Skip to main content
Cold Spring Harbor Perspectives in Biology logoLink to Cold Spring Harbor Perspectives in Biology
. 2013 Oct;5(10):a008979. doi: 10.1101/cshperspect.a008979

Receptor Tyrosine Kinases in the Nucleus

Graham Carpenter 1,, Hong-Jun Liao 1
PMCID: PMC3783051  PMID: 24086039

Abstract

To date, 18 distinct receptor tyrosine kinases (RTKs) are reported to be trafficked from the cell surface to the nucleus in response to ligand binding or heterologous agonist exposure. In most cases, an intracellular domain (ICD) fragment of the receptor is generated at the cell surface and translocated to the nucleus, whereas for a few others the intact receptor is translocated to the nucleus. ICD fragments are generated by several mechanisms, including proteolysis, internal translation initiation, and messenger RNA (mRNA) splicing. The most prevalent mechanism is intramembrane cleavage by γ-secretase. In some cases, more than one mechanism has been reported for the nuclear localization of a specific RTK. The generation and use of RTK ICD fragments to directly communicate with the nucleus and influence gene expression parallels the production of ICD fragments by a number of non-RTK cell-surface molecules that also influence cell proliferation. This review will be focused on the individual RTKs and to a lesser extent on other growth-related cell-surface transmembrane proteins.


Some receptor tyrosine kinases (RTKs) can be trafficked from the cell surface to the nucleus, in whole or in part. This occurs in parallel with portions of some non-RTK cell-surface molecules that also affect cell growth.


The localization of receptor tyrosine kinases (RTKs) in the nucleus is perhaps 20 years old and was for some time limited by the available techniques to descriptive experiments that suffered from potential artifacts. With improved technical approaches and clearer understanding of subcellular compartments more convincing and mechanistic data have appeared. The authors have previously reviewed this topic (Carpenter and Liao 2009) and will, therefore, concentrate on more recent and mechanistic advances.

Although there are several mechanisms involved in this trafficking pathway, the role of secretase-dependent processing of cell-surface molecules to the nucleus is the most clear and convincing in the case of Notch (Bray 2006). In that case, ligand binding initiates sequential proteolytic processing by α-secretase, which removes the ectodomain, and by γ-secretase, which cleaves within the transmembrane domain of the remaining cell-associated receptor fragment to release an intracellular domain (ICD) fragment into the cytosol. The ICD subsequently escorts a transcription activation factor into the nucleus to initiate cellular responses to the ligand. The Notch scenario is recapitulated to different extents by numerous RTKs, as indicated in Table 1, and other growth-related molecules as shown in Table 2.

Table 1.

Receptor tyrosine kinase in the nucleus

RTKa Mechanism
CSF-1 γ-Secretase-generated ICD
Eph B2 γ-Secretase-generated ICD
ErbB-1 Holoreceptor, mRNA splicing: rhomboid-generated ICD
ErbB-2 Holoreceptor, CTF
ErbB-3 Holoreceptor, RNA splicing
ErbB-4 γ-Secretase-generated ICD
FGFR1 Holoreceptor, granzyme-generated ICD
FGFR2 Holoreceptor
FGFR3 γ-Secretase-generated ICD
IGF-1 Holoreceptor, ICD
Insulin ICD
Met Holoreceptor, secretase- or caspase-generated ICD
PTK7 γ-Secretase-generated ICD
Ret γ-Secretase-generated ICD
Ron γ-Secretase-generated ICD
Ryk γ-Secretase-generated ICD
Tie1 γ-Secretase-generated ICD
VEGFR1 γ-Secretase-generated ICD, holoreceptor
VEGFR2 γ-Secretase-generated ICD, holoreceptor

ICD, intracellular domain; CTF, carboxy-terminal fragment.

aReferences can be found in the text or in Carpenter and Liao (2009).

Table 2.

Growth-related cell-surface molecules in the nucleus

Molecules Mechanism Referencesa
ADAM10 γ-Secretase-generated ICD Tousseyn et al. 2009
ADAM13 γ-Secretase-generated ICD Cousin et al. 2011
CD44 γ-Secretase-generated ICD Janiszewska et al. 2010; Miletti-González et al. 2012
EpCAM γ-Secretase-generated ICD Denzel et al. 2009; Maetzel et al. 2009; Ralhan et al. 2010; Huang et al. 2011b; Chaves-Pérez et al. 2013
Frizzled γ-Secretase-generated ICD Mathew et al. 2005
Growth hormone receptor γ-Secretase-generated ICD Wang et al. 2011a
IL-2R γ-Secretase-generated ICD Montes de Oca et al. 2010
Klotho γ-Secretase-generated ICD Bloch et al. 2009
LRP1 γ-Secretase-generated ICD Rebeck 2009
Neogenin (DDC) γ-Secretase-generated ICD Goldschneider et al. 2008; Bai et al. 2011
p75NTR γ-Secretase-generated ICD Ceni et al. 2010; Parkhurst et al. 2010; Wang et al. 2010a; Le Moan et al. 2011
Patched γ-Secretase-generated ICD Kagawa et al. 2011
Pre-β-cellulin γ-Secretase-generated ICD Stoeck et al. 2010
Protocadherin γ-Secretase-generated ICD Buchanan et al. 2010
PTPμ γ-Secretase-generated ICD Burgoyne et al. 2009
TβRI γ-Secretase-generated ICD, holoreceptor Mu et al. 2011; Chandra et al. 2012
Ephrin B2 γ-Secretase-generated ICD Tomita et al. 2006; Georgakopoulos et al. 2011
Notch γ-Secretase-generated ICD Fortini 2009

aEarlier references can be found in Carpenter and Liao (2009).

In reviewing intramembraneous cleavage of RTKs, two points are emphasized. First, is the cleavage process or nuclear localization of the ICD fragment stimulated by a ligand? Although a number of receptors and other cell-surface molecules are cleaved following the addition of protein kinase C agonists, such as phorbol esters, it remains to be shown in those cases whether natural receptor ligands also influence receptor trafficking to the nucleus. Second, what is the evidence that the released ICD fragment produces a relevant biologic activity? These issues are important as it has been hypothesized that secretase processing of transmembrane proteins may be a cellular housekeeping mechanism to degrade these molecules, as the presence of a transmembrane domain presents a barrier to other proteolytic systems (Small 2002; Kopan and Ilagan 2004). These are not, however, necessarily mutually exclusive interpretations. For example, α- or β-secretase release of an ectodomain fragment may be biologically important, whereas the γ-secretase degradation of the remaining cell-associated fragment may proceed as a housekeeping function. However, when the cleavage is stimulated by a ligand, especially the cognate ligand, then it seems very likely that the nuclear trafficking also represents a signal transduction mechanism. Although the nuclear function(s) of ICD fragments is unknown in many cases, the lessons of ErbB-4 and Notch indicate that ICDs may activate transcription by transporting transcription factors from the cytoplasm to the nucleus.

SECRETASE PROCESSING OF RTKs

ErbB-4 Receptor

Within the family of RTKs that are processed to the nucleus, ErbB-4 is the most completely understood in terms of mechanism and biological function and will be discussed in more detail as a model for the secretase cleavage of other RTKs (Fig. 1). Ectodomain proteolytic processing of ErbB-4 includes a basal level, which can be increased by 12-O-tetradecanoylphorbol-13-acetate (TPA) in all cells or by the addition of its cognate ligand, neuregulin (heregulin), to certain cells (Vecchi et al. 1996; Zhou and Carpenter 2000). This cleavage results in the formation of two receptor fragments: a 120-kDa ectodomain fragment that is released into the media and an 80-kDa membrane-bound fragment, termed m80 or CTF (carboxy-terminal fragment). Cleavage requires ADAM 17 (TACE) and it is likely this is the enzyme that executes cleavage of ErbB-4 between His651 and Ser652 within the extracellular stalk or ecto-juxtamembrane region (Rio et al. 2000; Cheng et al. 2003). Hence, the m80 fragment includes eight ectodomain residues, the transmembrane domain, and entire ICD.

Figure 1.

Figure 1.

Depicted is the general mechanism for generation and nuclear localization of RTK ICD fragments. Included are examples of canonical signal transduction pathways to the nucleus (i.e., MAPK and STAT pathways) as contrasted to the noncanonical ICD mechanism.

Sensitivity to ectodomain shedding is likely determined, at least in part, by the length of the stalk region as shown for the selectins (Migaki et al. 1995). There are two ErbB-4 isoforms termed Jm-a, in which the ectodomain is sensitive to cleavage, and Jm-b, which is not cleavable (Elenius et al.1997). The presence of these two isoforms is unique to ErbB-4. Because ADAM-mediated cleavage events do not involve a defined sequence or cleavage site in the substrate, it seems that longer stalk regions in substrates may simply permit accessibility of the protease. Interestingly, the stalk region in Jm-b is much shorter (six residues) than the corresponding region of Jm-a (16 residues), and ErbB-1, -2, and -3 also have relatively short stalk regions (6–9 residues) and are not subject to a significant level of metalloprotease-mediated ectodomain cleavage (Vecchi et al. 1996). Hence the unique sensitivity of the Jm-a ErbB-4 isoform to secretase-dependent processing and signaling seems likely owing to the length of its stalk region.

It seems probable that the shed ErbB-4 ectodomain may function to block receptor activation by binding neuregulin. The function of the m80 fragment, however, is known. The capacity of γ-secretase to cleave substrates requires that the substrate have a short ectodomain region of 50 or fewer residues (Struhl and Adachi 2000). Hence, the ADAM-mediated removal of a large portion of the ErbB-4 ectodomain is a prerequisite step for subsequent γ-secretase cleavage of the m80 fragment (Ni et al. 2001).

γ-Secretase is a complex of at least four distinct transmembrane proteins of which presenilin is the catalytic protease (Selkoe and Wolfe 2007). The nicastrin subunit of this complex recognizes transmembrane proteins with shortened or nublike ectodomains and thereby acts as a targeting subunit for intramembrane cleavage by presenilin (Shah et al. 2005). Presenilin activity converts the ErbB-4 m80 fragment to a soluble s80 or ICD fragment that is found in the cytoplasm, nucleus, and mitochondria (Ni et al 2001; Naresh et al. 2006). Recently, a fifth protein (GASP) has been added to the γ-secretase complex (He et al. 2010). GASP, which was first identified as a target of the tyrosine kinase inhibitor imatnib, is a 16 kDa protein that is required for efficient γ-secretase cleavage of the cell-surface protein APP. This raises the issue of whether a similar targeting protein may participate in the cleavage of other γ-secretase substrates.

