当前位置: 首页 > 期刊 > 《肿瘤与内分泌》 > 2005年第2期 > 正文
编号:11330260
The epidermal growth factor receptor family
http://www.100md.com 《肿瘤与内分泌》
     Cancer Biology Laboratory, Research School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK

    Abstract

    The epidermal growth factor receptor family consists of four receptor genes and at least 11 ligands, several of which are produced in different protein forms. They create an interacting system that has the ability to receive and process information that results in multiple outputs. The family has an important role in directing and coordinating many normal processes, including growth and development, normal tissue turnover and wound healing. Its members are also aberrantly activated by overexpression or mutation in many common human tumour types and as such have been the target for anticancer drug development.

    The epidermal growth factor receptor (EGFR) family

    The epidermal growth factor (EGF) receptor (R) belongs to the ErbB family of receptor tyrosine kinases (RTKs). These receptors possess protein tyrosine kinase (PTK) activity and are found only in metazoans, in contrast to many of the serine/threonine kinase families, which are conserved throughout eukaryotes and are found in both unicellular and multicellular organisms (Stein & Staros 2000). The four receptor genes, encoding the EGF receptor (EGFR/erbB-1), c-erbB-2/HER2, c-erbB3-/HER3 and c-erbB4/HER4, can be alternatively spliced to give rise to various partial protein products; in the case of c-erbB-4, these include four variations of the full-length receptor. Examination of the completed human genome sequence has determined that 58 of the 90 tyrosine kinase sequences identified are RTKs (Robinson et al. 2000). Furthermore, there are no additional sequences that are potentially new members of the growth factor receptor family (Robinson et al. 2000). The ErbB nexus, comprising the four interacting receptor types found in mammals, appears to be an evolutionary elaboration of the fundamental module found in the worm Caenorhabditis elegans (named LET-23 or lethal complementation group 23) and the equivalent single-gene homologue (expressed as three splice variants) found in the fruit fly Drosophila melanogaster, namely, DER (see Moghal & Sternberg (1999) for a comparative review). The roles played by the type 1 growth factors and receptors in normal development, differentiation, migration, wound healing and apoptosis are essential for the viability of multicellular organisms; the effect of aberrant function is demonstrated in disease states such as cancer.

    The 11 ligands currently identified for these receptors in mammals are EGF, transforming growth factor- (TGF-), HB-EGF (heparin binding), beta-cellulin, amphiregulin, epiregulin, epigen and the neuregulins (NRGs) 1–4 (Olayioye et al. 2000). A significant proportion of these are initially expressed as membrane-anchored proteins that require proteolytic cleavage either to achieve activity in solution or bind to cell surface proteoglycans from where they can act as a reservoir to be made available for receptor binding (Falls 2003). Two other genes, tomoregulin and epiglycan C, have recently been suggested to represent NRG 5 and 6 respectively, but further work is needed to confirm this assertion (Kinugasa et al. 2004). Determination of the ways in which this plethora of proteins interact and the resultant biological effects will be necessary to unravel the temporal and spatial nature of the mechanisms involved in this system (Jones et al. 1999).

    A simple model by which one can begin to understand the complex system of growth factor signalling is based upon perceiving the network as three individual, sequential layers (Gullick 2001). The initial, extracellular layer is composed of the ligands, and its nature is therefore determined by their concentration and bioavailability. These two parameters dictate whether the receptors that reside within the cell membrane (and comprise the second layer of the system) will dimerise to become active. If the information in the first layer is sufficient to induce receptor dimerisation and consequently increase catalytic activity, the third, intracellular layer of second messenger proteins can bind to specific sites on the receptors and initiate the signals required to induce the appropriate response. As discussed below, it is now evident that most or all of the ErbB family of receptors further aggregate into oligomers of several hundreds or a few thousand receptors that recruit one or more second messenger proteins. Clearly, there are opportunities for this to allow interactions in the ‘third layer’, creating a system capable of integrating many inputs and producing multiple outputs, in many respects resembling a computational device (Johnson et al. 2004).

    Modular structure of the EGFR

    The first RTK to be discovered was the EGFR (Carpenter et al. 1978), the study of which has subsequently yielded insight into the underlying mechanisms by which RTKs function (Schlessinger 2000), including the recruitment and activation of second messengers (reviewed by Jorissen et al. 2003). Furthermore, it was this growth factor receptor that first identified a relationship between an activated oncogene and cancer (Downward et al. 1984).

    Each member of the ErbB family comprises a conserved protein tyrosine kinase domain that resides within the cytoplasm, a transmembrane domain that makes a single pass through the plasma membrane, and a glycosylated, extracellular ligand-binding domain (Fig. 1). In the EGF receptor family, this last domain exhibits four subdomains denominated L1, S1 (CR1), L2 and S2 (CR2) (or, more simply, I, II, III and IV respectively) (Lax et al. 1989). Of these domains, S1 and S2 are homologous, cysteine-rich regions (CR1 and CR2), while L1 and L2 form the ligand-binding site (Garrett et al. 2002, Ogiso et al. 2002). It is likely that these are derived from an ancient gene duplication event (Stein & Staros 2000), and it is notable that the cysteine residues do not form disulphide bonds between the two S1/S2 domains.

    Receptor–receptor interactions

    In the absence of ligand, monomeric receptors reside within the cell membrane in an inactive state distributed fairly evenly over the cell membrane (although there are reports of concentrations of receptors in structures called calveolae and at the base of microspikes and in membrane ruffles). However, when ligand is present in the surrounding milieu, the receptor sites become occupied by monomeric EGF. Provided two stable 1:1 EGF:EGFR intermediate complexes persist, sequential receptor dimerisation and oligomerisation ensue (Ferguson et al. 2003). Receptor dimers, which have greater stability and ligand-binding affinity than their corresponding monomers (Ben-Levy et al. 1992, Zhou et al. 1993), are increasingly considered to be the minimal number of clustered receptors necessary for activation (Alroy & Yarden 1997). Such dimerisation is accomplished through the interaction of one EGFR S1 domain with that of a second ErbB receptor in a 2:2 receptor:ligand complex. Each S1 domain projects an extracellular ‘dimerisation loop’, and it is thought that the homophilic interactions between the two loops drive dimerisation. This scenario was elucidated by the crystal structure of ligand-bound EGFR that has recently been solved (Fig. 2) (Garrett et al. 2002, Ogiso et al. 2002).

