based on

    functional analyses, at least four mechanisms of RET

    dysfunction are responsible for the development of

    HSCR. First, most mutations identified in the extra-

    cellular domain markedly impaired the RET cell

    surface expression, probably because of incorrect

    folding of the RET protein [2325]. Secondly, a few

    mutations in the carboxy-terminal tail impaired the

    binding of adaptor proteins such as SHC to RET,resulting in the defect of RET-mediated RAS/ERK

    and PI3-K/AKT signalling [26, 27]. Thirdly, the

    mutations in the kinase domain, which were iden-

    tified in amino acids conserved amongst members of

    the tyrosine kinase family, almost completely abol-

    ished the RET tyrosine kinase activity [2831].

    Fourthly, some mutations in the kinase domain

    severely impaired the activation of PLCc pathway

    [31]. Taken together, these findings suggested that

    several intracellular signalling pathways via RET

    cooperatively function for the normal development

    of the enteric nervous system.

    Cell signalling mediated by RET

    RET is alternatively spliced to produce three isoforms

    (designated RET9, RET43 and RET51) that differ in

    the carboxy-terminal sequence [32, 33]. There are

    16 tyrosines in the intracellular domain of RET9 and

    RET43 and 18 tyrosines in that of RET51. Amongst

    these, tyrosines 1015 and 1062 (Y1015 and

    Y1062) have been shown to be important for cell

    signalling mediated by both ligand-dependent and

    -independent activation of RET [12, 34]. Y1015

    represents a binding site for PLCc [35], and Y1062

    represents a binding site for several adaptor proteinssuch as SHC [3638], SNT/FRS2 [39, 40], IRS1

    [41], DOK1 [42], DOK4 and 5 [43], and Enigma

    [44] (Fig. 1). As a result, various signalling path-

    ways including the RAS/ERK, PI-3K/AKT,

    p38MAPK, JNK [45] and ERK5 pathways [46] are

    activated mainly via phosphorylated Y1062 in RET.

    Consistent with these findings, mutation of Y1062

    markedly impaired the transforming activity of all

    types of MEN2 mutant proteins [36].

    When SHC binds to Y1062 following RET activa-

    tion, it further complexes with GRB2 and GAB1/

    GAB2 proteins, leading to activation of the PI3-K/AKT signalling pathway. On the contrary, asso-

    ciation of SHC with the GRB2/SOS complex is

    responsible for activation of the RAS/ERK pathway

    [45, 47] (Fig. 1). Activation of the PI3-K/AKT and

    RAS/ERK pathways resulted in the activation of

    transcription factors, NFjB and CREB, respectively

    [45]. Moreover, binding of SNT/FRS2 to Y1062

    leads to the RAS/ERK activation because it has

    GRB2 and SHP-2 binding sites in the carboxy-

    teminal region [39, 40].

    Using a yeast two-hybrid screen, we found that

    DOK1 also binds to tyrosine 1062 in RET [42, 48].DOK1 was more strongly associated with

    RET-MEN2B mutant proteins than RET-MEN2A

    mutant proteins. As a result, DOK1 was highly

    phosphorylated in the cells expressing RET-MEN2B.

    DOK1 has six tyrosine residues that represent

    binding sites for RAS-GTPase activating protein

    (RAS-GAP), leading to suppression of the RAS/ERK

    activation. In addition, DOK1 contains a tyrosine

    2003 Blackwell Publishing Ltd Journal of Internal Medicine 253: 627633

    6 28 K . K UR OK AW A et al.

  • 7/29/2019 Cell Signalling and Gene Expression Mediated by RET

    3/7

    residue that is a binding site for the NCK adaptor

    protein. The association of NCK with DOK1appeared to be responsible for JNK and c-JUN

    activation (Fig. 1). Salvatore et al. [49] reported

    that the level of phosphorylation of Y1062 is

    increased in PC12 cells expressing RET-MEN2B,

    resulting in enhancement of activation of the ERK

    and PI3-K/AKT pathways. Taken together with our

    findings indicating high levels of activation of JNK

    and AKT by RET-MEN2B [42, 48], it is suggested

    that the enhanced signalling via Y1062 may be

    involved in the MEN2B phenotype.