The carboxyl terminus of ErbB-4 encodes a PDZ domain recognition motif, which is required for presenilin cleavage of the m80 fragment (Ni et al. 2003). Deletion of this motif (TVV) does not influence ectodomain cleavage, but does attenuate presenilin association with the m80 fragment and production of the ICD fragment. Presenilin also contains a PDZ domain recognition motif, and it is possible that a scaffold of PDZ domain-containing proteins may be required for γ-secretase cleavage.

Presenilin cleavage of substrates occurs within the transmembrane domain and, based on APP and Notch processing, this may occur at multiple sites, producing several species of ICD fragments that may have differing levels of metabolic stability based on the N-end rule (Varshavsky 1997). Mutation within the transmembrane domain, which is often used to identify a cleavage site based on the lack of detectable ICD formation, can alter the metabolic stability of the ICD fragments. This is shown in the case of Notch where a transmembrane mutation appears to prevent cleavage, but actually results in a new ICD fragment that is very rapidly degraded owing to the presence of a metabolically destabilizing amino-terminal residue (Tagami et al. 2008).

It has been reported that the Val675Ala (Muraoka-Cook et al. 2006) or Val673Ile (Vidal et al. 2005) mutations within the ErbB-4 transmembrane domain abrogate γ-secretase cleavage, as judged by the inability to detect the ICD fragment. In view of the Notch mutagenesis data, it is not clear whether these mutations actually prevent cleavage or result in a less stable ICD fragment. Given the low level of ICD fragment normally detectable, a modest change in stability may render the fragment undetectable by the same methodology. Additional splice variants of ErbB-4 (Veikkolainen et al. 2011) produce two isoforms of the ICD fragment, CYT-1 and CYT-2. The two isoforms differ by 16 consecutive residues that are present in CYT-1, but absent in the CYT-2, and provide the CYT-1 isoform with overlapping docking sites for PI3 kinase and proteins containing WW domains. In a comparative study using Madin-Darby canine kidney (MDCK) cells, one group has shown that the CYT-1 ICD is less metabolically stable than the CYT-2 ICD (Zeng et al. 2009) and that the CYT-2 ICD is more readily detected in the nucleus (Zeng et al. 2007). These differences were due, at least in part, to association of the ubiquitin ligase Need 4, a WW-domain-containing protein, with selective sequence motifs present in the CYT-1 isoform. Using different cell backgrounds, similar results have been reported by others (Omerovic et al. 2007; Feng et al. 2009).

In terms of the physiological relevance, it is now clear that endogenous generation of the ErbB-4 ICD by γ-secretase is required for control of astrogenesis in the developing mouse (Sardi et al. 2006). In this system, the ICD fragment interacts with TAB2, an adaptor protein, and thereby with N-CoR, a corepressor, and chaperones this complex into the nucleus. A similar chaperone mechanism between the ErbB-4 ICD and STAT5 has been proposed to be operative during mammary differentiation (Williams et al. 2004). Also, the nuclear interaction of the ErbB-4 ICD with HIF-1α is reported to attenuate metabolic degradation of HIF-1α and thereby promote gene expression in mammary tissue (Paatero et al. 2012).

It is clear that ErbB-4 is functionally involved in mammary development in the animal (Jones 2008). ErbB-4 nuclear localization has been observed in normal and tumor mammary tissue and exogenous ICD expression provokes differentiation events (Muraoka-Cook et al. 2006). However, as a cautionary note studies using exogenous expression of the Notch ICD have shown that different levels of expression can provoke distinct biologic responses (Mazzone et al. 2010; Han et al. 2011). Nevertheless, analysis of an ErbB-4 noncleavable mutant (Rokicki et al. 2010) and the intracellular distribution of the ICD fragment in breast tumor tissue (Thor et al. 2009) are consistent with the conclusion that endogenous ErbB-4 cleavage is physiologically relevant to differentiation in this tissue and a positive factor for breast cancer patients.

Also, consistent with a role of the ErbB-4 ICD fragment in various differentiation systems is the report that γ-secretase inhibition prevents neuregulin generation of the ErbB-4 ICD in oligodendrocytes and maturation of this cell type (Lai and Feng 2004). Interestingly, there is a genetic correlation between ErbB-4 and schizophrenia (Pan et al. 2011) and the involvement of the ICD fragment in controlling gene expression in relevant cell types has been reported (Wong and Weickert 2009; Allison et al. 2011). Also, the maturation of fetal lung cells is reported to be dependent on secretase processing of ErbB-4 and ICD association with the transcription factors YAP (Hoeing et al. 2011), STAT 5a (Zscheppang et al. 2011a), and ERβ (Zscheppang et al. 2011b). It is suggested that these interactions allow ErbB-4 to regulate transcription of surfactant protein B and thereby promote lung maturation.

As mentioned above, the ErbB-4 ICD also has been localized in mitochondria and in that location may function as a proapoptotic protein. This is based on the capacity of the ICD to induce cell death, the presence of a BH3 domain in the ICD, the loss of apoptotic capacity following mutagenesis of this domain, and detection of an interaction with the antiapoptotic protein BCL-2, which, when overexpressed, abrogated ICD-induced cell death (Naresh et al. 2006).

Ephrin-B2

Addition of the ligand ephrin-B2 (Eph-B2) provokes the secretase cleavage of the EphB2 receptor releasing ectodomain and ICD fragments (Litterst et al. 2007; Lin et al. 2008). The cleavage events are also stimulated by ionomycin or by activation of the N-methyl-d-aspartate (NMDA) receptor, agents that mediate Ca2+ influx into cells. In this system, ephrin-mediated cleavage events require endocytosis, whereas cleavage mediated by ionomycin or NMDA receptor activation occurs on the cell surface. A similar endocytosis relationship between neuregulin- or TPA-mediated cleavage of ErbB-4 was noted (Zhou and Carpenter 2000). To date, it is unclear whether the Eph receptor ICD fragment is translocated from the cytoplasm to another organelle and there is no data related to its physiological function in mediating ligand responsiveness.

Colony Stimulating Factor-1 Receptor

Secretase cleavage of the colony-stimulating factor-1 receptor (CSF-1R) can be stimulated by CSF-1, LPS, or TPA (Wilhelmsen and van der Geer 2004; Glenn and van der Geer 2007, 2008). Lipopolysaccharide (LPS) is a ligand for the Toll4 receptor and agonists for other Toll-like receptors also stimulate cleavage of CSF-1R. This heterologous stimulation of CSF-1R cleavage may be related to the fact that in macrophages both receptor systems are thought to be involved in producing innate immune responses. Although the CSF-1 ICD fragment does appear in both cytoplasm and nucleus, a physiologic function has not been identified.

Vascular Endothelial Growth Factor-1

Pigment epithelium-derived factor (PEDF) binds to an unknown receptor and promotes an antiangiogenic response that can oppose the capacity of vascular endothelial growth factor (VEGF) to promote endothelial cell proliferation. The addition of PEDF to endothelial cells promotes the γ-secretase-mediated cleavage of VEGFR1 (Flt1) with release of its ICD fragment (Cai et al. 2006, 2011a; Rahimi et al. 2009). The ICD fragment is only present when cells are treated simultaneously with PEDF and VEGF, and the fragment was detected in the cytoplasm, but not in the nucleus. In this system, the intact VEGFR1 molecule is found in the nucleus following the addition of VEGF (see below) and PEDF reduces VEGF-induced angiogenesis and the nuclear level of intact VEGFR in a manner dependent on γ-secretase activity. This implies that the PEDF-stimulated production of the VEGFR1 ICD fragment negatively regulates intact VEGFR1 levels in the nucleus and VEGF-induced angiogenesis. Based on sensitivity to γ-secretase inhibitors, it is reported that the capacity of PEDF to prevent VEGF-induced changes in vascular permeability requires production of the VEGFR1 ICD fragment (Cai et al. 2011b). In contrast, Ablonczy et al. (2009) report that it is VEGFR2 that is cleaved by γ-secretase in this system.

Tie 1

Tie 1 is an orphan receptor that forms a hetero-oligomeric complex with Tie 2, the receptor for angiopoietin 1 (Ang 1). Addition of Ang 1 activates Tie 2 and provokes tyrosine phosphorylation of Tie 1. Ectodomain cleavage of Tie 1 is stimulated by a variety of agents (TPA, VEGF, TNFα, sheer stress) and increases Ang 1 activation of Tie 2, apparently by allowing greater access of the ligand to its Tie 2 binding site. Following ectodomain release, the Tie 1 cell-associated 45-kDa cleavage fragment is processed by γ-secretase to produce a 42-kDa cytoplasmic ICD fragment (Marron et al. 2007). In this receptor system, the ectodomain secretase action is physiologically important; however, the significance of γ-secretase activity may be simply to remove the highly tyrosine phosphorylated 45 kDa fragment. Although the addition of Ang 1 promotes rapid endocytosis and degradation of Tie 2, Tie 1 is not cleared from the cell surface by this same route. Therefore, a secretase mechanism may provide the means by which Ang1-phosphorylated Tie 1 is inactivated.

Insulin-like Growth Factor-1 and Insulin Receptors

In preliminary reports, it has been shown that the insulin and IGF-1 receptors can be cleavaged by secretase action to produce ICD fragments (Kasuga et al. 2007; McElroy et al. 2007). However, although TPA stimulated formation of the ICD fragments neither cognate ligand was shown to do so.

Ryk

Wnt proliferative signaling is mainly mediated by Frizzled receptors and low-density lipoprotein-related proteins (LRP) through a canonical pathway involving β-catenin and T-cell factor (TCF). However, it is known that Ryk, a receptor tyrosine kinase that lacks kinase activity, binds Wnt and participates in Wnt signaling in certain biologic contexts. Evidence has appeared demonstrating that γ-secretase cleavage of Ryk is required for Wnt 3-dependent neuronal differentiation (Lyu et al. 2008). Release of the Ryk ICD fragment does not require Wnt, but nuclear translocation of the ICD does require Wnt. Because the Ryk ICD lacks a nuclear localization signal, the mechanism for nuclear translocation is unclear. Cdc 37, a cochaperone of Hsp 90, interacts with the Ryk ICD, attenuates metabolic degradation, and promotes nuclear ICD levels (Lyu et al. 2009). Because this interaction has not been shown to be a result of Wnt signaling, the mechanism by which Wnt promotes nuclear translocation of the ICD fragment is not known. It is interesting to note that in Drosophila, Wnt binding induces the γ-secretase cleavage of D Frizzled, a heptahelical receptor, with nuclear localization of a CTF (Mathew et al. 2005). Also, the RTKs Ror1/2 participate in signaling by certain Wnt ligands (Grumolato et al. 2010), but it is not known whether ICD fragments are produced.