    Dimerisation may also involve direct contact between the helical transmembrane structures of the receptors, the attractive forces involved being either van der Waals interactions (Lemmon et al. 1997, White & Wimley 1999) or hydrogen bonding (Choma et al. 2000, Senes et al. 2001). Such close association between these domains is thought to be permitted by the amino-acid ‘dimerisation motifs’ contained within their 22–26 hydrophobic residues (Sternberg & Gullick 1989, 1990, Lemmon et al. 1994). These motifs were originally described as sequences of five residues, the first of which is a small side chain, the fourth an aliphatic chain, and the fifth either Gly or Ala (Sternberg & Gullick 1990). Particular point mutations within this sequence, such as the substitution of valine by glutamate, lead to constitutive dimerisation that, in the case of the c-erbB-2 receptor, is capable of inducing cell transformation (Brandt-Rauf et al. 1990, 1995, Sternberg & Gullick 1990, Smith et al. 1996, Gullick & Srinivasan 1998, Sharpe et al. 2000). The transmembrane domain is hypothesised to exert a high degree of regulation over receptor association, spatial arrangement and consequently signalling dynamics (Alroy & Yarden 1997, Gullick & Srinivasan 1998).

    It is now evident that further oligomerisation of these receptors occurs in response to ligand binding and second messenger recruitment. Even in the seminal experiments of Yarden and Schlessinger (1987), who first reported the phenomenon of dimerisation, it was evident that higher-order aggregates were seen, and the authors ‘concluded that receptor oligomerisation is an intrinsic property of the occupied EGF receptor’. Now, by green fluorescent protein tagging (Hayes et al. 2004), the process can be followed visually (for films and still images, see Gillham et al. (1999) and www.kent.ac.uk/bio/gullick/). We and other groups are investigating the significance of these ‘signalling platforms’, which, by recruiting different receptor types and multiple second messenger proteins, may add further levels of complexity and capability to the process.

    Homodimers and heterodimers

    Encoded by the ERBB2 proto-oncogene located on chromosome 17q21, c-erbB-2 is a 1255-amino-acid transmembrane glycoprotein of 185 kDa (Coussens et al. 1985), which is thought to function as a ligand-less coreceptor for the other three members of the ErbB family and therefore for the majority (if not all) of the ErbB ligands (Klapper et al. 1999, Rubin & Yarden 2001). In response to transactivation by EGF-like ligands, such as TGF- or epiregulin, c-erbB-2 receptors can form heterodimers with EGF receptors (Pinkas-Kramarski et al. 1996, 1997), or with c-erbB-3 and c-erbB-4 when activated by the NRGs (Tzahar et al. 1996, Burden & Yarden 1997, Pinkas-Kramarski et al. 1998). Indeed, c-erbB-2 is the preferred dimerisation partner in a distinct hierarchy that determines the interreceptor interactions (Tzahar et al. 1996) that are essential for normal Schwann cell (Morrissey et al. 1995) and certain epithelial cell behaviour (Klapper et al. 1997), and for determining specific cell lineages.

    The published crystal structure of the extracellular domain of c-erbB-2 (Cho et al. 2003) reveals that, in the absence of direct ligand binding, c-erbB-2 adopts a fixed conformation resembling a ligand-activated state and, as such, is permanently capable of interacting with other members of the ErbB family. In contrast to c-erbB-2, this conformation is adopted only by the EGFR (Ferguson et al. 2003) and c-erbB-3 (Cho et al. 2002) upon ligand binding, and is characterised by a long, extracellular, finger-like projection from domain II (the ‘dimerisation loop’) that mediates interaction with the corresponding structure on its dimerisation partner (Garrett et al. 2002, Ogiso et al. 2002). In the absence of ligand, the dimerisation loop makes intramolecular contact with a pocket on domain IV, thereby restraining the receptor in an autoinhibited conformation. The fixed, open structure of c-erbB-2 confers upon it an intrinsic capability to interact with other ligand-bound receptors by circumventing the requirement for ligand binding to release the dimerisation loop from an autoinhibited conformation (Cho et al. 2003). This absence of a barrier against auto-activation contributes to the transforming potential of c-erbB-2 when overexpressed in cell culture (Di Fiore et al. 1987, Hudziak et al. 1987, Cho et al. 2003) and may enable c-erbB-2 homodimers to function as a constitutively active kinase (Lonardo et al. 1990). Overexpression of c-erbB-2 and the spontaneous (hetero)- dimerisation that ensues has been shown not only to cause an increase in basal receptor phosphorylation and activation (D’Souza et al. 1993, Samanta et al. 1994, Ram & Ethier 1996, Worthylake et al. 1999), but also to inhibit downregulation by reducing the degradation rate of the transphosphorylated pool of receptors (Worthylake et al. 1999).

    It is interesting to note that the NRGs (ligands for the c-erbB-3 and c-erbB-4 receptors) are subject to alternative splicing that results in ligands with differing affinities for their receptors. Consequently, different receptors are recruited, so that this alternative splicing serves as a further mechanism by which the required heterodimers are achieved (Gullick & Srinivasan 1998). Such diversity of interaction between ligands and their receptors dictates the strength of the signalling kinetics and consequently the magnitude of the output and its specificity (as in growth or differentiation) (Marshall 1995).

    Activation of the EGFR cytoplasmic PTK domain

    The cytoplasmic region of the EGFR comprises three distinct domains: 1. the juxtamembrane domain, required for feedback by protein kinase C (PKC); 2. the noncatalytic carboxy-terminal tail, possessing the six tyrosine transphosphorylation sites mandatory for recruitment of adaptor/effector proteins (e.g. Grb2 and phospholipase C (PLC) respectively) containing SH2 domains (src homology domain 2) or PTB (phosphotyrosine binding) domains, plus the motifs necessary for internalisation and degradation of the receptor; 3. the central tyrosine kinase domain (src homology domain 1 (SH1)) that is responsible for mediating transphosphorylation of the six carboxyterminal tyrosine residues.