    DOK4 and 5 also bind to Y1062 of RET and

    induce ligand-dependent axonal outgrowth ofPC12 cells [43]. DOK4 is expressed broadly in

    many tissues, such as brain, heart, lung and

    kidney, whereas DOK5 is specifically expressed in

    the brain. Because DOK4 and DOK5 associate with

    neither RAS-GAP nor NCK, it is interesting to

    investigate which signalling pathways are activa-

    ted by them.

    Regulation of cell signalling via Y1062in RET

    Both SHC and SNT/FRS2 bind to pY1062 of RET viatheir phosphotyrosine-binding (PTB) domains, that

    recognize the common consensus motif NKLpY.

    Both PTB domains have conformational topology

    but do not have sequence homology [50]. So far,

    two types of the PTB domains are known. The PTB

    domain of SNT/FRS2 has sequence similarity to the

    PTB domain of IRS1, so these PTB domains are

    called phosphotyrosine-binding domain, insulin

    receptor substrate 1-like (PTBI). DOK-family pro-

    teins also contain PTBI.The PTB domain of SHC binds to TrkA, that has

    the canonical consensus motif for its binding

    (NPXpY, X is any amino acid) in the juxtamembrane

    region. The PTBI domain of SNT/FRS2 recognizes

    the same consensus motif in TrkA as the PTB

    domain of SHC. Consistent with this finding, the

    binding mode of SNT/FRS2 PTBI to RET in vitro

    resembles that of SHC PTB, depending on phos-

    phorylation of Y1062 [39]. Although the PTBI

    domain of SNT/FRS2 also binds to fibroblast growth

    factor receptor (FGFR) [5154], the consensus

    sequence for its binding in FGFR represents a uniquepeptide that does not contain phosphotyrosine. In

    addition, the binding is ligand-independent. The

    finding that SNT/FRS2 recognizes different receptors

    with different modes suggests the interference of

    signalling pathways activated by each receptor [52].

    Recently, it was reported that the microdomains of

    membrane, lipid rafts, play a role for regulating or

    separating cell signalling via SHC and SNT/FRS2

    from Y1062 of RET [55, 56]. As GFRas are GPI-

    anchored at the carboxy-terminus and SNT/FRS2

    has a myristylation sequence at the amino-terminus

    [57], these proteins locate on rafts. RET receptortyrosine kinase locates outside rafts under unstimu-

    lated conditions, and appears to be recruited to rafts

    by GPI-anchored coreceptor GFRas. As a result, after

    stimulation by GDNF, association of RET with SNT/

    FRS2 is supposed to lead to sustained ERK activation.

    On the contrary, SHC locates outside rafts, and may

    be involved in the transient ERK activation as well as

    the activation of the PI3-K/AKT pathway [56].

    Y1062Y1015

    PLC

    Signal sequence Cysteine-rich

    Transmembrane

    Kinase domain

    SHC

    GAB1/2

    GRB2

    PI3-K

    RET51

    RET9RET43

    AKT

    SOS

    SNT/FRS2 DOK1

    p38MAPK

    ERK5

    SHP-2

    NCK

    JNKRAS

    ERK

    c-JUN

    RAS-GAP

    NFB CREB

    GRB2++

    Fig. 1 Signaling pathways medi-

    ated by RET receptor tyrosine

    kinase.