Fibroblast Growth Factor 3

Activation of the fibroblast growth factor receptor 3 (FGFR3) by FGF-1 leads to an ErbB-4-like cleavage pathway (Degnin et al. 2011). Both FGFR3 and ErbB-4 can undergo regulated intramembrane proteolysis following addition of the homologous ligand or TPA, but in the former case autophosphorylation and endocytosis are required. Although internalized ErbB-4 is first cleaved by a metalloprotease, ectodomain cleavage of FGFR3 is catalyzed by a cathepsin protease. Subsequently, γ-secretase cleavage liberates a FGFR3 ICD fragment that translocates to the nucleus.

Met

Ligand-independent constitutive γ-secretase cleavage of Met in several cell types has been described (Foveau et al. 2009). Interestingly, an inhibitory antibody to Met (DN30), which induces receptor degradation and biologic activity of overexpressed Met, promotes the formation of Met ICD. The intracellular localization of this ICD fragment, however, was not reported.

Protein Tyrosine Kinase 7

Protein tyrosine kinase 7 (PRK7), a pseudokinase, is constitutively cleaved by ADAM metalloprotease and γ-secretase to produce an ICD fragment that can be found in the nucleus (Golubkov and Strongin 2012; Na et al. 2012). In these studies, exogenous expression of the ICD fragment promoted cell proliferation, migration, and colony formation. Although PTK7 functions as a coreceptor in the Wnt pathway, it has been implicated in a broad range of biologic processes. Whether the PTK7 ICD fragment has a role in these events is not known.

NONSECRETASE FORMATION OF RTK ICD FRAGMENTS

Caspase-Dependent Fragments

In the case of several RTKs (ErbB-2 [Tikhomirov and Carpenter 2001; Benoit et al. 2004; Strohecker et al. 2008], Ret [Bordeaux et al. 2000; Cabrera et al. 2011], ALK [Mourali et al. 2006; Racaud-Sultan et al. 2006], TrkC [Tauszig-Delamasure et al. 2007], and Met [Tulasne et al. 2004; Pozner-Moulis et al. 2006; Foveau et al. 2007; Deheuninck et al. 2009]) there is evidence that caspase activity cleaves the cytoplasmic domain to produce an ICD fragment. Because the fragment is often produced by two cleavage events within the cytoplasmic domain, the fragment is often considerably smaller than that produced by intramembrane proteolysis. In no reported case are these caspase proteolytic events stimulated by ligand binding or by TPA, and in some studies the presence of the cognate ligand prevents cleavage. The formation of caspase ICD fragments is functionally associated with the induction of apoptosis and in one instance (Strohecker et al. 2008) the fragment has been localized to the mitochondria.

Translation-Dependent Fragments

Metalloprotease activity (Codony-Servat et al. 1999) or internal translation initiation of ErbB-2 mRNA (Anido et al. 2006) leads to the production of ErbB-2 CTFs. The CTFs, which are generated at three methionine residues located on either side of the transmembrane domain, are found in the nucleus (Anido et al. 2006; Xia et al. 2011). These fragments promote cell migration (García-Castillo et al. 2009) and are reported to be oncogenic (Pedersen et al. 2009). Because two of the CTFs include the transmembrane domain, a mechanism for translocation of these CTFs to the nucleus is not clear. ErbB-2 is a major therapeutic target in breast cancer and the therapeutic agent used is an antibody to the ectodomain. Therefore the presence of these biologically active CTFs in tumor tissue represents a potential therapeutic problem (Arribas et al. 2011).

Splicing-Dependent Fragments

Another group has identified a spliced product of the epidermal growth factor (EGF) receptor (ErbB-1) that represents the splicing of exon 1 of the ectodomain to exon 23 of the cytoplasmic domain (Piccione et al. 2012). Hence, sequences required for ligand binding and tyrosine kinase activity are deleted. This variant, termed mLEEK, is constitutively produced in a variety of cell lines and tumor tissues, is present in the nucleus, and displays transcriptional activity. Because mLEEK retains a signal sequence and may be secreted, it remains to be shown how nuclear localization is achieved. Splicing of ErbB-3 produces at least 14 variants and a few of these encode sequences corresponding to an ICD. One of these encodes an ICD sequence encoding a protein 80 kDa, which is localized to the nucleus (Andrique et al. 2012). Data have been presented to implicate this protein in the regulation of cyclin D1 transcription. Another nuclear ErbB-3 variant has been reported to associate with active promoters in Schwann cells (Adilakshmi et al. 2011).

Granzyme-Dependent Fragments

The generation of an ICD fragment from FGFR1 in an FGF-dependent manner has been reported (Chioni and Grose 2012). The ICD fragment is localized to the nucleus and evidence is presented to argue that this fragment increases cell migration. Unexplained, however, was the mechanism by which the ICD fragment could be generated by granzyme B, a serine protease localized within cytoplasmic granules.

Rhomboid-Dependent Fragments

Rhomboids are intramembrane serine proteases that are best known for the regulation of the ectodomain shedding of transmembrane proteins, such as the precursor for TGF-α. The intramembrane cleavage of ErbB-1 (EGF receptor) by rhomboid activity has now been reported (Liao and Carpenter 2012). This cleavage is EGF-stimulated and produces an ICD-like fragment that is localized to membrane and nuclear fractions, but is not found in the cytosol. Because rhomboid cleavage occurs close to the ectodomain side of the transmembrane domain, this fragment may retain transmembrane domain residues at its amino terminus and therefore remain membrane associated.

INTACT RECEPTORS IN THE NUCLEUS

Listed in Table 1 are several receptors for which the data show that the intact or holoreceptor is present in the nucleus. That the nuclear species is the holoreceptor and not an ICD fragment is based on a nuclear fraction Mr value indicative of the mature receptor, which is significantly distinct from the size of an ICD fragment. Because cell fractionation can produce contamination of organelle preparations, this is a source of concern when nuclear extracts are used for size analysis. However, most localization studies are supplemented with imaging data of intact cells. In a few instances the data relies on immunohistochemistry alone and in those cases it is not clear that intact receptor is distinguishable from an ICD fragment unless both ectodomain and cytoplasmic domain antibodies are used. In nearly all cases, however, it does appear that the receptor is present in the nucleoplasm and not the nuclear envelope.

ErbB-1

The capacity of EGF to induce trafficking of the intact EGF receptor (ErbB-1) to the nucleus was first reported in 2001 and a nuclear target was identified (Lin et al. 2001). The nuclear receptor is reported to recognize the promoter of cyclin D1 and to transactivate this promoter in a reporter system. Other promoters are also reported to be recognized by the EGF receptor (Lo et al. 2005, 2010; Tao et al. 2005; Hanada et al. 2006). However, direct and specific binding to any promoter remains to be shown. Also, the nuclear receptor associates with proliferating cell nuclear antigen (PCNA) in the nucleus, modifying its stability (Wang et al. 2006), and with polynucleotide phosphorylase, increasing cellular radioresistance (Yu et al. 2012).

Trafficking from the cell surface to the nucleus requires receptor internalization through clathrin-coated pits (De Angelis Campos et al. 2011); retrograde transport from the Golgi to the endoplasmic reticulum (ER) (Wang et al. 2011b); phosphorylation of Ser 229, an ectodomain residue, by Akt (Huang et al. 2011a); phosphorylation of Thr 654 (Dittmann et al. 2010), a known cytoplasmic domain protein kinase C (PKC) phosphorylation site; and function of the lipid kinase PIKfyve (Kim et al. 2007). Importin β is required for ErbB-1 nuclear entry (Lo et al. 2006) and a nuclear localization sequence to facilitate this interaction is present in the cytoplasmic juxtamembrane region of the EGF receptor (Hsu and Hung 2007). The nuclear receptor has been identified by biochemical fractionation, immunohistochemistry, and live cell imaging methods and shown to be in a nonmembranous nuclear environment, based on its extractability with a high salt aqueous buffer.

The above trafficking steps, perhaps with the exception of Ser 229 phosphorylation, are reasonable given what is known about intracellular trafficking in general and more specifically about other molecules (e.g., bacterial toxins) (Sandvig and van Deurs 2002; Bonifacino and Rojas 2006), that are trafficked from the cell surface to the ER. The toxins are soluble molecules and after reaching the ER are translocated to the cytoplasm where they exert their biological activity. Therefore, a mechanism is required to facilitate movement of the membrane-bound EGF receptor in the ER to the nucleus as a nonmembrane localized molecule. Although no endocytic trafficking system was known to extract a transmembrane receptor from its lipid bilayer, it was suggested that an ER protein translocon could provide such a step (Carpenter 2003). The Sec61 translocon located in the ER is known to mediate the trafficking of extracellular toxins from the cell surface to the cytoplasm and, as part of the ERAD pathway, to retrotranslocate malfolded transmembrane proteins in the ER to the cytoplasm. Subsequent experiments showed that EGF induced trafficking of the EGF receptor to the ER where it interacted with the Sec61 translocon that mediated receptor retrotranslocation to the cytoplasm and import into the nucleus (Liao and Carpenter 2007). Experimental data showed that knockdown of a Sec61 subunit attenuated both EGF nuclear localization of the EGF receptor and EGF induction of cyclin D1. This trafficking pathway is depicted in Figure 2. A subsequent report has confirmed the role of the Sec61 translocon in this pathway, but places the interaction between the EGF receptor and the translocon at the inner nuclear membrane (Wang et al. 2010b, 2012).

Figure 2.

Figure 2.

Illustrated is the Sec61-dependent trafficking pathway for intact RTKs, such as the EGF receptor, to translocate from the cell surface to the nucleus.