    It is thought that prior to EGF binding, the activation loop that resides within the cytoplasmic PTK domain of the EGFR may adopt an inactive conformation that is inaccessible to both substrate and ATP. Consequent to ligand binding and receptor dimerisation, intracellular tyrosine kinase catalytic activity is increased. The majority of RTKs absolutely require tyrosine phosphorylation within their activation loop for catalytic activity and biological function (reviewed by Hubbard & Till 2000, Dibb et al. 2004). However, the major exception to this is the EGFR, which instead is thought to require dimerisation of the cytoplasmic domain for catalytic enhancement (Mohammadi et al. 1993, Sherrill 1997), and this appears to be sufficient to stimulate cross-phosphorylation between the two receptors at 3–6 specific tyrosine residues. Recently, it has been suggested that in c-erbB-4 the tyrosine residues that become phosphorylated may be determined by the type of growth factor that has bound the receptor (Sweeney & Carraway 2000); this, in turn, determines the identity of the second messenger recruited (Margolis 1992, van der Geer & Pawson 1995). The tyrosine residues and surrounding amino acids of each member of the ErbB family are specifically tailored to interact with a unique collection of second messengers such that the specific biological and biochemical response may be precisely induced (Carraway & Cantley 1994, Alroy & Yarden 1997, Olayioye et al. 2000). Further observations have indicated that these properties are not unique to c-erbB-4 but are characteristic of all the ErbB family members (Crovello et al. 1998, Sweeney & Carraway 2000) and may be required for tissue-specific biological activity.

    Second messengers

    Subsequent to transphosphorylation of the receptor dimer, second messenger proteins possessing one of the two main classes of domains that recognise site-specific phosphorylation (‘docking sites’) can interact with the receptors. The larger of the two groups of second messenger proteins interact with the receptor via an SH2 (src homology-2) structure, whereas the others exhibit what is known as a PTB domain (Pawson 1995). Both are modular proteins (Kuriyan & Cowburn 1997, Pawson & Scott 1997, Margolis 1999) and serve to transmit the signal received at the receptor to either the cytoplasm or the nucleus. The specificity of PTB domain binding is dictated by the particular sequence of 3-5 amino-acid residues in the N-terminal stretch of activated EGFR that precedes the phosphotyrosine (pTyr) moiety (Prigent & Gullick 1994, Margolis 1999). Conversely, SH2 domains bind to specific amino-acid sequences 1–6 residues long that occur in the C-terminal sequence directly after the pTyr (Songyang et al. 1993). Furthermore, many SH2- and PTB-containing proteins can interact with phospholipids or nucleic acids and/or possess intrinsic enzymatic activity.

    An example of a well-characterised second messenger/receptor interaction is the recruitment of the enzyme phospholipase C gamma (PLC) (Wang et al. 2001). In its inactive state, PLC is normally found in the cytosol. However, upon phosphorylation of the EGFR, the SH2 domain(s) of PLC are able to interact with the phosphorylated receptor. This causes not only its tyrosine phosphorylation by the activated receptor, but also relocation to the membrane, where it makes contact with the substrate PtdIns(3,4,5)P3 and ultimately generates the second messengers Ins(1,4,5)P3 and diacylglycerol (Falasca et al. 1998). A very similar redistribution occurs for several other second messengers (Gillham et al. 1999, Hayes et al. 2004). In addition to this effector protein activation by tyrosine phosphorylation, the two alternatives ‘activation by membrane translocation’ and ‘activation by a conformational change’ have been theorised, and the three are reviewed by Schlessinger (2000).

    Proteins that contain SH2 or SH3 domains but lack enzymatic activity mediate signalling by acting as a platform upon which a specific complement of signalling proteins can be recruited and assembled, thereby linking the receptor to a specific signalling cascade (Pawson & Schlessinger 1993). This is exemplified by the adaptor protein Grb2 interacting with activated EGFR and recruiting the guanine nucleotide release factor Son of sevenless (Sos). In so doing, Sos is brought into close proximity with the plasma membrane and consequently its target protein Ras (Schlessinger 1994, Pawson 1995). Alternatively, EGFR can phosphorylate docking proteins, such as Gab1 (Mattoon et al. 2004), which then interact with multiple effector proteins.

    In spite of the magnitude of information obtained, it has yet to be determined whether receptor dimers can recruit second messengers or whether receptor clustering is a prerequisite. Furthermore, it remains unknown whether a second messenger can bind more than one receptor at once, or indeed whether a receptor can bind multiple second messengers simultaneously (Gullick 2001).

    The strength of ligand binding and signalling by heterodimers containing c-erbB-2 is significantly greater than that of either homodimers or heterodimers that do not recruit this receptor (Sliwkowski et al. 1994, Karunagaran et al. 1996). Dissociation of ligand:receptor complexes is decelerated by c-erbB-2 such that perpetuated ligand binding to c-erbB-2, c-erbB-3 or EGFR causes enhanced and prolonged stimulation of the MAP kinase (ERK) and c-Jun kinase (SAPK) pathways by both the NRGs and EGF (Karunagaran et al. 1996). Signal duration and potency is further enhanced by a slow rate of internalisation compared with that of the EGFR (Sorkin et al. 1993, Baulida et al. 1996). Internalisation is a process that serves to reduce the number of functional receptors at the plasma membrane and attenuate the strength of the signal generated. However, the sequences contained within the carboxyl terminus of c-erbB-2 (Sorkin et al. 1993) confer only a weak and ineffective coupling between this receptor and c-Cbl (Levkowitz et al. 1996, Graus-Porta et al. 1997, Muthuswamy et al. 1999) and an inability to associate with AP-2 (a plasma membrane-coated pit adaptor complex) (Gilboa et al. 1995) such that internalisation is impaired.

    In terms of cell growth and transformation, the most potent heterodimer of the ErbB receptor family is composed of c-erbB-2 and c-erbB-3 (Wallasch et al. 1995, Pinkas-Kramarski et al. 1996), of which c-erbB-3 is a defective kinase (Guy et al. 1994) capable of binding some of the isoforms of the NRGs (Tzahar et al. 1994). Not only does the dimerisation of c-erbB-2 with c-erbB-3 increase the binding affinity of these sites, but the diversity of potential ligands to which they may bind is also extended to include EGF and betacellulin (Alimandi et al. 1997, Pinkas-Kramarski et al. 1998). This is particularly relevant to c-erbB-2-overexpressing human adenocarcinomas, in which c-erbB-3 is ubiquitously expressed at moderate or high levels (Lemoine et al. 1992) such that the combination of the two promotes ligand-binding promiscuity (Pinkas-Kramarski et al. 1998). An abundant supply of ErbB ligands, including EGF and betacellulin, originates from the mesenchyme-derived stroma underlying the layers of epithelial cells that are the precursors of adenocarcinomas. Consequently, this paracrine mesenchyme–epithelial interaction is thought to play a crucial role in tumour development (Salomon et al. 1995).