    2003 Blackwell Publishing Ltd Journal of Internal Medicine 253: 627633

    M I N I SY M P O SI U M : C E L L S I G N AL L I N G B Y R E T 6 2 9

  • 7/29/2019 Cell Signalling and Gene Expression Mediated by RET

    4/7

    The splicing site of three RET isoforms is present

    just downstream of Y1062, and their different roles

    are recently demonstrated. By NIH 3T3 transfection

    assay, we showed that RET51-MEN2A and RET51-

    MEN2B mutant proteins have stronger transforming

    activity than RET9-MEN2A and RET9-MEN2Bmutant proteins, respectively [36]. In addition, the

    activity of RET43 was very low [26]. These findings

    suggested that the sequences downstream of Y1062

    are important for binding of adaptor proteins and

    that RET51 may contribute more significantly to the

    tumour development associated with MEN 2 than

    RET9 and RET43. On the contrary, RET9 was

    shown to be critical for the development of the

    kidney and the enteric nervous system using

    targeted mutagenesis mice expressing RET9 or

    RET51 only [58], suggesting the possibility that

    RET9 and RET51 are involved in the activation of

    different signalling pathways in vivo [59].

    Computational analysis of SHC PTB domain

    binding to each RET isoform

    To obtain supportive information about the function

    of RET isoforms based on structural biology, we

    simulatedthe interaction of the SHCPTB domainwith

    the amino acids around Y1062 of each RET isoform

    (Fig. 2). The simulations were performed based on the

    model of the SHC PTB domain complexed with the

    tyrosine-phosphorylated peptide of TrkA [60].

    As shown in Fig. 2a, a hydrogen bond is formed

    between arginine (R) 67 in the SHC PTB domain

    RET51

    M1064

    2.37NN

    H

    R207 M1064

    2.37 NN

    H

    R207

    RET peptide with tyrosine 1062

    SHC PTB domain

    (b)

    (a)

    2.62

    R67

    pY1062

    Y1062F

    N

    H

    RET9

    R207R1064

    pY1062

    2.47

    NH

    R207R1064

    2.47

    NH

    RET43

    A1064R207

    A1064R207

    Fig. 2 Computational analyses of SHC phosphotyrosine-binding (PTB) domain binding to RET peptide with tyrosine 1062. Simulations

    of the interaction between SHC PTB domain and RET peptide with tyrosine 1062 were performed based on the model of the SHC PTB

    domain complexed with the tyrosine-phosphorylated peptide of TrkA [60]. The distances between the functional residues and the potential

    energy of compounds were calculated using the insight II software package. The compounds were simulated with force field parameters

    based on the consistent valence force field (CVFF). The group-based cut off, 0.95 nm for the van der Waals and 0.95 nm for Coulomb

    interactions, were introduced. The temperature was set at 298K. Calculations based on the simulation images were carried out using

    the insight II package. (a) A hydrogen bond between phosphotyrosine (pY) 1062 in RET9 and arginine (R) 67 in SHC (2.62 A ). When

    Y1062 is replaced with phenylalanine (Y1062F, light blue), the hydrogen bond cannot be formed. (b) The interaction between SHC

    PTB domain and three isoforms of RET. Arginine (R) 1064 in RET9 or methionine (M) 1064 in RET51 forms a hydrogen bond with

    arginine (R) 207 in the SHC PTB domain (2.47 A and 2.37 A respectively), but RET 43 does not.

    2003 Blackwell Publishing Ltd Journal of Internal Medicine 253: 627633

    6 30 K . K UR OK AW A et al.

  • 7/29/2019 Cell Signalling and Gene Expression Mediated by RET

    5/7

    and phosphotyrosine (pY) 1062 in RET (2.62A).

    When Y1062 is replaced by phenylalanine

    (Y1062F), the hydrogen bond cannot be formed.

    In addition, arginine (R) 1064 in RET9 or methion-

    ine (M) 1064 in RET51 forms a hydrogen bond with

    arginine (R) 207 in the SHC PTB domain (2.47 A

    and 2.37 A respectively), but RET 43 does not (Fig.

    2b). These findings are consistent with our previous

    data using in vitro binding assay [26, 39]. The

    current simulations suggested that the conformation

    of the SHC PTB domain binding to RET9 is almost

    the same as that to TrkA [60]. However, the actual

    conformation of the SHC PTB domain binding to

    RET51 might be changed to some degree, because

    the distance between methionine (M) 1064 in

    RET and arginine (R) 207 in SHC appears to be

    too close to form stable interactions between these

    molecules.Amongst three RET isoforms, only RET9 has the

    consensus sequence for binding of the SHC SH2

    domain [38, 61]. This may contribute to the stable

    binding of SHC to RET9 in addition to the stable

    mode of binding of the SHC PTB domain to RET9.