Additional reports have shown that nuclear localization of the EGF receptor is provoked by radiation (Dittmann et al. 2005; Liccardi et al. 2011) or treatment with the receptor antibody C225/Cetuximab (Liao and Carpenter 2009). The nuclear receptor is reported to interact with MUC1 and increase the level of chromatin-bound EGF receptor (Bitler et al. 2010; Merlin et al. 2011). MUC1 is a cell-surface protein that is known to interact with the receptor and promote receptor internalization. It is also known to be trafficked to the nucleus (Carson 2008).

ErbB-2 and ErbB-3

Using methodologies similar to those used to localize the EGF receptor to the nucleus (Lin et al. 2001), the same group showed that the intact ErbB-2 molecule translocated from the cell surface to the nucleus and thereby facilitates the expression of the Cox-2 mRNA (Wang et al. 2004; Giri et al. 2005) and ribosomal RNA (Li et al. 2011). The mechanism of ErbB-2 trafficking to the nucleus is reported to include a Sec61-dependent step analogous to that of ErbB-1 (Wang et al. 2012). Because ErbB-2 does not bind a known ligand, the studies represent a constitutive pathway. Progesterone, however, transactivates ErbB-2 and thereby initiates ErbB-2 translocation to the nucleus (Béguelin et al. 2010). This translocation of ErbB-2 chaperones STAT3 into the nucleus where the complex, together with the progesterone receptor, interacts with the cyclin D promoter to bring about progesterone-induced cyclin D expression. Less is known about the nuclear form of ErbB-3 (Offterdinger et al. 2002).

IGF-1

An initial report of intact IGF-1 receptors present in the nucleus (Chen and Roy 1996) has been confirmed and extended in recent reports from two groups (Aleksic et al. 2010; Deng et al. 2010; Sehat et al. 2010). Both groups indicate that this relocalization from plasma membrane to nucleus is IGF-1 dependent and that the nuclear receptor may function as a transcriptional coactivator. One report showed that nuclear localization and the capacity to increase transcription in reporter assays require sumoylation at three lysine residues in the β chain of the receptor. Chromatin-immunoprecipitation (ChIP) assays indicated the nuclear IGF-1R interacts with DNA sequences identified as predominately intergenic, with characteristics of enhancers and having the capacity to increase luciferase transcription in reporter assays.

Ron

Nuclear localization of the Ron RTK is reported to occur in bladder cancer cells and is not stimulatable by the Ron ligand MSP (Liu et al. 2010). Rather nuclear levels of Ron are increased during serum starvation. Interestingly, the increase in nuclear Ron requires the EGF receptor as a heterodimerization partner. ChIP assays have identified potential gene targets, including a number of stress-responsive genes.

Met

Nuclear localization of Met ICD fragments produced by either secretase or caspase cleavage are discussed above. One group has reported that the intact Met receptor is very rapidly translocated from the cell surface to the nucleus following the addition of HGF, and this translocation event is required for HGF induction of a nuclear Ca2+ signal (Gomes et al. 2008).

FGFR1 and 2

There is a large background of published reports describing the ligand-dependent translocation of intact FGFR1 to the nucleus (reviewed in Stachowiak et al 2007). There is a report detailing the dynamics of nuclear FGFR1 movement among receptor populations having different motilities taken to represent free receptor or receptor associated with chromatin or nuclear matrix (Dunham-Ems et al. 2009). The interaction of FGFR1 and the transcription factor Nurr 1 in the nucleus of dopaminergic neurons has been described (Baron et al. 2012). Morphologic studies have recognized FGFR2 in the nucleus of tissue samples (Lu et al. 2004; Schmahl et al. 2004; Giulianelli et al. 2008; Martin et al. 2011), but without analysis of the size it is not possible to distinguish whether this represents an intact receptor or a receptor fragment.

VEGFR1 and R2

Two reports have identified intact VEGFR1 in the nucleus and one indicates that the localization is increased by ligand (Cai et al. 2006; Lee et al. 2007). Several studies have found that intact VEGFR2 (KDR) is localized in the nucleus and in some reports this is ligand sensitive (Stewart et al. 2003; Fox et al. 2004; Santos and Dias 2004; Zhang et al. 2005; Santos et al. 2007). Domingues et al. (2001) conclude that nuclear VEGFR2 regulates its own transcription.

NUCLEAR LOCALIZATION OF OTHER GROWTH-REGULATING RECEPTORS

Although Notch is an obvious example of a cell-surface receptor that uses nuclear localization of the receptor to affect its biologic activity, there are a number of other examples for growth-related cell-surface proteins and these are presented in Table 2. This list is limited to recent additions; a more comprehensive list was previously published and should be consulted for earlier references (Carpenter and Liao 2009).

CONCLUDING REMARKS

The role of regulated intramembrane proteolysis in signal transduction by activated RTKs is reasonably well established biochemically and biologically. One open question revolves around the role of tyrosine kinase activity within the nuclear compartment. Are there nuclear substrates or does kinase activity not have a role in ICD function? A second issue is whether any ICD, regardless of its source, actually forms a direct and biologically important contact with DNA. To date, there is no convincing data to resolve either of those questions.

It might be important to note that the ligand-dependent trafficking of any RTK or its ICD to the nucleus would represent a noncanonical signaling pathway for that particular ligand. This would be unique as other signaling elements are canonical (i.e., common to multiple ligand–receptor systems).

Last, the reader is referred to other recent reviews on this topic (Ancot et al. 2009; Borlido et al. 2009; McCarthy et al. 2009; De Strooper and Annaert 2010; López-Otin and Hunter 2010; Wang et al. 2010c; Lal and Caplan 2011; Lemberg 2011).

ACKNOWLEDGMENT

The authors acknowledge support of the National Institutes of Health (grant RO1 CA 125649).

Footnotes

Editors: Joseph Schlessinger and Mark A. Lemmon

Additional Perspectives on Signaling by Receptor Tyrosine Kinases available at www.cshperspectives.org