    The collaboration of c-erbB-2 and c-erbB-3 leads to the activation of several of the major signal transduction pathways. Subsequent to ligand binding and dimerisation, c-erbB-3 becomes transphosphorylated by c-erbB-2, causing several carboxyl-terminus phosphotyrosine residues in each receptor to undergo the phosphorylation (Wallasch et al. 1995) required to permit interaction with intracellular signalling proteins (second messengers). Consequently, many of the c-erbB-3 phosphotyrosine moieties are able to recruit PI-3 kinase and thereby activate the Akt pathway (Prigent & Gullick 1994), whereas a single phosphotyrosine residue of c-erbB-2 is required for the recruitment of Shc and stimulation of the mitogen-activated protein kinase (MAPK) pathway, an interaction sufficient for cell transformation (Ben-Levy et al. 1994).

    Downstream signalling pathways

    The MAPK and PI3K/Akt pathways promote cell proliferative and survival/antiapoptotic signals via the activation of transcription factors and upregulation of cyclin D1 (Diehl et al. 1998, Lee et al. 2000, Lenferink et al. 2001, Wulf et al. 2001, Citri et al. 2003). Enhanced levels of functional c-erbB-2 cause an increase in cyclin D1 that functions to sequester the cyclin kinase inhibitor p27 and release Cdk2 (Neve et al. 2000). Subsequently, Cdk2 becomes positively regulated via its association with cyclin E and causes deregulation of the G1/S checkpoint such that the cell-cycle progression is promoted and leads to malignant transformation (reviewed in Harari & Yarden 2000). Overexpression of cyclin D1 at a 40% incidence has led to its identification as a significant factor in the development of breast cancer (Bartkova et al. 1994).

    The downstream effectors of Akt also serve to sequester p27 such that the constitutive activation of Akt that arises from c-erbB-2-overexpression is thought to confer resistance to tumour necrosis factor-induced apoptosis (Zhou et al. 2000). Antiapoptotic signalling is further mediated by significant upregulation of the CDK inhibitor p21Waf1 (Waf1, Cip1, Sdil) (Fan et al. 1995, 1997, Bacus et al. 1996, Fiddes et al. 1998, Yu et al. 1998). It is this upregulation and consequent decrease in cyclinB-Cdc2 activation that is thought to protect against the paclitaxel-induced cell death exhibited by c-erbB-2-overexpressing cells (Yu et al. 1998).

    Additional targets of c-erbB-2/c-erbB-3 signalling are the PLC pathway and the JAK-STAT pathway. PLC is activated by docking at specific sites solely on c-erbB-2 (Fedi et al. 1994) and hydrolyses PIP2 into the signalling molecules IP3 and DAG required for calcium/calmodulin-dependent kinases and stimulation of protein kinase C.

    The full-length c-erbB-2 protein (p185) overexpressed by cultured tumour cells is subject to slow proteolytic cleavage that releases the Mr 110 000 (p110) extracellular domain (ECDc-erbB-2) into the conditioned medium (Lin & Clinton 1991, Zabrecky et al. 1991, Pupa et al. 1993). Soluble ECDc-erbB-2 was subsequently found in the serum of patients with advanced breast cancer in a manner that correlated well with c-erbB-2 overexpression in the primary tumour; in contrast, the sera of those patients in the early stages of the disease appear to be negative for this marker (Pupa et al. 1993). The shedding of ECDc-erbB-2 is thought to be a process regulated by tyrosine phosphorylation (Codony-Servat et al. 1999) that results in the constitutive activation of the remaining membrane-associated c-erbB-2 domains (Segatto et al. 1988, Huang et al. 1997). Such potentially increased signalling may explain the association of high ECDc-erbB-2 serum levels with a poor prognosis and limited response to endocrine therapy and chemotherapy in patients with advanced breast cancer (Di Fiore et al. 1987, Segatto et al. 1988).

    A second marker of Mr 95 000 was later discovered in cancer tissues that correlated with the levels of p110 shed from the surface of c-erbB-2-overexpressing tumour cells. This was determined to be an NH2-terminally truncated c-erbB-2 product possessing in vitro kinase activity (Christianson et al. 1998). Subsequently, a relationship was revealed between the expression of this truncated protein and metastasis to the lymph nodes in p95-positive breast cancer patients; p95-negative tumours were associated with node-negative patients (Christianson et al. 1998). However, this study found no correlation between p185 or p95 enrichment in patients and tumour size or hormone receptor status.

    Summary

    The complexity of EGFR signalling is great. However, new technologies are evolving, such as fluorescently labelled receptors, ligands and second messengers expressed by live cells that can be detected by digital microscopy, which offer the opportunity of visualising not only the interplay between these signalling components but also their trafficking and positioning within the cell (Hayes et al. 2004). Moreover, prototype computational models of growth factor receptor clustering have been developed (Johnson et al. 2004) that will in time also contribute to obtaining further information about signalling interactions. Genetic manipulation (knockins and -outs) and the ability to suppress protein levels (knockdowns) will be critical tools in cell biology experiments. Finally, the growing use of antibodies and small molecule receptor tyrosine kinase inhibitors in large numbers of cancer patients will reinforce and inform laboratory experiments. Ultimately, however, the problem is soluble and not only will lead to a comprehensive description of the system and better cancer treatments but also represents an example where the new methods can be applied to other complex systems.

    Acknowledgements

    Laura Bazley is the recipient of a postgraduate studentship from the Breast Cancer Campaign.

    Funding

    Laura Bazley declares no conflict of interest. Bill Gullick has received payments from AstraZeneca for work associated with several advisory boards.

    References

    Alimandi M, Wang LM, Bottaro D, Lee CC, Kuo A, Frankel M, Fedi P, Tang C, Lippman M & Pierce JH 1997 Epidermal growth factor and betacellulin mediate signal transduction through co-expressed ErbB2 and ErbB3 receptors. EMBO Journal 16 5608–5617.

    Alroy I & Yarden Y 1997 The ErbB signaling network in embryogenesis and oncogenesis: signal diversification through combinatorial ligand–receptor interactions. FEBS Letters 410 83–86.