    Gene expression and disease phenotype

    We performed a differential display analysis of gene

    expression using NIH3T3 cells expressing the

    RET-MEN2A or RET-MEN2B mutant proteins [62].

    As a result, we identified 130 known genes and 13previously unidentified sequences whose expression

    was affected by each mutant protein. Of 130 known

    genes, 29 genes were confirmed to be expressed

    differentially by Northern blotting (Table 1). Based

    on their expression patterns, they were classified

    into four types: (i) 10 genes induced by both RET-

    MEN2A and RET-MEN2B mutant proteins (type I),

    (ii) six genes induced predominantly by RET-MEN2A

    (type II), (iii) five genes induced predominantly by

    RET-MEN2B (type III), (iv) eight genes repressed by

    RET-MEN2A and RET-MEN2B (type IV). Type I

    includes cyclin D1, cofilin, and cathepsin L and Bgenes that are known to be involved in cell growth,

    tumour progression and invasion. In contrast, type

    IV includes type I collagen, lysyl oxidase, annexin I,

    and tissue inhibitor of matrix metalloproteinase 3

    (TIMP3) genes that have been implicated in tumour

    suppression. Type II and type III include various

    genes with different physiological functions, and it is

    unknown whether these gene expressions are really

    involved in the tumour development or diseasephenotype.

    To see the physiological significance of inducible

    genes, we investigated whether these inducible

    genes are also induced by GDNF stimulation.

    Amongst 21 genes induced by RET-MEN2A and/or

    RET-MEN2B, six genes including cyclin D1, cathep-

    sin B, cofilin, ring finger protein 11 (RNF11),

    integrin-a6, and stanniocalcin 1 (STC1) genes were

    also induced in TGW human neuroblastoma cells in

    response to GDNF stimulation, although the time

    course of the induction was different depending on

    the genes [62]. The STC1 gene was highly inducedby both MEN2B mutant protein and GDNF stimu-

    lation. Because STC1 was suggestive of a role in

    early skeletal development that is affected in MEN2B

    patients, we stained paraffin sections of human

    medullary thyroid carcinoma, arising from MEN2A,

    MEN2B and sporadic cases. Interestingly, MEN2B

    MTC was strongly stained with the STC1 antibody,

    whereas MEN2A MTC was weakly stained and

    Table 1 Gene expression induced or suppressed by RET-MEN

    2A/2B mutant proteins

    Type I Type II Type III Type IV

    Cyclin D1 EMK2 STC1 Annexin I

    RNF11 ITGA6 PheX Annexin IV

    Cofilin PKA-RI EIF4G3 COLIA1Foocen TACC3 Neuropsin COLIA2

    Cathepsin L PRNPA PLOD2 Lysyl oxidase

    Cathepsin B MPT TIMP3

    Decorin Pleiotrophin

    INFb SDF1a

    TB-2 like 1 protein

    HSC73

    Type I, the genes induced by both RET-MEN2A and MEN2B

    mutant proteins.

    Type II, the genes induced predominantly by RET-MEN2A mutant

    protein.

    Type III, the genes induced predominantly by RET-MEN2B

    mutant protein.

    Type IV, the genes repressed by both RET-MEN2A and MEN2Bmutant proteins.

    RNF11, ring finger protein 11; INFb, interferon b; HSC73, heat

    shock protein 73; EMK2, ELKL motif kinase 2; ITGA6, integrin-a6;

    PKA-RI, protein kinase A regulatory subunit I; TACC3, trans-

    forming acidic coiled coil-containing gene family 3; PRNPA, pri-

    on-related protein A; MPT, mitochondrial phosphate transporter;

    STC1, stanniocalcin 1; PheX, phosphate-regulating gene with

    homology to endopeptidases on the X chromosome; EIF4G3,

    eukaryotic translation initiation factor 4G3; PLOD2, procollagen-

    lysine, 2-oxyglutarate, 5-dioxygenase 2; COLIA1, type I collagen

    a1 chain; COLIA2, type I collagen a2 chain; TIMP3, tissue

    inhibitor of metalloproteinases 3; SDF1a, stromal cell-derived

    factor-1a.