REFERENCES

  1. Ablonczy Z, Prakasam A, Fant J, Fauq A, Crosson C, Sambamurti K 2009. Pigment epithelium-derived factor maintains retinal pigment epithelium function by inhibiting vascular endothelial growth factor-R2 signaling through γ-secretase. J Biol Chem 284: 30177–30286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Adilakshmi T, Ness-Myers J, Madrid-Aliste C, Fiser A, Tapinos N 2011. A nuclear variant of ErbB3 receptor tyrosine kinase regulates ezrin distribution and Schwann cell myelination. J Neurosci 31: 5106–5119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Aleksic T, Chitnis MM, Perestenko OV, Gao S, Thomas PH, Turner GD, Protheroe AS, Howarth M, Macaulay VM 2010. Type 1 insulin-like growth factor receptor translocates to the nucleus of human tumor cells. Cancer Res 70: 6412–6419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Allison JG, Das PM, Ma J, Inglis FM, Jones FE 2011. The ERBB4 intracellular domain (4ICD) regulates NRG1-induced gene expression in hippocampal neurons. Neurosci Res 70: 155–163 [DOI] [PubMed] [Google Scholar]
  5. Ancot F, Foveau B, Lefebvre J, Leroy C, Tulasne D 2009. Proteolytic cleavages give receptor tyrosine kinases the gift of ubiquity. Oncogene 28: 2185–2195 [DOI] [PubMed] [Google Scholar]
  6. Andrique L, Fauvin D, El Maassarani M, Colasson H, Vannier B, Séité P 2012. ErbB3(80 kDa), a nuclear variant of the ErbB3 receptor, binds to the Cyclin D1promoter to activate cell proliferation but is negatively controlled by p14ARF. Cell Signal 24: 1074–1085 [DOI] [PubMed] [Google Scholar]
  7. Anido J, Scaltriti M, Bech Serra JJ, Santiago Josefat B, Todo FR, Baselga J, Arribas J 2006. Biosynthesis of tumorigenic HER2 C-terminal fragments by alternative initiation of translation. EMBO J 25: 3234–3244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Arribas J, Baselga J, Pedersen K, Parra-Palau JL 2011. p95HER2 and breast cancer. Cancer Res 71: 1515–1519 [DOI] [PubMed] [Google Scholar]
  9. Bai G, Chivatakarn O, Bonanomi D, Lettieri K, Franco L, Xia C, Stein E, Ma L, Lewcock JW, Pfaff SL 2011. Presenilin-dependent receptor processing is required for axon guidance. Cell 144: 106–118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Baron O, Förthmann B, Lee YW, Terranova C, Ratzka A, Stachowiak EK, Grothe C, Claus P, Stachowiak MK 2012. Cooperation of nuclear fibroblast growth factor receptor 1 and Nurr1 offers new interactive mechanism in postmitotic development of mesencephalic dopaminergic neurons. J Biol Chem 287: 19827–19840 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Béguelin W, Díaz Flaqué MC, Proietti CJ, Cayrol F, Rivas MA, Tkach M, Rosemblit C, Tocci JM, Charreau EH, et al. 2010. Progesterone receptor induces ErbB-2 nuclear translocation to promote breast cancer growth via a novel transcriptional effect: ErbB-2 function as a coactivator of Stat3. Mol Cell Biol 30: 5456–5472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Benoit V, Chariot A, Delacroix L, Deregowski V, Jacobs N, Merville MP, Bours V 2004. Caspase-8-dependent HER-2 cleavage in response to tumor necrosis factor α stimulation is counteracted by nuclear factor κB through c-FLIP-L expression. Cancer Res 64: 2684–2691 [DOI] [PubMed] [Google Scholar]
  13. Bitler BG, Goverdhan A, Schroeder JA 2010. MUC1 regulates nuclear localization and function of the epidermal growth factor receptor. J Cell Sci 123: 1716–1723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bloch L, Sineshchekova O, Reichenbach D, Reiss K, Saftig P, Kuro-o M, Kaether C 2009. Klotho is a substrate for α-, β- and γ-secretase. FEBS Lett 583: 3221–3224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bonifacino JS, Rojas R 2006. Retrograde transport from endosomes to the trans-Golgi network. Nat Rev Cell Mol Biol 7: 563–579 [DOI] [PubMed] [Google Scholar]
  16. Bordeaux MC, Forcet C, Granger L, Corset V, Bidaud C, Billaud M, Bredesen DE, Edery P, Mehlen P 2000. The RET proto-oncogene induces apoptosis: A novel mechanism for Hirschsprung disease. EMBO J 19: 4056–4063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Borlido J, Zecchini V, Mills IG 2009. Nuclear trafficking and functions of endocytic proteins implicated in oncogenesis. Traffic 10: 1209–1220 [DOI] [PubMed] [Google Scholar]
  18. Bray SJ 2006. Notch signalling: A simple pathway becomes complex. Nat Rev Mol Cell Biol 7: 678–689 [DOI] [PubMed] [Google Scholar]
  19. Buchanan SM, Schalm SS, Maniatis T 2010. Proteolytic processing of protocadherin proteins requires endocytosis. Proc Natl Acad Sci 107: 17774–17779 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Burgoyne AM, Phillips-Mason PJ, Burden-Gulley SM, Robinson S, Sloan AE, Miller RH, Brady-Kalnay SM 2009. Proteolytic cleavage of protein tyrosine phosphatase μ regulates glioblastoma cell migration. Cancer Res 69: 6960–6968 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Cabrera JR, Bouzas-Rodriguez J, Tauszig-Delamasure S, Mehlen P 2011. RET modulates cell adhesion via its cleavage by caspase in sympathetic neurons. J Biol Chem 286: 14628–14638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cai J, Jiang WG, Grant MB, Boulton M 2006. Pigment epithelium-derived factor inhibits angiogenesis via regulated intracellular proteolysis of vascular endothelial growth factor receptor 1. J Biol Chem 281: 3604–3613 [DOI] [PubMed] [Google Scholar]
  23. Cai J, Chen Z, Ruan Q, Han S, Liu L, Qi X, Boye SL, Hauswirth WW, Grant MB, Boulton ME 2011a. γ-Secretase and presenilin mediate cleavage and phosphorylation of vascular endothelial growth factor receptor-1. J Biol Chem 286: 42514–42523 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Cai J, Wu L, Qi X, Li Calzi S, Caballero S, Shaw L, Ruan Q, Grant MB, Boulton ME 2011b. PEDF regulates vascular permeability by a γ-secretase-mediated pathway. PLoS ONE 6: e21164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Carpenter G 2003. Nuclear localization and possible functions of receptor tyrosine kinases. Curr Opin Cell Biol 15: 143–148 [DOI] [PubMed] [Google Scholar]
  26. Carpenter G, Liao HJ 2009. Trafficking of receptor tyrosine kinases to the nucleus. Exp Cell Res 315: 1556–1566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Carson DD 2008. The cytoplasmic tail of MUC1: A very busy place. Sci Signal 1: e35. [DOI] [PubMed] [Google Scholar]
  28. Chandra M, Zang S, Li H, Zimmerman LJ, Champer J, Tsuyada A, Chow A, Zhou W, Yu Y, Gao H, et al. 2012. Nuclear translocation of type I transforming growth factor β receptor confers a novel function in RNA processing. Mol Cell Biol 32: 2183–2195 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Chaves-Pérez A, Mack B, Maetzel D, Kremling H, Eggert C, Harréus U, Gires O 2013. EpCAM regulates cell cycle progression via control of cyclin D1 expression. Oncogene 32: 641–650 [DOI] [PubMed] [Google Scholar]
  30. Ceni C, Kommaddi RP, Thomas R, Vereker E, Liu X, McPherson PS, Ritter B, Barker PA 2010. The p75NTR intracellular domain generated by neurotrophin-induced receptor cleavage potentiates Trk signaling. J Cell Sci 123: 2299–22307 [DOI] [PubMed] [Google Scholar]
  31. Chen CW, Roy D 1996. Up-regulation of nuclear IGF-I receptor by short term exposure of stilbene estrogen, diethylstilbestrol. Mol Cell Endocrinol 118: 1–8 [DOI] [PubMed] [Google Scholar]
  32. Cheng QC, Tikhomirov O, Zhou W, Carpenter G 2003. Ectodomain cleavage of ErbB-4: Characterization of the cleavage site and m80 fragment. J Biol Chem 278: 38421–38427 [DOI] [PubMed] [Google Scholar]
  33. Chioni AM, Grose R 2012. FGFR1 cleavage and nuclear translocation regulates breast cancer cell behavior. J Cell Biol 197: 801–817 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Codony-Servat J, Albanell J, Lopez-Talavera JC, Arribas J, Baselga J 1999. Cleavage of the HER2 ectodomain is a pervanadate-activable process that is inhibited by the tissue inhibitor of metalloproteases-1 in breast cancer cells. Cancer Res 59: 1196–11201 [PubMed] [Google Scholar]
  35. Cousin H, Abbruzzese G, Kerdavid E, Gaultier A, Alfandari D 2011. Translocation of the cytoplasmic domain of ADAM13 to the nucleus is essential for Calpain8-a expression and cranial neural crest cell migration. Dev Cell 20: 256–263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. De Angelis Campos AC, Rodrigues MA, de Andrade C, de Goes AM, Nathanson MH, Gomes DA 2011. Epidermal growth factor receptors destined for the nucleus are internalized via a clathrin-dependent pathway. Biochem Biophys Res Commun 412: 341–346 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Degnin CR, Laederich MB, Horton WA 2011. Ligand activation leads to regulated intramembrane proteolysis of fibroblast growth factor receptor 3. Mol Biol Cell 22: 3861–3873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Deheuninck J, Goormachtigh G, Foveau B, Ji Z, Leroy C, Ancot F, Villeret V, Tulasne D, Fafeur V 2009. Phosphorylation of the MET receptor on juxtamembrane tyrosine residue 1001 inhibits its caspase-dependent cleavage. Cell Signal 21: 1455–1463 [DOI] [PubMed] [Google Scholar]
  39. Deng H, Lin Y, Badin M, Vasilcanu D, Strömberg T, Jernberg-Wiklund H, Sehat B, Larsson O 2010. Over-accumulation of nuclear IGF-1 receptor in tumor cells requires elevated expression of the receptor and the SUMO-conjugating enzyme Ubc9. Biochem Biophys Res Commun 404: 667–671 [DOI] [PubMed] [Google Scholar]
  40. Denzel S, Maetzel D, Mack B, Eggert C, Bärr G, Gires O 2009. Initial activation of EpCAM cleavage via cell-to-cell contact. BMC Cancer 9: 402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. De Strooper B, Annaert W 2010. Novel research horizons for presenilins and γ-secretases in cell biology and disease. Annu Rev Cell Dev Biol 26: 235–260 [DOI] [PubMed] [Google Scholar]
  42. Dittmann K, Mayer C, Fehrenbacher B, Schaller M, Raju U, Milas L, Chen DJ, Kehlbach R, Rodemann HP 2005. Radiation-induced epidermal growth factor receptor nuclear import is linked to activation of DNA-dependent protein kinase. J Biol Chem 280: 31182–31189 [DOI] [PubMed] [Google Scholar]
  43. Dittmann K, Mayer C, Fehrenbacher B, Schaller M, Kehlbach R, Rodemann HP 2010. Nuclear EGFR shuttling induced by ionizing radiation is regulated by phosphorylation at residue Thr654. FEBS Lett 584: 3878–3884 [DOI] [PubMed] [Google Scholar]