    Bacus SS, Yarden Y, Oren M, Chin DM, Lyass L, Zelnick CR, Kazarov A, Toyofuku W, Gray-Bablin J, Beerli RR, Hynes NE, Nikiforov M, Haffner R, Gudkov A & Keyomarsi K 1996 Neu differentiation factor (heregulin) activates a p53-dependent pathway in cancer cells. Oncogene 12 2535–2547.

    Bartkova J, Lukas J, Muller H, Lutzhoft D, Strauss M & Bartek J 1994 Cyclin D1 protein expression and function in human breast cancer. International Journal of Cancer 57 353–361.

    Baulida J, Kraus MH, Alimandi M, Di Fiore PP & Carpenter G 1996 All ErbB receptors other than the epidermal growth factor receptor are endocytosis impaired. Journal of Biological Chemistry 271 5251–5257.

    Ben-Levy R, Peles E, Goldman-Michael R & Yarden Y 1992 An oncogenic point mutation confers high affinity ligand binding to the neu receptor. Implications for the generation of site heterogeneity. Journal of Biological Chemistry 267 17304–17313.

    Ben-Levy R, Paterson HF, Marshall CJ & Yarden Y 1994 A single autophosphorylation site confers oncogenicity to the Neu/ErbB-2 receptor and enables coupling to the MAP kinase pathway. EMBO Journal 13 3302–3311.

    Brandt-Rauf PW, Rackovsky S & Pincus MR 1990 Correlation of the structure of the transmembrane domain of the neu oncogene-encoded p185 protein with its function. PNAS 87 8660–8664.

    Brandt-Rauf PW, Pincus MR & Monaco R 1995 Conformation of the transmembrane domain of the c-erbB-2 oncogene-encoded protein in its monomeric and dimeric states. Journal of Protein Chemistry 14 33–40.

    Burden S & Yarden Y 1997 Neuregulins and their receptors: a versatile signaling module in organogenesis and oncogenesis. Neuron 18 847–855.

    Carpenter G 2000 The EGF receptor: a nexus for trafficking and signaling. Bioessays 22 697–707.

    Carpenter G, King L Jr & Cohen S 1978 Epidermal growth factor stimulates phosphorylation in membrane preparations in vitro. Nature 276 409–410.

    Carraway KL 3rd & Cantley LC 1994 A neu acquaintance for ErbB3 and ErbB4: a role for receptor heterodimerisation in growth signaling. Cell 78 5–8.

    Cho HS & Leahy DJ 2002 Structure of the extracellular region of HER3 reveals an interdomain tether. Science 297 1330–1333.

    Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney DW Jr & Leahy DJ 2003 Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature 421 756–760.

    Choma C, Gratkowski H, Lear JD & DeGrado WF 2000 Asparagine-mediated self-association of a model transmembrane helix. Nature Structural Biology 7 161–166.

    Christianson TA, Doherty JK, Lin YJ, Ramsey EE, Holmes R, Keenan EJ & Clinton GM 1998 NH2- terminally truncated HER-2/neu protein: relationship with the shedding of the extracellular domain and the prognostic factors in breast cancer. Cancer Research 58 5123–5129.

    Citri A, Skaria KB & Yarden Y 2003 The deaf and the dumb: the biology of ErbB-2 and ErbB-3. Experimental Cell Research 284 54–65.

    Codony-Servat J, Albanell J, Lopez-Talavera JC, Arribas J & Baselga J 1999 Cleavage of the HER2 ectodomain is a pervandate-activable process that is inhibited by the tissue inhibitor TIMP-1 in breast cancer cells. Cancer Research 59 1196–1201.

    Coussens L, Yang-Feng TL, Liao Y-C, Chen E, Gray A, McGrath J, Seeburg PH, Libermann TA, Schlessinger J & Francke U 1985 Tyrosine kinase receptor with extensive homology to EGF receptor shares chromosomal location with neu oncogene. Science 230 1132–1139.

    Crovello CS, Lai C, Cantley LC & Carraway KL 3rd 1998 Differential signaling by the epidermal growth factor-like growth factors neuregulin-1 and neuregulin 2. Journal of Biological Chemistry 273 26954–26961.

    Dibb NJ, Dilworth SM & Mol CD 2004 Switching on kinases oncogenic activation of BRAF and the PDGFR family. Nature Reviews Cancer 4 718–727.

    Diehl JA, Cheng M, Roussel MF & Sherr CJ 1998 Glycogen synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular localization. Genes and Development 12 3499–3511.

    Di Fiore PP, Pierce JH, Kraus MH, Segatto O, King CR & Aaronson SA 1987 erbB-2 is a potent oncogene when overexpressed in NIH/3T3 cells. Science 237 178–182.

    Downward J, Yarden Y, Mayes E, Scrace G, Totty N, Stockwell P, Ullrich A, Schlessinger J & Waterfield MD 1984 Close similarity of epidermal growth factor receptor and v-erb-B oncogene protein sequences. Nature 307 521–527.

    D’Souza B, Berdichevsky F, Kyprianou N & Taylor-Papadimitriou J 1993 Collagen-induced morphogenesis and expression of the alpha 2-integrin subunit is inhibited in c-erbB2-transfected human mammary epithelial cells. Oncogene 8 1797–1806.

    Falasca M, Logan SK, Lehto VP, Baccante G, Lemmon MA & Schlessinger J 1998 Activation of phospholipase C gamma by PI 3-kinase-induced PH domain-mediated membrane targeting. EMBO Journal 17 414–422.

    Falls DL 2003 Neuregulins: functions, forms, and signaling strategies. Experimental Cell Research 284 14–30.

    Fan Z, Lu Y, Wu X, DeBlasio A, Koff A & Mendelsohn J 1995 Prolonged induction of p21Cip1/WAF1/CDK2/PCNA complex by epidermal growth factor receptor activation mediates ligand-induced A431 cell growth inhibition. Journal of Cell Biology 131 235–242.

    Fan Z, Shang BY, Lu Y, Chou JL & Mendelsohn J 1997 Reciprocal changes in p27(Kip1) and p21(Cip1) in growth inhibition mediated by blockade or overstimulation of epidermal growth factor receptors. Clinical Cancer Research 3 1943–1948.

    Fedi P, Pierce JH, di Fiore PP & Kraus MH 1994 Efficient coupling with phosphatidylinositol 3-kinase, but not phospholipase C gamma or GTPase-activating protein, distinguishes ErbB-3 signaling from that of other ErbB/EGFR family members. Molecular Cell Biology 14 492–500.

    Ferguson KM, Berger MB, Mendrola JM, Cho H-S, Leahy DJ & Lemmon MA 2003 EGF activates its receptor by removing interactions that autoinhibit ectodomain dimerization. Molecular Cell 11 507–517.

    Fiddes RJ, Janes PW, Sivertsen SP, Sutherland RL, Musgrove EA & Daly RJ 1998 Inhibition of the MAP kinase cascade blocks heregulin-induced cell cycle progression in T-47D human breast cancer cells. Oncogene 16 2803–2813.

    Garrett TP, McKern NM, Lou M, Elleman TC, Adams TE, Lovrecz GO, Zhu H-J, Walker F, Frenkel MJ, Hoyne PA, Jorissen RN, Nice EC, Burgess AW & Ward CW 2002 Crystal structure of a truncated epidermal growth factor receptor extracellular domain bound to transforming growth factor alpha. Cell 110 763–773.

    Gilboa L, Ben-Levy R, Yarden Y & Henis YI 1995 Roles for a cytoplasmic tyrosine and tyrosine kinase activity in the interactions of Neu receptors with coated pits. Journal of Biological Chemistry 270 7061–7067.

    Gillham H, Golding MC, Pepperkok R & Gullick WJ 1999 Intracellular movement of green fluorescent proteintagged phosphatidylinositol 3-kinase in response to growth factor receptor signaling. Journal of Cell Biology 146 869–880.

    Graus-Porta D, Beerli RR, Daly JM & Hynes NE 1997 ErbB-2, the preferred heterodimerisation partner of all ErbB receptors, is a mediator of lateral signaling. EMBO Journal 16 1647–1655.

    Gullick WJ 2001 The type 1 growth factor receptors and their ligands considered as a complex system. Endocrine-Related Cancer 8 75–82.

    Gullick WJ & Srinivasan R 1998 The type 1 growth factor receptor family: new ligands and receptors and their role in breast cancer. Breast Cancer Research and Treatment 52 43–53.

    Guy PM, Platko JV, Cantley LC, Cerione RA & Carraway KL 3rd 1994 Insect cell-expressed p180erbB3 possesses an impaired tyrosine kinase activity. PNAS 91 8132–8136.

    Harari D & Yarden Y 2000 Molecular mechanisms underlying ErbB2/HER2 action in breast cancer. Oncogene 19 6102–6114.

    Hayes N, Howard-Cofield E & Gullick WJ 2004 Green fluorescent protein as a tool to study epidermal growth factor receptor function. Cancer Letters 206 129–135.

    Huang HS, Nagane M, Klingbeil CK, Lin H, Nishikawa R, Ji XD, Huang CM, Gill GN, Wiley HS & Cavenee WK 1997 The enhanced tumorigenic activity of a mutant epidermal growth factor receptor common in human cancers is mediated by threshold levels of constitutive tyrosine phosphorylation and unattenuated signaling. Journal of Biological Chemistry 272 2927–2935.

    Hubbard SR & Till JH 2000 Protein tyrosine kinase structure and function. Annual Review of Biochemistry 69 373–398.

    Hudziak RM, Schlessinger J & Ullrich A 1987 Increased expression of the putative growth factor receptor p185HER2 causes transformation and tumorigenesis of NIH 3T3 cells. PNAS 84 7159–7163.

    Johnson CG, Goldman JP & Gullick WJ 2004 Simulating complex intracellular processes using object-oriented computational modelling. Progress in Biophysics and Molecular Biology 86 379–406.

    Jones JT, Akita RW & Sliwkowski MX 1999 Binding specificities and affinities of EGF domains for ErbB receptors. FEBS Letters 447 227–231.

    Jorissen RN, Walker F, Pouliot N, Garrett TP, Ward CW & Burgess AW 2003 Epidermal growth factor receptor: mechanisms of activation and signaling. Experimental Cell Research 284 31–53.

    Karunagaran D, Tzahar E, Beerli RR, Chen X, Graus-Porta D, Ratzkin BJ, Seger R, Hynes NE & Yarden Y 1996 ErbB-2 is a common auxiliary subunit of NDF and EGF receptors: implications for breast cancer. EMBO Journal 15 254–264.

    Kinugasa Y, Ishiguro H, Tokita Y, Oohira A, Ohmoto H & Higashiyama S 2004 Neuroglycan C, a novel member of the neuregulin family. Biochemical and Biophysical Research Communications 321 1045–1049.

    Klapper LN, Vaisman N, Hurwitz E, Pinkas-Kramarski R, Yarden Y, Sela M 1997 A subclass of tumor-inhibitory monoclonal antibodies to ErbB-2/HER2 blocks crosstalk with growth factor receptors. Oncogene 14 2099–2109.

    Klapper LN, Glathe S, Vaisman N, Hynes NE, Andrews GC, Sela M, Yarden Y 1999 The ErbB-2/HER2 oncoprotein of human carcinomas may function solely as a coreceptor for multiple stroma-derived growth factors. PNAS 96 4995–5000.

    Kuriyan J & Cowburn D 1997 Modular peptide recognition domains in eukaryotic signaling. Annual Review of Biophysics and Biomolecular Structure 26 259–288.

    Lane HA, Beuvink I, Motoyama AB, Daly JM, Neve RM & Hynes NE 2000 ErbB2 potentiates breast tumour proliferation through modulation of p27Kip1-Cdk2 complex formation: receptor overexpression does not determine growth factor dependency. Molecular and Cellular Biology 20 3210–3223.

    Lax I, Bellot F, Howk R, Ullrich A, Givol D & Schlessinger J 1989 Functional analysis of the ligand binding site of EGF receptor utilising chimeric chicken/human receptor molecules. EMBO Journal 8 421–427.

    Lee RJ, Albanese C, Fu M, D’Amico M, Lin B, Watanabe G, Haines GK 3rd, Siegel PM, Hung MC, Yarden Y, Horowitz JM, Muller WJ & Pestell RG 2000 Cyclin D1 is required for transformation by activated Neu and is induced through an E2F-dependent signaling pathway. Molecular and Cellular Biology 20 672–683.

    Lemmon MA, Treutlein HR, Adams PD, Brunger AT & Engelman DM 1994 A dimerization motif for transmembrane alpha helices. Nature Structural Biology 1 157–163.

    Lemmon MA, Bu Z, Ladbury JE, Zhou M, Pinchasi D, Lax I, Engelman DM & Schlessinger J 1997 Two EGF molecules contribute additively to stabilization of the EGF receptor dimer. EMBO Journal 16 281–294.

    Lemoine NR, Lobresco M, Leung H, Barton C, Hughes CM, Prigent SA, Gullick WJ & Kloppel G 1992 The ErbB-3 gene in pancreatic cancer. Journal of Pathology 168 269–273.

    Lenferink AE, Busse D, Flanagan WM, Yakes FM & Arteaga CL 2001 ErbB2/neu kinase modulates cellular p27(Kip1) and cyclin D1 through multiple signaling pathways. Cancer Research 61 6583–6591.

    Levkowitz G, Klapper LN, Tzahar E, Freywald A, Sela M & Yarden Y 1996 Coupling of the c-Cbl protooncogene product to ErbB-1/EGF-receptor but not to other ErbB proteins. Oncogene 12 1117–1125.

    Lin YZ & Clinton GM 1991 A soluble protein related to the HER-2 proto-oncogene product is released from human breast carcinoma cells. Oncogene 6 639–643.

    Lonardo F, Di Marco E, King CR, Pierce JH, Segatto O, Aaronson SA & Di Fiore PP 1990 The normal erbB-2 product is an atypical receptor-like tyrosine kinase with constitutive activity in the absence of ligand. New Biology 2 992–1003.

    Margolis B 1992 Proteins with SH2 domains: transducers in the tyrosine kinase signaling pathway. Cell Growth and Differentiation 3 73–80.

    Margolis B 1999 The PTB domain: the name doesn’t say it all. Trends in Endocrinology and Metabolism 10 262–267.

    Marshall CJ 1995 Specificity of receptor tyrosine kinase signaling: transient versus sustained extracellular signal-regulated kinase activation. Cell 80 179–185.

    Mattoon DR, Lamothe B, Lax I & Schlessinger J 2004 The docking protein Gab1 is the primary mediator of EGF-stimulated activation of the PI-3K/Akt cell survival pathway. BMC Biology 2 24.

    Moghal N & Sternberg PW 1999 Multiple positive and negative regulators of signaling by EGF-receptor. Current Opinion in Cell Biology 11 190–196.

    Mohammadi M, Honegger A, Sorokin A, Ullrich A, Schlessinger J & Hurwitz DR 1993 Aggregation-induced activation of the epidermal growth factor receptor protein tyrosine kinase. Biochemistry 32 8742–8748.

    Morrissey TK, Levi AD, Nuijens A, Sliwkowski MX, Bunge RP 1995 Axon-induced mitogenesis of human Schwann cells involves heregulin and p185erbB2. PNAS 92 1431–1435.

    Muthuswamy SK, Gilman M & Brugge JS 1999 Controlled dimerization of ErbB receptors provides evidence for differential signaling by homo- and heterodimers. Molecular and Cellular Biology 19 6845–6857.

    Neve RM, Sutterlüty H, Pullen N, Lane HA, Daly JM, Krek W & Hynes NE 2000 Effects of oncogenic ErbB2 on G1 cell cycle regulators in breast tumour cells. Oncogene 19 1647–1656.

    Ogiso H, Ishitani R, Nureki O, Fukai S, Yamanaka M, Kim JH, Saito K, Sakamoto A, Inoue M, Shirouzu M & Yokoyama S 2002 Crystal structure of the complex of human epidermal growth factor and receptor extracellular domains. Cell 110 775–787.

    Olayioye MA, Neve RM, Lane HA & Hynes NE 2000 The ErbB signaling network: receptor heterodimerization in development and cancer. EMBO Journal 19 3159–3167.

    Osoba D & Burchmore M 1999 Health-related quality of life in women with metastatic breast cancer treated with trastuzumab (Herceptin). Seminars in Oncology 26 (4 Suppl 12) 84–88.

    Pawson T 1995 Protein modules and signaling networks. Nature 373 573–580.

    Pawson T & Schlessinger J 1993 SH2 and SH3 domains. Current Biology 3 434–442.

    Pawson T & Scott JD 1997 Signaling through scaffold, anchoring and adaptor proteins. Science 278 2075–2080.

    Pinkas-Kramarski R, Shelly M, Glathe S, Ratzkin BJ & Yarden Y 1996 Neu differentiation factor/neuregulin isoforms activate distinct receptor combinations. Journal of Biological Chemistry 271 19029–19032.

    Pinkas-Kramarski R, Eilam R, Alroy I, Levkowitz G, Lonai P & Yarden Y 1997 Differential expression of NDF/neuregulin receptors ErbB-3 and ErbB-4 and involvement in inhibition of neuronal differentiation. Oncogene 15 2803–2815.

    Pinkas-Kramarski R, Lenferink AE, Bacus SS, Lyass L, van de Poll ML, Klapper LN, Tzahar E, Sela M, van Zoelen EJ & Yarden Y 1998 The oncogenic ErbB 2/ErbB-3 heterodimer is a surrogate receptor of the epidermal growth factor and betacellulin. Oncogene 16 1249–1258.

    Prigent SA & Gullick WJ 1994 Identification of c-erbB-3 binding sites for phosphatidylinositol 3'-kinase and SHC using an EGF receptor/c-erbB-3 chimera. EMBO Journal 13 2831–2841.

    Pupa SM, Menard S, Morelli D, Pozzi B, De Palo G & Colnaghi MI 1993 The extracellular domain of the c-erbB-2 oncoprotein is released from tumour cells by proteolytic cleavage. Oncogene 8 2917–2923.

    Ram TG & Ethier SP 1996 Phosphatidylinositol 3-kinase recruitment by p185erbB-2 and erbB-3 is potently induced by neu differentiation factor/heregulin during mitogenesis and is constitutively elevated in growth factor-independent breast carcinoma cells with c-erbB-2 gene amplification. Cell Growth and Differentiation 7 551–561.

    Robinson DR, Wu YM & Lin SF 2000 The protein tyrosine kinase family of the human genome. Oncogene 19 5548–5557.

    Rubin I & Yarden Y 2001 The basic biology of HER2. Annals of Oncology 12 (Suppl 1) S3–S8.

    Salomon DS, Brandt R, Ciardiello F & Normanno N 1995 Epidermal growth factor-related peptides and their receptors in human malignancies. Critical Reviews in Oncology/Hematology 19 183–232.

    Samanta A, LeVea CM, Dougall WC, Qian X & Greene MI 1994 Ligand and p185cneu density govern receptor interactions and tyrosine kinase activation. PNAS 91 1711–1715.

    Schlessinger J 1994 SH2/SH3 signaling proteins. Current Opinion in Genetics and Development 4 25–30.

    Schlessinger J 2000 Cell signaling by receptor tyrosine kinases. Cell 103 211–225.

    Schroff RW, Foon KA, Beatty SM, Oldham RK & Morgan AC Jr 1985 Human anti-murine immunoglobulin responses in patients receiving monoclonal antibody therapy. Cancer Research 45 879–885.

    Segatto O, King CR, Pierce JH, Di Fiore PP & Aaronson SA 1988 Different structural alterations upregulate in vitro tyrosine kinase activity and transforming potency of the erbB-2 gene. Molecular and Cellular Biology 8 5570–5574.

    Senes A, Ubarretxena-Belandia I & Engelman DM 2001 The Calpha–H...O hydrogen bond: a determinant of stability and specificity in transmembrane helix interactions. PNAS 98 9056–9061.

    Sharpe S, Barber KR & Grant CW 2000 Val(659)–> Glu mutation within the transmembrane domain of ErbB-2: effects measured by (2)H NMR in fluid phospholipid bilayers. Biochemistry 39 6572–6580.

    Sherrill JM 1997 Insufficiency of self-phosphorylation for the activation of epidermal growth factor receptor. Biochemistry 36 5677–5684.

    Sliwkowski MX, Schaefer G, Akita RW, Lofgren JA, Fitzpatrick VD, Nuijens A, Fendly BM, Cerione RA, Vandlen RL & Carraway KL 3rd 1994 Coexpression of erbB2 and erbB3 proteins reconstitutes a high affinity receptor for heregulin. Journal of Biological Chemistry 269 14661–14665.

    Smith SO, Smith CS & Bormann BJ 1996 Strong hydrogen bonding interactions involving a buried glutamic acid in the transmembrane sequence of the neu/erbB-2 receptor. Nature Structural Biology 3 252–258.

    Songyang Z, Shoelson SE, Chaudhuri M, Gish G, Pawson T, Haser WG, King F, Roberts T, Ratnofsky S, Lechleider RJ, Neel BG, Birge RB, Fajardo JE, Chou MM, Hanafusa H, Schaffhausen B & Cantley LC 1993 SH2 domains recognise specific phosphopeptide sequences. Cell 72 767–778.

    Sorkin A, Di Fiore PP & Carpenter G 1993 The carboxyl terminus of epidermal growth factor receptor/erbB-2 chimerae is internalization impaired. Oncogene 8 3021–3028.

    Stein RA & Staros JV 2000 Evolutionary analysis of the ErbB receptor and ligand families. Journal of Molecular Evolution 50 397–412.

    Sternberg MJ & Gullick WJ 1989 Neu receptor dimerization. Nature 339 587.

    Sternberg MJ & Gullick WJ 1990 A sequence motif in the transmembrane region of growth factor receptors with tyrosine kinase activity mediates dimerization. Protein Engineering 3 245–248.

    Sweeney C & Carraway KL 3rd 2000 Ligand discrimination by ErbB receptors: differential signaling through differential phosphorylation site usage. Oncogene 19 5568–5573.

    Tzahar E, Levkowitz G, Karunagaran D, Yi L, Peles E, Lavi S, Chang D, Liu N, Yayon A, Wen D & Yarden Y 1994 ErbB-3 and ErbB-4 function as the respective low and high affinity receptors of all Neu differentiation factor/heregulin isoforms. Journal of Biological Chemistry 269 25226–25233.

    Tzahar E, Waterman H, Chen X, Levkowitz G, Karunagaran D, Lavi S, Ratzkin BJ & Yarden Y 1996 A hierarchical network of inter-receptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Molecular and Cellular Biology 16 5276–5287.

    van der Geer P & Pawson T 1995 The PTB domain: a new protein module implicated in signal transduction. Trends in Biochemical Sciences 20 277–280.

    Wallasch C, Weiss FU, Niederfellner G, Jallal B, Issing W & Ullrich A 1995 Heregulin-dependent regulation of HER2/neu oncogenic signaling by heterodimerisation with HER3. EMBO Journal 14 4267–4275.

    Wang XJ, Liao HJ, Chattopadhyay A & Carpenter G 2001 EGF-dependent translocation of green fluorescent protein-tagged PLC-gamma1 to the plasma membrane and endosomes. Experimental Cell Research 267 28–36.

    White SH & Wimley WC 1999 Membrane protein folding and stability: physical principles. Annual Review of Biophysics and Biomolecular Structure 28 319–365.

    Worthylake R, Opresko LK & Wiley HS 1999 ErbB-2 amplification inhibits down-regulation and induces constitutive activation of both ErbB-2 and epidermal growth factor receptors. Journal of Biological Chemistry 274 8865–8874.

    Wulf GM, Ryo A, Wulf GG, Lee SW, Niu T, Petkova V & Lu KP 2001 Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcriptional activity of c-Jun towards cyclin D1. EMBO Journal 20 3459–7342.

    Yarden Y & Schlessinger J 1987 Epidermal growth factor induces rapid, reversible aggregation of the purified epidermal growth factor receptor. Biochemistry 26 1443–1451.

    Yu D, Jing T, Liu B, Yao J, Tan M, McDonnell TJ & Hung MC 1998 Overexpression of ErbB2 blocks Taxol-induced apoptosis by upregulation of p21Cip1, which inhibits p34Cdc2 kinase. Molecular Cell 2 581–591.

    Zabrecky JR, Lam T, McKenzie SJ & Carney W 1991 The extracellular domain of p185/neu is released from the surface of human breast carcinoma cells, SK-BR-3. Journal of Biological Chemistry 266 1716–1720.

    Zhou BP, Hu MC, Miller SA, Yu Z, Xia W, Lin SY & Hung MC 2000 HER-2/neu blocks tumour necrosis factor-induced apoptosis via the Akt/NF-kappaB pathway. Journal of Biological Chemistry 275 8027–8031.

    Zhou M, Felder S, Rubinstein M & Hurwitz DR, Ullrich A, Lax I & Schlessinger J 1993 Real time measurements of kinetics of EGF binding to soluble EGF receptor monomers and dimers support the dimerization model for receptor activation. Biochemistry 32 8193–8198.(L A Bazley and W J Gullic)