    2003 Blackwell Publishing Ltd Journal of Internal Medicine 253: 627633

    M I N I SY M P O SI U M : C E L L S I G N AL L I N G B Y R E T 6 3 1

  • 7/29/2019 Cell Signalling and Gene Expression Mediated by RET

    6/7

    sporadic MTC without RET mutation was almost

    unstained [62], suggesting a possible role for STC1

    in the development of MEN2B phenotype.

    Conflict of interest statement

    No conflict of interest was declared.

    Acknowledgements

    We would like to thank members of our laboratory

    for helpful scientific advice. We are grateful to

    K. Imaizumi and M. Kozuka for their technical

    assistance. This work was supported in part by a

    grant-in-aid for COE (Center of Excellence) research

    from the Ministry of Education, Culture, Science,

    Sports and Technology of Japan.

    References

    1. Takahashi M, Buma Y, Iwamoto T, Inaguma Y, Ikeda H, Hiai

    H. Cloning and expression of the ret proto-oncogene enco-

    ding a tyrosine kinase with two potential transmembrane

    domains. Oncogene 1988; 3: 5718.

    2. Takahashi M, Buma Y, Hiai H. Isolation of ret proto-

    oncogene cDNA with an amino-terminal signal sequence.

    Oncogene 1989; 4: 8056.

    3. Schuchardt A, DAgati V, Larsson-Blomberg L, Costantini F,

    Pachnis V. Defects in the kidney and enteric nervous system

    of mice lacking the tyrosine kinase receptor Ret. Nature

    1994; 367: 3803.

    4. Airaksinen MS, Titievsky A, Saarma M. GDNF family neu-

    rotrophic factor signalling: four masters, one servant? Mol

    Cell Neurosci 1999; 13: 3125.

    5. Airaksinen MS, Saarma M. The GDNF family: signalling,

    biological functions and therapeutic value. Nat Rev Neurosci

    2002; 3: 3894.

    6. Grieco M, Santoro M, Berlingieri MT et al. PTC is a novel

    rearranged form of the ret proto-oncogene and is frequently

    detected in vivo in human thyroid papillary carcinomas. Cell

    1990; 60: 5563.

    7. Mulligan LM, Kwok JBJ, Healey CS et al. Germ-line mutations

    of the RET proto-oncogene in multiple endocrine neoplasia

    type 2A. Nature 1993; 363: 45860.

    8. Donis-Keller H, Dou SS, Chi D et al. Mutations in the RET

    proto-oncogene are associated with MEN 2A and FMTC.

    Hum Mol Genet 1993; 2: 856.

    9. Hofstra RMW, Landsvater RM, Ceccherini I et al. A mutation

    in the RET proto-oncogene associated with multiple endo-

    crine neoplasia type 2B and sporadic medullary thyroid

    carcinoma. Nature 1994; 367: 376.

    10. Carlson KM, Dou SS, Chi D et al. Single missense mutation in

    the tyrosine kinase catalytic domain of the RET protoonco-

    gene is associated with multiple endocrine neoplasia type 2B.

    Proc Natl Acad Sci USA 1994; 91: 157983.

    11. Jhiang SM. The RET proto-oncogene in human cancers.

    Oncogene 2000; 19: 5597.

    12. Takahashi M. The GDNF/RET signalling pathway and hu-

    man diseases Cytokine Growth Factor Rev 2001; 12: 3673.

    13. Eng C. RET proto-oncogene in the development of human

    cancer. J Clin Oncol 1999; 17: 38093.