  44. Domingues I, Rino J, Demmers JA, de Lanerolle P, Santos SC 2001. VEGFR2 translocates to the nucleus to regulate its own transcription. PLoS ONE 6: e25668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Dunham-Ems SM, Lee YW, Stachowiak EK, Pudavar H, Claus P, Prasad PN 2009. Stachowiak MK. Fibroblast growth factor receptor-1 (FGFR1) nuclear dynamics reveal a novel mechanism in transcription control. Mol Biol Cell 20: 2401–2412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Elenius K, Corfas G, Paul S, Choi CJ, Rio C, Plowman GD, Klagsbrun M 1997. A novel juxtamembrane domain isoform of HER4/ErbB4. Isoform-specific tissue distribution and differential processing in response to phorbol ester. J Biol Chem 272: 26761–26768 [DOI] [PubMed] [Google Scholar]
  47. Feng SM, Muraoka-Cook RS, Hunter D, Sandahl MA, Caskey LS, Miyazawa K, Atfi A, Earp HS III 2009. The E3 ubiquitin ligase WWP1 selectively targets HER4 and its proteolytically derived signaling isoforms for degradation. Mol Cell Biol 29: 892–906 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Fortini ME 2009. Notch signaling: The core pathway and its posttranslational regulation. Dev Cell 16: 633–647 [DOI] [PubMed] [Google Scholar]
  49. Foveau B, Leroy C, Ancot F, Deheuninck J, Ji Z, Fafeur V, Tulasne D 2007. Amplification of apoptosis through sequential caspase cleavage of the MET tyrosine kinase receptor. Cell Death Differ 14: 752–764 [DOI] [PubMed] [Google Scholar]
  50. Foveau B, Ancot F, Leroy C, Petrelli A, Reiss K, Vingtdeux V, Giordano S, Fafeur V, Tulasne D 2009. Down-regulation of the met receptor tyrosine kinase by presenilin-dependent regulated intramembrane proteolysis. Mol Biol Cell 20: 2495–2507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Fox SB, Turley H, Cheale M, Blázquez C, Roberts H, James N, Cook N, Harris A, Gatter K 2004. Phosphorylated KDR is expressed in the neoplastic and stromal elements of human renal tumours and shuttles from cell membrane to nucleus. J Pathol 202: 313–320 [DOI] [PubMed] [Google Scholar]
  52. García-Castillo J, Pedersen K, Angelini PD, Bech-Serra JJ, Colomé N, Cunningham MP, Parra-Palau JL, Canals F, Baselga J, Arribas J 2009. HER2 carboxyl-terminal fragments regulate cell migration and cortactin phosphorylation. J Biol Chem 284: 25302–25313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Georgakopoulos A, Xu J, Xu C, Mauger G, Barthet G, Robakis NK 2011. Presenilin1/γ-secretase promotes the EphB2-induced phosphorylation of ephrinB2 by regulating phosphoprotein associated with glycosphingolipid-enriched microdomains/Csk binding protein. FASEB J 25: 3594–3604 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Giri DK, Ali-Seyed M, Li LY, Lee DF, Ling P, Bartholomeusz G, Wang SC, Hung MC 2005. Endosomal transport of ErbB-2: Mechanism for nuclear entry of the cell surface receptor. Mol Cell Biol 25: 11005–11018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Giulianelli S, Cerliani JP, Lamb CA, Fabris VT, Bottino MC, Gorostiaga MA, Novaro V, Góngora A, Baldi A, Molinolo A, et al. 2008. Carcinoma-associated fibroblasts activate progesterone receptors and induce hormone independent mammary tumor growth: A role for the FGF-2/FGFR-2 axis. Int J Cancer 123: 2518–2531 [DOI] [PubMed] [Google Scholar]
  56. Glenn G, van der Geer P 2007. CSF-1 and TPA stimulate independent pathways leading to lysosomal degradation or regulated intramembrane proteolysis of the CSF-1 receptor. FEBS Lett 581: 5377–5381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Glenn G, van der Geer P 2008. Toll-like receptors stimulate regulated intramembrane proteolysis of the CSF-1 receptor through Erk activation. FEBS Lett 582: 911–915 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Goldschneider D, Rama N, Guix C, Mehlen P 2008. The neogenin intracellular domain regulates gene transcription via nuclear translocation. Mol Cell Biol 28: 4068–4079 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Golubkov VS, Strongin AY 2012. Insights into ectodomain shedding and processing of protein-tyrosine pseudokinase 7 (PTK7). J Biol Chem 287: 2009–42018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Gomes DA, Rodrigues MA, Leite MF, Gomez MV, Varnai P, Balla T, Bennett AM, Nathanson MH 2008. c-Met must translocate to the nucleus to initiate calcium signals. J Biol Chem 283: 4344–4351 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Grumolato L, Liu G, Mong P, Mudbhary R, Biswas R, Arroyave R, Vijayakumar S, Economides AN, Aaronson SA 2010. Canonical and noncanonical Wnts use a common mechanism to activate completely unrelated coreceptors. Genes Dev 24: 2517–2530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Han J, Allalunis-Turner J, Hendzel MJ 2011. Characterization and comparison of protein complexes initiated by the intracellular domain of individual Notch paralogs. Biochem Biophys Res Commun 407: 479–485 [DOI] [PubMed] [Google Scholar]
  63. Hanada N, Lo HW, Day CP, Pan Y, Nakajima Y, Hung MC 2006. Co-regulation of B-Myb expression by E2F1 and EGF receptor. Mol Carcinog 45: 10–17 [DOI] [PubMed] [Google Scholar]
  64. He G, Luo W, Li P, Remmers C, Netzer WJ, Hendrick J, Bettayeb K, Flajolet M, Gorelick F, Wennogle LP, et al. 2010. γ-Secretase activating protein is a therapeutic target for Alzheimer’s disease. Nature 467: 95–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hoeing K, Zscheppang K, Mujahid S, Murray S, Volpe MV, Dammann CE, Nielsen HC 2011. Presenilin-1 processing of ErbB4 in fetal type II cells is necessary for control of fetal lung maturation. Biochim Biophys Acta 1813: 480–491 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Hsu SC, Hung MC 2007. Characterization of a novel tripartite nuclear localization sequence in the EGFR family. J Biol Chem 282: 10432–10440 [DOI] [PubMed] [Google Scholar]
  67. Huang WC, Chen YJ, Li LY, Wei YL, Hsu SC, Tsai SL, Chiu PC, Huang WP, Wang YN, Chen CH, et al. 2011a. Nuclear translocation of epidermal growth factor receptor by Akt-dependent phosphorylation enhances breast cancer-resistant protein expression in gefitinib-resistant cells. J Biol Chem 286: 20558–20568 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Huang HP, Chen PH, Yu CY, Chuang CY, Stone L, Hsiao WC, Li CL, Tsai SC, Chen KY, Chen HF, et al. 2011b. Epithelial cell adhesion molecule (EpCAM) complex proteins promote transcription factor-mediated pluripotency reprogramming. J Biol Chem 286: 33520–33532 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Janiszewska M, De Vito C, Le Bitoux MA, Fusco C, Stamenkovic I 2010. Transportin regulates nuclear import of CD44. J Biol Chem 285: 30548–30557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Jones FE 2008. HER4 intracellular domain (4ICD) activity in the developing mammary gland and breast cancer. J Mammary Gland Biol Neoplasia 13: 247–258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Kagawa H, Shino Y, Kobayashi D, Demizu S, Shimada M, Ariga H, Kawahara H 2011. A novel signaling pathway mediated by the nuclear targeting of C-terminal fragments of mammalian Patched 1. PLoS ONE 6: e18638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Kasuga K, Kaneko H, Nishizawa M, Onodera O, Ikeuchi T 2007. Generation of intracellular domain of insulin receptor tyrosine kinase by γ-secretase. Biochem Biophys Res Commun 360: 90–96 [DOI] [PubMed] [Google Scholar]
  73. Kim J, Jahng WJ, Di Vizio D, Lee JS, Jhaveri R, Rubin MA, Shisheva A, Freeman MR 2007. The phosphoinositide kinase PIKfyve mediates epidermal growth factor receptor trafficking to the nucleus. Cancer Res 67: 9229–9237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Kopan R, Ilagan MX 2004. γ-Secretase: Proteasome of the membrane? Nat Rev Mol Cell Biol 5: 499–504 [DOI] [PubMed] [Google Scholar]
  75. Lai C, Feng L 2004. Implication of γ-secretase in neuregulin-induced maturation of ligodendrocytes. Biochem Biophys Res Commun 314: 535–542 [DOI] [PubMed] [Google Scholar]
  76. Lal M, Caplan M 2011. Regulated intramembrane proteolysis: Signaling pathways and biological functions. Physiology (Bethesda) 26: 34–44 [DOI] [PubMed] [Google Scholar]
  77. Lee TH, Seng S, Sekine M, Hinton C, Fu Y, Avraham HK, Avraham S 2007. Vascular endothelial growth factor mediates intracrine survival in human breast carcinoma cells through internally expressed VEGFR1/FLT1. PLoS Med 4: e186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Lemberg MK 2011. Intramembrane proteolysis in regulated protein trafficking. Traffic 12: 1109–1118 [DOI] [PubMed] [Google Scholar]
  79. Le Moan N, Houslay DM, Christian F, Houslay MD, Akassoglou K 2011. Oxygen-dependent cleavage of the p75 neurotrophin receptor triggers stabilization of HIF-1α. Mol Cell 44: 476–490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Li LY, Chen H, Hsieh YH, Wang YN, Chu HJ, Chen YH, Chen HY, Chien PJ, Ma HT, Tsai HC, et al. 2011. Nuclear ErbB2 enhances translation and cell growth by activating transcription of ribosomal RNA genes. Cancer Res 71: 4269–4279 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Liao HJ, Carpenter G 2007. Role of the Sec61 translocon in EGF receptor trafficking to the nucleus and gene expression. Mol Biol Cell 18: 1064–1072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Liao HJ, Carpenter G 2009. Cetuximab/C225-induced intracellular trafficking of epidermal growth factor receptor. Cancer Res 69: 6179–6183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Liao HJ, Carpenter G 2012. Regulated intramembrane cleavage of the EGF receptor. Traffic 13: 1106–1112 [DOI] [PubMed] [Google Scholar]
  84. Liccardi G, Hartley JA, Hochhauser D 2011. EGFR nuclear translocation modulates DNA repair following cisplatin and ionizing radiation treatment. Cancer Res 71: 1103–1114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Lin SY, Makino K, Xia W, Matin A, Wen Y, Kwong KY, Bourguignon L, Hung MC 2001. Nuclear localization of EGF receptor and its potential new role as a transcription factor. Nat Cell Biol 3: 802–808 [DOI] [PubMed] [Google Scholar]
  86. Lin KT, Sloniowski S, Ethell DW, Ethell IM 2008. Ephrin-B2-induced cleavage of EphB2 receptor is mediated by matrix metalloproteinases to trigger cell repulsion. J Biol Chem 283: 28969–28979 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Litterst C, Georgakopoulos A, Shioi J, Ghersi E, Wisniewski T, Wang R, Ludwig A, Robakis NK 2007. Ligand binding and calcium influx induce distinct ectodomain/γ-secretase-processing pathways of EphB2 receptor. J Biol Chem 282: 16155–16163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Liu HS, Hsu PY, Lai MD, Chang HY, Ho CL, Cheng HL, Chen HT, Lin YJ, Wu TJ, Tzai TS, et al. 2010. An unusual function of RON receptor tyrosine kinase as a transcriptional regulator in cooperation with EGFR in human cancer cells. Carcinogenesis 31: 1456–1464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Lo HW, Hsu SC, Ali-Seyed M, Gunduz M, Xia X, Wei Y, Bartholomeusz G, Shih JY, Hung MC 2005. Nuclear interaction of EGFR and STAT3 in the activation of the iNOS/NO pathway. Cancer Cell 7: 575–589 [DOI] [PubMed] [Google Scholar]
  90. Lo HW, Ali-Seyed M, Wu Y, Bartholomeusz G, Hsu SC, Hung MC 2006. Nuclear-cytoplasmic transport of EGFR involves receptor endocytosis, importin β1 and CRM1. J Cell Biochem 98: 1570–1583 [DOI] [PubMed] [Google Scholar]
  91. Lo HW, Cao X, Zhu H, Ali-Osman F 2010. Cyclooxygenase-2 is a novel transcriptional target of the nuclear EGFR-STA3 and EGFRvIII-STAT3 signaling axes. Mol Cancer Res 8: 232–245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. López-Otín C, Hunter T 2010. The regulatory crosstalk between kinases and proteases in cancer. Nat Rev Cancer 10: 278–292 [DOI] [PubMed] [Google Scholar]
  93. Lu P, Ewald AJ, Martin GR, Werb Z 2004. Genetic mosaic analysis reveals FGF receptor 2 function in terminal end buds during mammary gland branching morphogenesis. Dev Biol 321: 77–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Lyu J, Yamamoto V, Lu W 2008. Cleavage of the Wnt receptor Ryk regulates neuronal differentiation during cortical neurogenesis. Dev Cell 15: 773–780 [DOI] [PubMed] [Google Scholar]
  95. Lyu J, Wesselschmidt RL, Lu W 2009. Cdc37 regulates Ryk signaling by stabilizing the cleaved Ryk intracellular domain. J Biol Chem 284: 12940–12948 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Maetzel D, Denzel S, Mack B, Canis M, Went P, Benk M, Kieu C, Papior P, Baeuerle PA, Munz M, et al. 2009. Nuclear signalling by tumour-associated antigen EpCAM. Nat Cell Biol 11: 162–171 [DOI] [PubMed] [Google Scholar]
  97. Marron MB, Singh H, Tahir TA, Kavumkal J, Kim HZ, Koh GY, Brindle NP 2007. Regulated proteolytic processing of Tie1 modulates ligand responsiveness of the receptor-tyrosine kinase Tie2. J Biol Chem 282: 30509–30517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Martin AJ, Grant A, Ashfield AM, Palmer CN, Baker L, Quinlan PR, Purdie CA, Thompson AM, Jordan LB, Berg JN 2011. FGFR2 protein expression in breast cancer: Nuclear localization and correlation with patient genotype. BMC Res Notes 4: 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Mathew D, Ataman B, Chen J, Zhang Y, Cumberledge S, Budnik V 2005. Wingless signaling at synapses is through cleavage and nuclear import of receptor DFrizzled2. Science 310: 1344–1347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Mazzone M, Selfors LM, Albeck J, Overholtzer M, Sale S, Carroll DL, Pandya D, Lu Y, Mills GB, Aster JC, et al. 2010. Dose-dependent induction of distinct phenotypic responses to Notch pathway activation in mammary epithelial cells. Proc Natl Acad Sci 107: 5012–5017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. McCarthy JV, Twomey C, Wujek P 2009. Presenilin-dependent regulated intramembrane proteolysis and γ-secretase activity. Cell Mol Life Sci 66: 1534–1555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. McElroy B, Powell JC, McCarthy JV 2007. The insulin-like growth factor 1 (IGF-1) receptor is a substrate for γ-secretase-mediated intramembrane proteolysis. Biochem Biophys Res Commun 358: 1136–1141 [DOI] [PubMed] [Google Scholar]
  103. Merlin J, Stechly L, de Beaucé S, Monté D, Leteurtre E, van Seuningen I, Huet G, Pigny P 2011. Galectin-3 regulates MUC1 and EGFR cellular distribution and EGFR downstream pathways in pancreatic cancer cells. Oncogene 30: 2514–2525 [DOI] [PubMed] [Google Scholar]
  104. Migaki GI, Kahn J, Kishimoto TK 1995. Mutational analysis of the membrane-proximal cleavage site of l-selectin: Relaxed sequence specificity surrounding the cleavage site. J Exp Med 182: 549–557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Miletti-González KE, Murphy K, Kumaran MN, Ravindranath AK, Wernyj RP, Kaur S, Miles GD, Lim E, Chan R, Chekmareva M, et al. 2012. Identification of function for CD44 intracytoplasmic domain (CD44-ICD): Modulation of matrix metalloproteinase 9 (MMP-9) transcription via novel promoter response element. J Biol Chem 287: 18995–19007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Montes de Oca P, Malardé V, Proust R, Dautry-Varsat A, Gesbert F 2010. Ectodomain shedding of interleukin-2 receptor β and generation of an intracellular functional fragment. J Biol Chem 285: 22050–22058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Mourali J, Bénard A, Lourenço FC, Monnet C, Greenland C, Moog-Lutz C, Racaud-Sultan C, Gonzalez-Dunia D, Vigny M, Mehlen P, et al. 2006. Anaplastic lymphoma kinase is a dependence receptor whose proapoptotic functions are activated by caspase cleavage. Mol Cell Biol 26: 6209–6222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Mu Y, Sundar R, Thakur N, Ekman M, Gudey SK, Yakymovych M, Hermansson A, Dimitriou H, Bengoechea-Alonso MT, Ericsson J, et al. 2011. TRAF6 ubiquitinates TGF-β type I receptor to promote its cleavage and nuclear translocation in cancer. Nat Commun 2: 330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Muraoka-Cook RS, Sandahl M, Husted C, Hunter D, Miraglia L, Feng SM, Elenius K, Earp HS 2006. The intracellular domain of ErbB4 induces differentiation of mammary epithelial cells. Mol Biol Cell 17: 4118–4129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Na HW, Shin WS, Ludwig A, Lee ST 2012. The cytosolic domain of protein-tyrosine kinase 7 (PTK7), generated from sequential cleavage by a disintegrin and metalloprotease 17 (ADAM17) and γ-secretase, enhances cell proliferation and migration in colon cancer cells. J Biol Chem 287: 25001–25009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Naresh A, Long W, Vidal GA, Wimley WC, Marrero L, Sartor CI, Tovey S, Cooke TG, Bartlett JM, Jones FE 2006. The ERBB4/HER4 intracellular domain 4ICD is a BH3-only protein promoting apoptosis of breast cancer cells. Cancer Res 66: 6412–6420 [DOI] [PubMed] [Google Scholar]
  112. Ni CY, Murphy MP, Golde TE, Carpenter G 2001. γ-Secretase cleavage and nuclear localization of ErbB-4 receptor tyrosine kinase. Science 294: 2179–2181 [DOI] [PubMed] [Google Scholar]
  113. Ni CY, Yuan H, Carpenter G 2003. Role of the ErbB-4 carboxyl terminus in γ-secretase cleavage. J Biol Chem 278: 4561–4565 [DOI] [PubMed] [Google Scholar]
  114. Offterdinger M, Schofer C, Weipoltshammer K, Grunt TW 2002. c-erbB-3: A nuclear protein in mammary epithelial cells. J Cell Biol 157: 929–939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Omerovic J, Santangelo L, Puggioni EM, Marrocco J, Dall’Armi C, Palumbo C, Belleudi F, Di Marcotullio L, Frati L, Torrisi MR, et al. 2007. The E3 ligase Aip4/Itch ubiquitinates and targets ErbB-4 for degradation. FASEB J 21: 2849–2862 [DOI] [PubMed] [Google Scholar]
  116. Paatero I, Jokilammi A, Heikkinen PT, Iljin K, Kallioniemi OP, Jones FE, Jaakkola PM, Elenius K 2012. Interaction with ErbB4 promotes hypoxia-inducible factor-1α signaling. J Biol Chem 287: 9659–9671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Pan B, Huang XF, Deng C 2011. Antipsychotic treatment and neuregulin 1-ErbB4 signalling in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 35: 924–930 [DOI] [PubMed] [Google Scholar]
  118. Parkhurst CN, Zampieri N, Chao MV 2010. Nuclear localization of the p75 neurotrophin receptor intracellular domain. J Biol Chem 285: 5361–5368 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Pedersen K, Angelini PD, Laos S, Bach-Faig A, Cunningham MP, Ferrer-Ramón C, Luque-García A, García-Castillo J, Parra-Palau JL, Scaltriti M, et al. 2009. A naturally occurring HER2 carboxy-terminal fragment promotes mammary tumor growth and metastasis. Mol Cell Biol 29: 3319–3331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Piccione EC, Lieu TJ, Gentile CF, Williams TR, Connolly AJ, Godwin AK, Koong AC, Wong AJ 2012. A novel epidermal growth factor receptor variant lacking multiple domains directly activates transcription and is overexpressed in tumors. Oncogene 31: 2953–2967 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Pozner-Moulis S, Pappas DJ, Rimm DL 2006. Met, the hepatocyte growth factor receptor, localizes to the nucleus in cells at low density. Cancer Res 66: 7976–7982 [DOI] [PubMed] [Google Scholar]
  122. Racaud-Sultan C, Gonzalez-Dunia D, Vigny M, Mehlen P, Delsol G, Allouche M 2006. Anaplastic lymphoma kinase is a dependence receptor whose proapoptotic functions are ctivated by caspase cleavage. Mol Cell Biol 26: 6209–6222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Rahimi N, Golde TE, Meyer RD 2009. Identification of ligand-induced proteolytic cleavage and ectodomain shedding of VEGFR-1/FLT1 in leukemic cancer cells. Cancer Res 69: 2607–2614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Ralhan R, He HC, So AK, Tripathi SC, Kumar M, Hasan MR, Kaur J, Kashat L, MacMillan C, Chauhan SS, et al. 2010. Nuclear and cytoplasmic accumulation of Ep-ICD is frequently detected in human epithelial cancers. PLoS ONE 5: e14130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Rebeck GW 2009. Nontraditional signaling mechanisms of lipoprotein receptors. Sci Signal 2: e28. [DOI] [PubMed] [Google Scholar]
  126. Rio C, Buxbaum JD, Peschon JJ, Corfas G 2000. Tumor necrosis factor-α-converting enzyme is required for cleavage of erbB4/HER4. J Biol Chem 275: 10379–10387 [DOI] [PubMed] [Google Scholar]
  127. Rokicki J, Das PM, Giltnane JM, Wansbury O, Rimm DL, Howard BA, Jones FE 2010. The ERα coactivator, HER4/4ICD, regulates progesterone receptor expression in normal and malignant breast epithelium. Mol Cancer 9: 150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Sandvig K, van Deurs B 2002. Transport of protein toxins into cells: Pathways used by ricin, cholera toxin and Shiga toxin. FEBS Lett 529: 49–53 [DOI] [PubMed] [Google Scholar]
  129. Santos SC, Dias S 2004. Internal and external autocrine VEGF/KDR loops regulate survival of subsets of acute leukemia through distinct signaling pathways. Blood 103: 3883–3889 [DOI] [PubMed] [Google Scholar]