    14. Asai N, Iwashita T, Matsuyama M, Takahashi M. Mechanism

    of activation of the ret proto-oncogene by multiple

    endocrine neoplasia 2A mutations. Mol Cell Biol 1995; 15:

    1619.

    15. Santoro M, Carlomagno F, Romano A et al. Activation of RET

    as a dominant transforming gene by germline mutations of

    MEN2A and MEN2B. Science 1995; 267: 383.

    16. Borrello MG, Smith DP, Pasini B et al. RET activation by

    germline MEN2A and MEN2B mutations. Oncogene 1995;

    11: 24127.

    17. Iwashita T, Asai N, Murakami H, Matsuyama M, Takahashi

    M. Identification of tyrosine residues that are essential for

    transforming activity of the ret proto-oncogene with MEN2A

    or MEN2B mutation. Oncogene 1996; 12: 487.

    18. Romeo G, Ronchetto P, Luo Y et al. Point mutations affecting

    the tyrosine kinase domain of the RET proto-oncogene in

    Hirschprungs disease. Nature 1994; 367: 378.

    19. Edery P, Lyonnet S, Mulligan LM et al. Mutations of the RET

    protooncogene in Hirschsprungs disease. Nature 1994; 367:

    3780.

    20. Angrist M, Bolk S, Thiel B et al. Mutation analysis of the RET

    receptor tyrosine kinase in Hirschsprung disease. Hum Mol

    Genet 1995; 4: 8230.

    21. Attie T, Pelet A, Edery P et al. Diversity of RET proto-onco-

    gene mutations in familial and sporadic Hirschsprung

    disease. Hum Mol Genet 1995; 4: 1386.

    22. Chakravarti A. Endothelin receptor-mediated signalling in

    Hirschsprung disease. Hum Mol Genet 1996; 5: 307.

    23. Carlomagno F, De Vita G, Berlingieri MT et al. Molecular

    heterogeneity of RET loss of function in Hirschsprungs

    disease. EMBO J 1996; 15: 27125.

    24. Iwashita T, Murakami H, Asai N, Takahashi M. Mechanism

    of Ret dysfunction by Hirschsprung mutations affecting its

    extracellular domain. Hum Mol Genet 1996; 5: 15780.

    25. Cosma MP, Cardone M, Carlomagno F, Colantuoni V.

    Mutations in the extracellular domain cause RET loss of

    function by a dominant negative mechanism. Mol Cell Biol

    1998; 18: 3329.

    26. Ishiguro Y, Iwashita T, Murakami H et al. The role of amino

    acids surrounding tyrosine 1062 in Ret in specific binding of

    the Shc phosphotyrosine-binding domain. Endocrinology

    1999; 140: 3998.

    27. Geneste O, Bidaud C, De Vita G et al. Two distinct mutations

    of the RET receptor causing Hirschsprungs disease impair

    the binding of signalling effecters to a multifunctional

    docking site. Hum Mol Genet 1999; 8: 19899.

    28. Pasini B, Borrello MG, Greco A et al. Loss of function effect of

    RETmutations causing Hirschsprung disease

    Nature Genet

    1995; 10: 340.

    29. Pelet A, Geneste O, Edery P et al. Various mechanisms cause

    RET-mediated signalling defects in Hirschsprungs disease.

    J Clin Invest 1998; 101: 14123.

    30. Cosma MP, Panariello L, Quadro L, Dathan NA, Fattoruso O,

    Colantuoni V. A mutation in the RET proto-oncogene in

    Hirschsprungs disease affects the tyrosine kinase activity

    associated with multiple endocrine neoplasia type 2A and

    2B. Biochem J 1996; 314: 397400.

    2003 Blackwell Publishing Ltd Journal of Internal Medicine 253: 627633

    6 32 K . K UR OK AW A et al.

  • 7/29/2019 Cell Signalling and Gene Expression Mediated by RET

    7/7

    31. Iwashita T, Kurokawa K, Qiao S et al. Functional analysis of

    RET with Hirschsprung mutations affecting its kinase do-

    main. Gastroenterology 2001; 121: 233.

    32. Tahira T, Ishizaka Y, Itoh F, Sugimura T, Nagao M. Char-

    acterization of ret proto-oncogene mRNAs encoding two

    isoforms of the protein product in a human neuroblastoma

    cell line. Oncogene 1990; 5: 9102.

    33. Myers SM, Eng C, Ponder BAJ, Mulligan LM. Characteriza-

    tion of RET proto-oncogene 3-splicing variants and polya-

    denylation sites: a novel C-terminus for RET. Oncogene 1995;

    11: 20345.

    34. Coulpier M, Anders J, Ibanez CF. Coordinated activation of

    autophosphorylation sites in the RET receptor tyrosine kin-

    ase importance of tyrosine 1062 for GDNF mediated

    neuronal differentiation and survival. J Biol Chem 2002;

    277: 1999.

    35. Borrello MG, Alberti L, Arighi E et al. The full oncogenic

    activity of Ret/ptc2 depends on tyrosine 539, a docking site

    for phospholipase C-c. Mol Cell Biol 1996; 16: 21563.

    36. Asai N, Murakami H, Iwashita T, Takahashi M. A mutation

    at tyrosine 1062 in MEN2A-Ret and MEN2B-Ret impairs

    their transforming activity and association with Shc adaptor

    proteins. J Biol Chem 1996; 271: 17649.

    37. Arighi E, Alberti L, Torriti F et al. Identification of Shc docking

    site on Ret tyrosine kinase. Oncogene 1997; 14: 7782.

    38. Lorenzo MJ, Gish GD, Houghton C et al. RET alternate spli-

    cing influences the interaction of activated RET with the SH2

    and PTB domains of Shc, and the SH2 domain of Grb2.

    Oncogene 1997; 14: 7671.

    39. Kurokawa K, Iwashita T, Murakami H, Hayashi H, Kawai K,

    Takahashi M. Identification of SNT/FRS2 docking site on

    RET receptor tyrosine kinase and its role for signal trans-

    duction. Oncogene 2001; 20: 19238.

    40. Melillo RM, Santoro M, Ong SH et al. Docking protein FRS2

    links the protein tyrosine kinase RET and its oncogenic forms

    with the mitogen-activated protein kinase signalling cascade.

    Mol Cell Biol 2001; 21: 41787.

    41. Melillo RM, Carlomagno F, De Vita G et al. The insulin re-

    ceptor substrate (IRS)-1 recruits phosphatidylinositol 3-kin-

    ase to Ret: evidence for a competition between Shc and IRS-1

    for the binding to Ret. Oncogene 2001; 20: 2018.

    42. Murakami H, Yamamura Y, Shimono Y, Kawai K, Kurokawa

    K, Takahashi M. Role of Dok1 in cell signalling mediated by

    RET tyrosine kinase. J Biol Chem 2002; 277: 327890.

    43. Grimm J, Sachs M, Britsch S et al. Novel p62dok family

    members, dok-4 and dok-5, are substrates of the c-Ret

    receptor tyrosine kinase and mediate neuronal differenti-

    ation. J Cell Biol 2001; 154: 3454.

    44. Durick K, Gill GN, Taylor SS. Shc and Enigma are both re-

    quired for mitogenic signalling by Ret/ptc2. Mol Cell Biol

    1998; 18: 229308.

    45. Hayashi H, Ichihara M, Iwashita Tet al.

    Characterization ofintracellular signals via tyrosine 1062 in RET activated by

    glial cell line-derived neurotrophic factor. Oncogene 2000;

    19: 44675.

    46. Hayashi Y, Iwashita T, Murakamai H et al. Activation of

    BMK1 via tyrosine 1062 in RET by GDNF and MEN2A

    mutation. Biochem Biophys Res Commun 2001; 281: 689.

    47. Besset V, Scott RP, Ibanez CF. Signaling complexes and

    protein-protein interactions involved in the activation of the

    Ras and phosphatidylinositol 3-kinase pathways by the c-Ret

    receptor tyrosine kinase. J Biol Chem 2000; 275: 391566.

    48. Murakami H, Iwashita T, Asai N et al. Enhanced phospha-

    tidylinositol 3-kinase activity and high phosphorylation state

    of its downstream signalling molecules mediated by Ret with

    the MEN 2B mutation. Biochem Biophys Res Commun 1999;

    262: 675.

    49. Salvatore D, Melillo RM, Monaco C et al. Increased in vivo

    phosphorylation of Ret tyrosine 1062 is a potential patho-

    genetic mechanism of multiple endocrine neoplasia type 2B.

    Cancer Res 2001; 61: 142631.

    50. Forman-Kay JD, Pawson T. Diversity in protein recognition

    by PTB domains. Curr Opin Struct Biol 1999; 9: 695.

    51. Xu H, Lee KW, Goldfarb M. Novel recognition motif on fi-

    broblast growth factor receptor mediates direct association

    and activation of SNT adapter proteins. J Biol Chem 1998;

    273: 179890.

    52. Ong SH, Guy GR, Hadari YR et al. FRS2 proteins recruit

    intracellular signalling pathways by binding to diverse tar-

    gets on fibroblast growth factor and nerve growth factor

    receptors. Mol Cell Biol 2000; 20: 9789.

    53. Dhalluin C, Yan KS, Plotnikova O et al. Structural basis of

    SNT PTB domain interactions with distinct neurotrophic

    receptors. Mol Cell 2000; 6: 929.

    54. Yan KS, Kuti M, Yan S et al. FRS2 PTB domain conformation

    regulates interactions with divergent neurotrophic receptors.

    J Biol Chem 2002; 277: 170894.

    55. Tansey MG, Baloh RH, Milbrandt J, Johnson EM. GFRa-me-

    diated localization of RET to lipid rafts is required for effective

    downstream signalling, differentiation, and neuronal survi-

    val. Neuron 2000; 25: 6123.

    56. Paratcha G, Ledda F, Baars L et al. Released GFRa1 poten-

    tiates downstream signalling, neuronal survival, and differ-

    entiation via a novel mechanism of recruitment of c-Ret to

    lipid rafts. Neuron 2001; 29: 1784.

    57. Kouhara H, Hadari YR, Spivak-Kroizman T et al. A lipid-

    anchored Grb2-binding protein that links FGF-receptor ac-

    tivation to the Ras/MAPK signalling pathway. Cell 1997; 89:

    69702.

    58. de Graaff E, Srinivas S, Kilkenny C et al. Differential activities

    of the RET tyrosine kinase receptor isoforms during mam-

    malian embryogenesis Genes Dev 2001; 15: 24344.

    59. Tsui-Pierchala BA, Ahrens RC, Crowder RJ, Milbrandt J,

    Johnson EM. The long and short isoforms of Ret function as

    independent signalling complexes. J Biol Chem 2002; 277:

    346125.

    60. Zhou MM, Ravichandran KS, Olejniczak ET et al. Structure

    and ligand recognition of the phosphotyrosine binding do-

    main of Shc. Nature 1995; 378: 5892.

    61. Ohiwa M, Murakami H, Iwashita T et al. Characterization of

    Ret-Shc-Grb2 complex induced by GDNF, MEN 2A, and MEN

    2B mutations. Biochem Biophys Res Commun 1997; 237: 74

    51.

    62. Watanabe T, Ichihara M, Hashimoto Met al.

    Characteriza-tion of gene expression induced by RET with MEN2A or

    MEN2B mutation. Am J Pathol 2002; 161: 2456.

    Received 12 March 2003; accepted 20 March 2003.

    Correspondence: Masahide Takahashi MD, PhD, Department of

    Pathology, Nagoya University Graduate School of Medicine,

    65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan (fax:

    181 52 7442098; e-mail: [email protected]).

    2003 Blackwell Publishing Ltd Journal of Internal Medicine 253: 627633

    M I N I SY M P O SI U M : C E L L S I G N AL L I N G B Y R E T 6 3 3