  130. Santos SC, Miguel C, Domingues I, Calado A, Zhu Z, Wu Y, Dias S 2007. VEGF and VEGFR-2 (KDR) internalization is required for endothelial recovery during wound healing. Exp Cell Res 313: 1561–1574 [DOI] [PubMed] [Google Scholar]
  131. Sardi SP, Murtie J, Koirala S, Patten BA, Corfas G 2006. Presenilin-dependent ErbB4 nuclear signaling regulates the timing of astrogenesis in the developing brain. Cell 127: 185–197 [DOI] [PubMed] [Google Scholar]
  132. Schmahl J, Kim Y, Colvin JS, Ornitz DM, Capel B 2004. Fgf9 induces proliferation and nuclear localization of FGFR2 in Sertoli precursors during male sex determination. Development 131: 3627–3636 [DOI] [PubMed] [Google Scholar]
  133. Sehat B, Tofigh A, Lin Y, Trocmé E, Liljedahl U, Lagergren J, Larsson O 2010. SUMOylation mediates the nuclear translocation and signaling of the IGF-1 receptor. Sci Signal 3: ra10. [DOI] [PubMed] [Google Scholar]
  134. Selkoe DJ, Wolfe MS 2007. Presenilin: Running with scissors in the membrane. Cell 131: 215–221 [DOI] [PubMed] [Google Scholar]
  135. Shah S, Lee SF, Tabuchi K, Hao HY, Yu C, LaPlant Q, Ball H, Dann CE, Sudhof T, Yu G 2005. Nicastrin functions as a γ-secretase-substrate receptor. Cell 122: 435–447 [DOI] [PubMed] [Google Scholar]
  136. Small DH 2002. Is γ-secretase a multienzyme complex for membrane protein degradation? Models and speculations. Peptides 23: 1317–1321 [DOI] [PubMed] [Google Scholar]
  137. Stachowiak MK, Maher PA, Stachowiak EK 2007. Integrative nuclear signaling in cell development—A role for FGF receptor-1. DNA Cell Biol 26: 811–826 [DOI] [PubMed] [Google Scholar]
  138. Stewart M, Turley H, Cook N, Pezzella F, Pillai G, Ogilvie D, Cartlidge S, Paterson D, Copley C, Kendrew J, et al. 2003. The angiogenic receptor KDR is widely distributed in human tissues and tumours and relocates intracellularly on phosphorylation. An immunohistochemical study. Histopathology 43: 33–39 [DOI] [PubMed] [Google Scholar]
  139. Stoeck A, Shang L, Dempsey PJ 2010. Sequential and γ-secretase-dependent processing of the β-cellulin precursor generates a palmitoylated intracellular-domain fragment that inhibits cell growth. J Cell Sci 123: 2319–2331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Strohecker AM, Yehiely F, Chen F, Cryns VL 2008. Caspase cleavage of HER-2 releases a bad-like cell death effector. J Biol Chem 283: 18269–18282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Struhl G, Adachi A 2000. Requirements for presenilin-dependent cleavage of notch and other transmembrane proteins. Mol Cell 6: 625–636 [DOI] [PubMed] [Google Scholar]
  142. Tagami S, Okochi M, Yanagida K, Ikuta A, Fukumori A, Matsumoto N, Ishizuka-Katsura Y, Nakayama T, Itoh N, Jiang J, et al. 2008. Regulation of Notch signaling by dynamic changes in the precision of S3 cleavage of Notch-1. Mol Cell Biol 28: 165–176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Tao Y, Song X, Deng X, Xie D, Lee LM, Liu Y, Li W, Li L, Deng L, Wu Q, et al. 2005. Nuclear accumulation of epidermal growth factor receptor and acceleration of G1/S stage by Epstein–Barr-encoded oncoprotein latent membrane protein 1. Exp Cell Res 303: 240–251 [DOI] [PubMed] [Google Scholar]
  144. Tauszig-Delamasure S, Yu LY, Cabrera JR, Bouzas-Rodriguez J, Mermet-Bouvier C, Guix C, Bordeaux MC, Arumae U, Mehlen P 2007. The TrkC receptor induces apoptosis when the dependence receptor notion meets the neurotrophin paradigm. Proc Natl Acad Sci 104: 13361–13366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Thor AD, Edgerton SM, Jones FE 2009. Subcellular localization of the HER4 ntracellular domain, 4ICD, identifies distinct prognostic outcomes for breast cancer patients. Am J Pathol 175: 1802–1809 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Tikhomirov O, Carpenter G 2001. Caspase-dependent cleavage of ErbB-2 by geldanamycin and staurosporin. J Biol Chem 276: 33675–33680 [DOI] [PubMed] [Google Scholar]
  147. Tomita T, Tanaka S, Morohashi Y, Iwatsubo T 2006. Presenilin-dependent intramembrane cleavage of ephrin-B1. Mol Neurodegener 1: 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Tousseyn T, Thathiah A, Jorissen E, Raemaekers T, Konietzko U, Reiss K, Maes E, Snellinx A, Serneels L, Nyabi O, et al. 2009. ADAM10, the rate-limiting protease of regulated intramembrane proteolysis of Notch and other proteins, is processed by ADAMS-9, ADAMS-15, and the γ-secretase. J Biol Chem 284: 11738–11747 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Tulasne D, Deheuninck J, Lourenco FC, Lamballe F, Ji Z, Leroy C, Puchois E, Moumen A, Maina F, Mehlen P, et al. 2004. Proapoptotic function of the MET tyrosine kinase receptor through caspase cleavage. Mol Cell Biol 24: 10328–10339 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Varshavsky A 1997. The N-end rule pathway of protein degradation. Genes Cells 2: 13–28 [DOI] [PubMed] [Google Scholar]
  151. Vecchi M, Baulida J, Carpenter G 1996. Selective cleavage of the heregulin receptor ErbB-4 by protein kinase C activation. J Biol Chem 271: 18989–18995 [DOI] [PubMed] [Google Scholar]
  152. Veikkolainen V, Vaparanta K, Halkilahti K, Iljin K, Sundvall M, Elenius K 2011. Function of ERBB4 is determined by alternative splicing. Cell Cycle 10: 2647–2657 [DOI] [PubMed] [Google Scholar]
  153. Vidal GA, Naresh A, Marrero L, Jones FE 2005. Presenilin-dependent γ-secretase processing regulates multiple ERBB4/HER4 activities. J Biol Chem 280: 19777–19783 [DOI] [PubMed] [Google Scholar]
  154. Wang SC, Lien HC, Xia W, Chen IF, Lo HW, Wang Z, Ali-Seyed M, Lee DF, Bartholomeusz G, Ou-Yang F, et al. 2004. Binding at and transactivation of the COX-2 promoter by nuclear tyrosine kinase receptor ErbB-2. Cancer Cell 6: 251–261 [DOI] [PubMed] [Google Scholar]
  155. Wang SC, Nakajima Y, Yu YL, Xia W, Chen CT, Yang CC, McIntush EW, Li LY, Hawke DH, Kobayashi R, et al. 2006. Tyrosine phosphorylation controls PCNA function through protein stability. Nat Cell Biol 8: 1359–1368 [DOI] [PubMed] [Google Scholar]
  156. Wang X, Cui M, Wang L, Chen X, Xin P 2010a. Inhibition of neurotrophin receptor p75 intramembran proteolysis by γ-secretase inhibitor reduces medulloblastoma spinal metastasis. Biochem Biophys Res Commun 403: 264–269 [DOI] [PubMed] [Google Scholar]
  157. Wang YN, Yamaguchi H, Huo L, Du Y, Lee HJ, Lee HH, Wang H, Hsu JM, Hung MC 2010b. The translocon Sec61β localized in the inner nuclear membrane transports membrane-embedded EGF receptor to the nucleus. J Biol Chem 285: 38720–38729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Wang YN, Yamaguchi H, Hsu JM, Hung MC 2010c. Nuclear trafficking of the epidermal growth factor receptor family membrane proteins. Oncogene 29: 3997–4006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Wang X, Cowan JW, Gerhart M, Zelickson BR, Jiang J, He K, Wolfe MS, Black RA, Frank SJ 2011a. γ-Secretase-mediated growth hormone receptor proteolysis: Mapping of the intramembranous cleavage site. Biochem Biophys Res Commun 408: 432–436 [DOI] [PubMed] [Google Scholar]
  160. Wang YN, Wang H, Yamaguchi H, Lee HJ, Lee HH, Hung MC 2011b. COPI-mediated retrograde trafficking from the Golgi to the ER regulates EGFR nuclear transport. Biochem Biophys Res Commun 399: 498–504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Wang YN, Lee HH, Lee HJ, Du Y, Yamaguchi H, Hung MC 2012. Membrane-bound trafficking regulates nuclear transport of integral epidermal growth factor receptor (EGFR) and ErbB-2. J Biol Chem 287: 16869–16879 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Wilhelmsen K, van der Geer P 2004. Phorbol 12-myristate 13-acetate-induced release of the colony-stimulating factor 1 receptor cytoplasmic domain into the cytosol involves two separate cleavage events. Mol Cell Biol 24: 454–464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Williams CC, Allison JG, Vidal GA, Burow ME, Beckman BS, Marrero L, Jones FE 2004. The ERBB4/HER4 receptor tyrosine kinase regulates gene expression by functioning as a STAT5A nuclear chaperone. J Cell Biol 167: 469–478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Wong J, Weickert CS 2009. Transcriptional interaction of an estrogen receptor splice variant and ErbB4 suggests convergence in gene susceptibility pathways in schizophrenia. J Biol Chem 284: 18824–18832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Xia W, Liu Z, Zong R, Liu L, Zhao S, Bacus SS, Mao Y, He J, Wulfkuhle JD, Petricoin EF III, et al. 2011. Truncated ErbB2 expressed in tumor cell nuclei contributes to acquired therapeutic resistance to ErbB2 kinase inhibitors. Mol Cancer Ther 10: 1367–1374 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Yu YL, Chou RH, Wu CH, Wang YN, Chang WJ, Tseng YJ, Chang WC, Lai CC, Lee HJ, Huo L, et al. 2012. Nuclear EGFR suppresses ribonuclease activity of polynucleotide phosphorylase through DNAPK-mediated phosphorylation at serine 776. J Biol Chem 287: 31015–31026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Zeng F, Zhang MZ, Singh AB, Zent R, Harris RC 2007. ErbB4 isoforms selectively regulate growth factor induced Madin-Darby canine kidney cell tubulogenesis. Mol Biol Cell 18: 4446–4456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Zeng F, Xu J, Harris RC 2009. Nedd4 mediates ErbB4 JM-a/CYT-1 ICD ubiquitination and degradation in MDCK II cells. FASEB J 23: 1935–1945 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Zhang Y, Pillai G, Gatter K, Blázquez C, Turley H, Pezzella F, Watt SM 2005. Expression and cellular localization of vascular endothelial growth factor A and its receptors in acute and chronic leukemias: An immunohistochemical study. Hum Pathol 36: 797–805 [DOI] [PubMed] [Google Scholar]
  170. Zhou W, Carpenter G 2000. Heregulin-dependent trafficking and cleavage of ErbB-4. J Biol Chem 275: 34737–34743 [DOI] [PubMed] [Google Scholar]
  171. Zscheppang K, Dörk T, Schmiedl A, Jones FE, Dammann CE 2011a. Neuregulin receptor ErbB4 functions as a transcriptional cofactor for the expression of surfactant protein B in the fetal lung. Am J Respir Cell Mol Biol 45: 761–767 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Zscheppang K, Konrad M, Zischka M, Huhn V, Dammann CE 2011b. Estrogen-induced upregulation of Sftpb requires transcriptional control of neuregulin receptor ErbB4 in mouse lung type II epithelial cells. Biochim Biophys Acta 1813: 1717–1727 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Biology are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES