You are on page 1of 9

Neuron, Vol.

18, 847–855, June, 1997, Copyright 1997 by Cell Press

Neuregulins and Their Receptors: Meeting Report


A Versatile Signaling Module
in Organogenesis and Oncogenesis
Steve Burden* and Yosef Yarden† domains. Likewise, the single ligand is synthesized as
* Molecular Neurobiology Program a transmembrane precursor containing a characteristic
Skirball Institute epidermal growth factor (EGF) -like motif with six cyste-
New York University Medical Center ine residues arranged in three loops (reviewed by Korn-
New York, New York 10016 feld, 1997). The Drosophila relative of ErbB (DER), how-
† Department of Molecular Cell Biology ever, binds four different ligands, one of which, vein,
The Weizmann Institute of Science has no transmembrane precursor, but like certain NRG
Rehovot 76100 isoforms, carries an immunoglobulin (Ig) -like motif. An-
Israel other ligand, argos, is an antagonist (reviewed by Perri-
mon and Perkins, 1997). A mammalian antagonist has
There has been rapid growth in our understanding of yet to be identified. Like their mammalian counterparts,
neuregulins (NRGs), a large group of structurally related the invertebrate ErbB receptors also signal primarily
polypeptide factors, and their receptors, four transmem- through the Ras-Raf-MAP-kinase pathway. In both in-
brane tyrosine kinases of the ErbB family. This new vertebrates and mammals, the signaling module is nec-
knowledge reveals a complex signaling system, used in essary for development of multiple and seemingly unre-
different tissues to regulate cell survival, proliferation, lated cell lineages and organs, and its requirement
and differentiation. Only 5 years ago, independent ef- coincides with more than one developmental phase.
forts to identify activities responsible for stimulating Another lesson that is mirrored in mammals is the tightly
Neu/ErbB-2 phosphorylation, Schwann cell proliferation, controlled availability of the ligands, whose range of
and acetylcholine receptor synthesis resulted in the action is relatively short; the synthesizing cell and the
striking discovery that the peptide factors, Neu differen- responsive tissue are juxtaposed or only a few cell layers
tiation factor (NDF/heregulin), glial growth factor (GGF), apart.
and acetylcholine receptor inducing activity (ARIA) were
the products of the same gene. The name neuregulin, The ErbB Signaling Network of Mammals
originally introduced by Mark Marchionni, represents Over the last 800 million years, the ErbB-NRG module
a composite of the initial names and recognizes the has undergone remarkable expansion through gene du-
importance of these factors in the nervous system. In- plication events to reach its present state of four recep-
tensive efforts have now clarified not only an enormous tors and a few dozen ligands. It is likely that this expan-
functional diversity of the NRG-ErbB signaling network, sion reflects an increasing physiological demand for
but also its role in midgestation organogenesis as a key control of additional cell lineages. The diversification
determinant in differentiation of several cell lineages potential of the module is enhanced by combinatorial
(Figure 1). Instructive illustrations of this diversity were protein–protein interactions. The diversity of responses
provided by presentations at a recent workshop orga- is generated by three levels of interacting molecules:
nized in the Banbury Center at the Cold Spring Harbor first, there are multiple ligands whose receptor-binding
Laboratory by Mark A. Marchionni (Cambridge Neuro- motifs, an EGF-like domain, are flanked by variable do-
Science) and Greg Lemke (Salk Institute, La Jolla, CA) mains. Second, all ligands bind to a set of receptors
on “Neuregulins and Neuregulin Receptors.” that can generate all possible homo- and heterodimers.
Neuregulins (NRGs) and their receptors are evolution- Third, each different receptor dimer can couple to a
arily conserved in both structure and function and were distinct set of cytoplasmic signaling proteins.
present more than a billion years ago. Therefore, we Ligands
start this review with lessons learned from invertebrates. A newly isolated family of NRG-like ligands, termed
We then discuss the structural basis of signaling by NRG2, was described by three groups at the workshop
these ligands and their receptors, including a newly (H. Chang, Stanford University, Palo Alto, CA; C. Lai,
discovered second NRG gene, and review current Scripps Institute, La Jolla, CA; and A. Goodearl, Millen-
thoughts on how diversity is generated through combi- nium Pharmaceuticals, Boston). Like NRG isoforms,
natorial protein interactions. Individual organs and cell these ligands come in several variants, which probably
lineages, whose development is critically regulated by arose via alternative splicing from a gene on chromo-
NRGs, are then described in light of recent gene tar- some 5, which is distinct from the human NRG gene
geting, by in vivo and in vitro experiments. (located on chromosome 8 and now termed NRG1). The
structure of these new ligands includes an EGF-like
Invertebrate ErbB Signaling Modules motif, which has two variants, a and b, as well as a
The simplest version of the NRG-ErbB network is found collection of other recognizable extracellular motifs that
in C. elegans; it is comprised of a single receptor, the are variably present: a signal peptide, two variants of a
product of the let-23 gene, and a single ligand encoded cysteine-containing N-terminal domain, either a cyste-
by lin-3. Nevertheless, the major structural determinants ine-rich domain (CRD) or a domain distantly related to
of mammalian NRGs and ErbBs are already present: the a kringle motif, an Ig-like loop, a glycosylation domain,
receptor’s ectodomain is a duplicated structure carrying and a transmembrane domain. The presence or absence
two cysteine-rich blocks, and its cytoplasmic portion of the latter domain determines whether the precursor
is a tyrosine kinase flanked by noncatalytic regulatory is made as a transmembrane protein or as a secreted
Neuron
848

Figure 1. Physiological Functions of the NRG-ErbB Signaling Module


The center panel schematically shows the four ErbB proteins crossing the plasma membrane (horizontal bar). ErbB-3 and ErbB-4 are the
direct NRG receptors, whereas ErbB-1 and ErbB-2 function as coreceptors. The tyrosine kinase domain of ErbB-3 is inactive (crossed). The
color code of the receptors is maintained in the other panels, each presenting one of the known roles of NRGs (functions symbolized by thick
arrows). Note juxtaposition of the sources of NRG (blue) and the responsive cells (red). AChR, acetylcholine receptor; SC, Schwann cell; and
NRG, neuregulin.

polypeptide. According to a new nomenclature, the vari- Thus, the transmembrane precursor forms of NRGs may
ous isoforms are grouped into three types: type I in- also function independently of ErbB proteins.
cludes variants with an Ig loop and a glycosylation do- Both NRG1 and NRG2 isoforms bind directly to ErbB-3
main (e.g., isoforms originally named NDF and ARIA); and to ErbB-4, but they recruit ErbB-1 and ErbB-2 as
NRGs with an Ig loop but no glycosylation domain are coreceptors. Whereas ErbB-2 has no known direct li-
termed type II (e.g., the NRG isoform originally called gand, ErbB-1 binds six mammalian EGF-like molecules:
GGF-II); and the CRD domain-containing factors, which EGF, transforming growth factor a, heparin-binding
were originally named sensory and motor neuron– EGF-like growth factor (HB-EGF), amphiregulin, epi-
derived factor (SMDF; Ho et al., 1995), are now termed regulin, and betacellulin. The latter, along with HB-EGF
type III NRGs. The cytoplasmic tails of some proNRG (Elenius et al., 1997) and possibly additional ligands,
isoforms are longer than some of the extracellular por- also binds to ErbB-4 (D. Stern, Yale University, New
tions, and their lengths are variable. They may function Haven, CT). The exact role of the structural heterogene-
in “reverse signaling” since the region common to all ity of NRGs is still unknown. Best understood is the
transmembrane NRG1s appears to associate specifi- variation in the EGF-like receptor-binding domain. The
cally with certain cytoplasmic signaling proteins (D. C-terminal loop of NRG1 determines the strength of
Falls, Emory University, Atlanta). Further, the transmem- receptor binding and has a role in determining which
brane isoform proNRG1-b2 has recently been isolated in receptor heterodimers will form: the higher affinity b
a screen for apoptosis-inducing genes, but its function is isoform stabilizes both ErbB-3/ErbB-2 and ErbB-3/
apparently cell autonomous (Grimm and Leder, 1997). ErbB-1 heterodimers, whereas the a isoform recruits
Meeting Review
849

only the former complex (Pinkas-Kramarski et al., 1996). is a dimer interface, and therefore may act as a dimeriza-
In addition, each NRG isoform has a distinct spatial tion site (Stern).
and temporal pattern of expression (C. Birchmeier, Max According to an alternative model (Y. Yarden, Weiz-
Delbrück Center, Berlin; N. Ratner, University of Cincin- mann Institute, Rehovot, Israel), NRGs are bivalent
nati). For example, type I isoforms, which are the only ligands; they first bind a direct receptor (either ErbB-3
NRG1 isoforms detectable at early embryonic ages, are or ErbB-4) through an N-terminally located high affinity
the predominant species expressed in the endocardium, site, and then a second site of NRG, whose affinity is
while type II NRGs are the major isoforms expressed in lower and specificity broader, selects a coreceptor with
embryonic skeletal muscle, and type III NRGs are pres- some preference for ErbB-2. High resolution NMR stud-
ent in sympathetic, motor, and sensory neurons (X. Yang ies (Jacobsen et al., 1996) and application of phage
and L. Role, Columbia University, New York). On the display technology to NRG (Sliwkowski) indirectly sup-
other hand, all NRG1 types are expressed in the ventric- port the existence of two receptor contact sites. His2
ular zone of the telencephalon. In addition, stromal cells (178) and Leu3, two invariant residues located at the
underlying many epithelial cell layers of parenchymal N-terminus of the EGF-like domain, are essential for
organs are a rich source of certain NRGs. NRG binding to ErbB-3 (see also Barbacci et al., 1995),
Receptors whereas several C-terminal residues dictate binding to
The two direct NRG receptors, ErbB-3 and ErbB-4, and both ErbB-3 and ErbB-4. Consistent with bivalency of
their two coreceptors, ErbB-1 and ErbB-2, qualify as another ErbB ligand, EGF, recent biophysical measure-
distinct components of a signaling network in that they ments determined several possible EGF:ErbB-1 stoi-
differ in most functional aspects, yet are capable of chiometries in solution, of which the 2:2 complex pre-
extensive intermolecular interactions. In addition to li- dominates (Lemmon et al., 1997). In addition, affinity
gand specificity and affinity, expression patterns, and labeling with EGF indicates that the C-terminus of this
intracellular trafficking following ligand binding, the four ligand contacts the receptor’s N-terminus, whereas the
NRG receptors differ in kinase activity and substrate C-tail of EGF is juxtaposed to the sequence intervening
selectivity. Most remarkable is ErbB-3, whose catalytic the two cysteine-rich domains of ErbB-1 (Summerfield
function and endocytosis are both impaired by compari- et al., 1996). Thus, NRGs may drive receptor dimerization
son to the other three receptors (Baulida et al., 1996). through a mechanism that combines conformational al-
The deficiency in catalytic function, however, can be terations in receptors with bivalent ligand interactions
overcome by the process of ligand-induced receptor generating a series of molecular complexes, including
dimerization, an essential step that precedes receptor a stable quaternary complex of two ligands bridging two
phosphorylation and intracellular signaling. Although receptors. Presumably, each NRG isoform is character-
ErbB-3 homodimers are functionally defective, this re- ized by a unique combination of the two sites, thereby
ceptor is recruited into heterodimers by the three other favoring certain receptor combinations.
ErbB proteins. Thus, ErbB-3 signaling depends not only Effector Signaling Pathways
on NRG binding, but also requires association with other As is the case for other receptor tyrosine kinases, ligand-
ErbB proteins. Ligand-induced formation of homo- and induced autophosphorylation of ErbB proteins on spe-
heterodimers is shared by the other ErbB proteins, and cific tyrosine residues establishes docking sites for cyto-
all 10 possible receptor combinations can exist, includ- plasmic signaling proteins sharing a phosphotyrosine
ing a homodimer of the ligand-less ErbB-2, which is binding motif (SH2 or PTB domains). Due to differences
formed as a consequence of either overexpression or an in the amino acid sequences surrounding phosphorylation
oncogenic mutation within the transmembrane domain. sites of each receptor, different sets of signaling pro-
Selection of a coreceptor by a ligand-occupied direct teins become associated with each of the 10 possible
receptor is clearly a nonrandom process that follows ErbB complexes. Several substrates, like Grb-2 and Shc,
hierarchical rules. For example, ErbB-2-containing het- are shared by most ErbB species. However, others like
erodimers are preferred, and heterodimers that can be the negative regulator c-Cbl (Levkowitz et al., 1996) and
driven by two types of ligands (e.g., ErbB-1/ErbB-3 com- the phosphatidylinositol-specific phospholipase (PLCg),
plexes) are better stabilized by NRG than by EGF (Tzahar are recruited only to ErbB-1, and possibly ErbB-2 (Co-
et al., 1996). How ligand binding promotes different re- hen et al., 1996), resulting in calcium mobilization only
ceptor combinations is currently an open question. Li- in response to EGF.
gand affinities of the various dimer combinations differ ErbB specificity of other substrates, such as Src and
significantly, even when dimers are artificially stabilized PI-3 kinase, must still be examined in cells whose reper-
with immunoglobulin Fc tails (ErbB-IgG; M. Sliwkowski, toire of these receptors is genetically manipulated.
Genentech). This is exemplified by heterodimeric com- Nevertheless, analyses of several cellular systems ex-
plexes of ErbB-3 or ErbB-4 with ErbB-2, whose affinities pressing individual, or pairs of, ErbB proteins have already
for NRGs exceed those of other combinations, including reached a consensus on the complementarity of various
an ErbB-3/ErbB-4 heterodimer. Studying the ErbB-2/ pairs (Riese et al., 1995; Cohen et al., 1996; Pinkas-
ErbB-3 heterodimer with an anti-ErbB-3 monoclonal an- Kramarski et al., 1996; Tzahar et al., 1996; Zhang et al.,
tibody and the corresponding ErbB-IgG, Sliwkowski 1996). Homodimers are in general less active than hetero-
supported the possibility that a conformational alter- dimers in stimulating cell proliferation, and ErbB-2-con-
ation takes place after NRG binding, exposing a cryptic taining heterodimers are superior to other heterodimeric
dimerization site that recruits ErbB-2. Replacements of complexes. The least effective and most unstable dimer
specific residues of ErbB-2 with a cysteine indicate that appears to be the NRG-driven ErbB-3/ErbB-4 complex,
the juxtamembrane portion of the extracellular domain and dimers formed between ErbB-1 and either NRG
Neuron
850

receptor display an intermediate signaling potency. The heterodimers of NRG receptors. Apparently, by requir-
remarkable mitogenic superiority of the ligand-less pro- ing heterodimers, cardiac development gains an addi-
tein, ErbB-2, attracted significant attention because this tional control level: the availability of a coreceptor. As
protein was implicated, more than other members of the for other growth factors, a clinical connection is in sight.
family, in cancer development. Functional inactivation of Since expression levels of NRG, ErbB-2, and ErbB-3
ErbB-2 demonstrated that ErbB-2 amplifies and pro- increase following treatments that induce cardiac hyper-
longs signaling by NRG and EGF through deceleration trophy, an adaptive or pathological thickening of the
of the rate of ligand dissociation (Graus-Porta et al., heart’s wall, the possibility that this hypertrophy may
1995; Karunagaran et al., 1996). Presumably, complexes be stimulated by increased NRG/ErbB signaling was
containing a ligand bound to a direct receptor and ErbB-2 raised by K.-F. Lee.
as a coreceptor are long-lived due to retarded ligand
dissociation and a relatively slow rate of ErbB-2 endocy-
tosis. On the other hand, because ErbB-2 is a strong NRGs in Schwann Cell and
activator of the MAP-kinase pathway, its heterodimers Oligodendrocyte Differentiation
are most potent in signaling. Most myelin-forming cells of the peripheral nervous sys-
tem develop from the neural crest in a multistep process
NRGs in Cardiac Development requiring contact with neurons. NRGs are important
Quite unexpectedly, but in agreement with the notion neuron-derived factors, which control Schwann cell dif-
that NRGs act through a signaling network rather than ferentiation, and are likely to have a critical role in regu-
via a direct receptor, targeting of the genes encoding lating Schwann cell number. In this case, a heterodimer
NRG (C. Birchmeier), ErbB-2 (K.-F. Lee, Salk Institute), of ErbB-2 with ErbB-3, rather than with ErbB-4, is the
and ErbB-4 (M. Gassman, Salk Institute) in mice resulted active receptor (Levi et al., 1995; Grinspan et al., 1996).
in an almost identical cardiac malformation (Gassmann In fact, ErbB-2 involvement in proliferation of Schwann
et al., 1995; Lee et al., 1995; Meyer and Birchmeier, cells was inferred long ago on the basis of Schwannoma
1995; Kramer et al., 1996). Homozygous embryos die in induction by an oncogenic mutant of the rodent ErbB-2.
utero at around embryonic day 10, presumably due to In cell culture, NRGs are the only factors able to mimic
defects resulting from the absence of heart trabeculae, neurites in supporting survival of Schwann cell precur-
the finger-like extensions of the heart’s wall. The only sors, and a soluble ErbB-4 can block neuronal support
previous clue for involvement of NRGs in cardiac devel- (K. Jessen, University College, London). The precursors
opment was derived from expression studies that local- first appear around embryonic day 12.5 in the mouse,
ized NRG to the endothelial cells of the endocardium and and by embryonic day 15, they complete differentiation
both ErbB-2 and ErbB-4 to the adjacent heart muscle of to Schwann cells expressing the calcium binding protein
the ventricular wall, the myocardium. In the absence of S100 and the transcription factor Krox-20 (Murphy et al.,
any of the three components, the inductive effect of the 1996). The latter, along with the cyclic-AMP responsive
endocardium on the myocardium is defective: the wall element binding protein (CREB), is activated by NRG in
does not protrude into the chamber to form the trabecu- Schwann cells. Consistent with the role of NRG in precur-
lae. Interestingly, inactivation of NRG in chick embryos, sor survival, mice lacking NRG expression displayed se-
using a novel ribozyme-based antisense strategy, verely reduced numbers of presumptive Schwann cell pre-
resulted in trabeculation defects similar to those ob- cursors along trunk projections of nerves that normally
served in mice (Lemke). The short-range inductive action express NRG and become ensheathed by Schwann cells.
of NRG in mammalian heart development absolutely Unlike mice that are homozygous for the EGF-like do-
depends on the presence of both the receptor, ErbB-4, main of NRG (NRGEGF2/2), mice that are homozygous
and the coreceptor, ErbB-2. Thus, contrary to in vitro mutant in the Ig-like domain (NRGIg2/ 2) but express other
studies implying that ErbB-4 can transmit signals in the NRG isoforms, contain Schwann cells along axonal pro-
absence of ErbB-2, homodimers of ErbB-4 are inactive jections of cranial nerves (C. Birchmeier). These results
in ventricle differentiation. indicate that Schwann cells do not depend on the Ig-
The other direct NRG receptor, ErbB-3, is not involved like isoform of NRG. Since the most abundant NRG iso-
in trabeculation as its expression is restricted to the form in motor and sensory neurons, CRD-NRG, lacks
mesenchyme of the endocardial cushion, which devel- the Ig domain, but instead has a CRD (Yang and Role),
ops into valves. Indeed, the hearts of ErbB-32/2 mice it seems likely that isoforms containing the CRD are
display delayed but otherwise normal trabeculation, but sufficient to initiate Schwann cell differentiation. Inter-
their atrioventricular valves are rudimentary and thinned, estingly, Schwann cells are present in trigeminal and
leading to death in utero at E13.5 (S. Erickson, Genen- dorsal root ganglia of ErbB-32/2 animals (E13.5), but
tech). As the ErbB-3-defective mice survive longer than their numbers are reduced in both the ganglia and their
the other mutants, many additional sites of NRG-ErbB-3 nerve projections (Erickson). In addition, sciatic nerves
functions were observed in these embryos (see below). from adult ErbB-31/2 mice displayed reduced conduc-
The endocardium-derived NRG mediates formation of tion velocity, in agreement with a Schwann cell defect.
cushion mesenchyme, from which valve primordia evolve. On maturation, Schwann cells respond to NRGs by
The identity of the ErbB-3 coreceptor in valvuloseptal enhanced proliferation, probably reflecting the ability of
differentiation is unknown, as neither ErbB-1 nor ErbB-2 axons to maintain an appropriate ratio to myelin-forming
are expressed in the cushion. Thus, two different as- Schwann cells (Syroid et al., 1996). Although mature
pects of cardiac development, trabeculation and valvu- Schwann cells synthesize certain transmembrane iso-
loseptal formation, are critically regulated by distinct forms of NRG and display constitutive phosphorylation
Meeting Review
851

of ErbB-3 (Rosenbaum et al., 1997), factor concentration during the disease reduced the frequency of clinical
may not be sufficiently high to maintain proliferation via relapses. The mechanism of action is unclear; indeed,
an autocrine mechanism (Ratner), indicating that other it is possible that NRG acts indirectly on immune cells
factors may have a role in regulating postnatal cells. rather than directly on oligodendrocytes. Nevertheless,
Nevertheless, NRG at low concentrations increases di- NRG clearly enhanced the extent of myelination in these
rected migration of Schwann cells cultured in vitro on mice, suggesting the need for further study of a potential
peripheral nerves (Mahanthappa et al., 1996). therapeutic role for NRG in multiple sclerosis.
NRG expression and NRG-mediated signaling change An interesting interplay between NRG and sonic
following nerve injury. Wallerian degeneration of the sci- hedgehog (Shh) is emerging from studies of oligoden-
atic nerve causes Schwann cells to up-regulate expres- drocyte development in the chick spinal cord (R.H.
sion of certain NRG isoforms, along with ErbB-3 and Miller, Case Western Reserve University, Cleveland).
ErbB-2 (Carroll et al., 1997). Because these Schwann Since oligodendrocyte precursors are located in the
cells are more intensely stained with antibodies specific ventral neural tube, oligodendrocytes do not develop in
for a major tyrosine autophosphorylation site in ErbB-2 explants of the dorsal portion of the tube. Although nei-
(C.D. Stiles, Dana Farber Cancer Institute, Boston), ther NRG nor low concentrations of Shh could induce
nerve degeneration is apparently accompanied by in- oligodendrocytes in explants of dorsal neural tube, the
creased NRG signaling. Schwann cells at the neuromus- two ligands together collaborated to induce oligoden-
cular synapse provide another example for a trophic drocyte differentiation in these explants.
neural effect, as will be discussed below.
Like Schwann cells, differentiation of oligodendro-
cytes, the myelin-forming cells of the central nervous NRGs in the Neuromuscular Synapse
system, is profoundly influenced by neurons. While mi- NRGs play an important role at neuromuscular syn-
grating from the subventricular zone to tracts of the white apses, where they are thought to act as a signal that
matter, the progenitor cell progressively assumes a ma- stimulates transcription of certain genes, including
ture oligodendrocyte phenotype. J. Salzer (New York those encoding acetylcholine receptor (AChR) subunits,
University) reported that NRG is mitogenic to both cell in nuclei near the synaptic site. The first in vivo evidence
types in vitro. Moreover, NRG reverses differentiation of supporting this notion was presented by G. Fischbach
myelin-forming oligodendrocytes, and under certain in (Harvard Medical School), who reported that mice that
vitro conditions, it can inhibit commitment to both an oligo- are heterozygous for the Ig allele of NRG (NRGIg1/2) are
dendrocyte lineage (at the pro-oligodendrocyte stage) and mildly myasthenic. These heterozygous mice have fewer
to type 2 astrocytes. Strikingly, NRG similarly inhibits (50%) AChRs at their neuromuscular synapses, support-
expression of myelin-specific proteins by Schwann cells ing the idea that NRG has a role in stimulating AChR
(A. Mudge, University College, London; Cheng and synthesis in synaptic nuclei, and that the concentration
Mudge, 1996). While these activities should be verified of NRG at the synapse is limiting. The mutant mice
in vivo, they suggest that NRG has a common role in apparently compensate for the deficit in AChR number
the myelinating cells of both the central and peripheral by releasing more acetylcholine from their nerve termi-
nervous systems. It enables neurons to enlarge the pool nals, measured as an increase in quantal content, and
of premyelinating glial cells, and only after NRG-ErbB this result may explain why the heterozygous mice are
signaling has been attenuated can myelination proceed. behaviorally normal. Nevertheless, tetanic stimulation
Indeed, studies presented by J. Grinspan and S. Scherer of their motor nerves in vitro showed that synaptic trans-
(University of Pennsylvania, Philadelphia) indicate that mission fails more readily in NRGIg1/ 2 mice, further indi-
the pool of premyelinated Schwann cells is the one regu- cating that NRG has a role in neuromuscular function.
lated by axonal NRG during early postnatal develop- The Ig isoform of NRG is not the major NRG isoform
ment. After axotomy, which removes the source of axonal in motor neurons, and indeed the spinal cord of NRGIg 1/2
NRG, they observed enhanced apoptosis of premy- mice have nearly normal levels of NRG. Since the Ig-
elinating Schwann cells, and the increase in apoptosis like domain is crucial for binding to heparan sulfate pro-
was completely prevented by the application of exoge- teoglycans, it may promote accumulation of this isoform
nous NRG (Grinspan et al., 1996). Similar rescue of in the extracellular matrix of the synaptic cleft, even
Schwann cells was observed at neuromuscular junc- during stages of development in which the abundance
tions, Golgi tendon organs, and Pacinian corpuscle (W. of this mRNA is relatively low (Fischbach). An alternative
Thompson, University of Texas, Austin). explanation for the apparent paradox of reduced neuro-
The interaction of NRG with myelinating cells has im- muscular function in mice containing nearly normal lev-
plications for demyelinating disorders of the nervous els of NRG in their spinal cord was raised by S. Burden
system. Evidence was presented indicating that NRG (Skirball Institute, New York). NRG is synthesized not
may have a clinically beneficial role as a therapeutic only by motor neurons but also by skeletal muscle cells
agent for multiple sclerosis. C. Raine (Albert Einstein (Moscoso et al., 1995; Rimer et al., 1996, Soc. Neurosci.
College of Medicine, New York) used an adoptive trans- abstract), and the persistence of the membrane-bound
fer model of experimental allergic encephalomyelitis form of NRG at denervated synapses demonstrates that
(EAE) to determine whether NRG might reduce the rate muscle cells contribute NRG to the synaptic site (Rimer
or extent of demyelination. In this model, clinical re- et al., 1996, Soc. Neurosci. abstract). Because the Ig iso-
lapses are episodic, and they are associated with inflam- form is the major NRG isoform in skeletal muscle cells
mation, demyelination, and remyelination. Raine re- (X. Yang and Burden), the phenotype of the NRGIg1/ 2
ported that repeated injections of NRG at different times mice may be explained by a reduction in muscle-derived
Neuron
852

rather than in motor neuron-derived NRG. Moreover, NRG Actions on Neurons and Cranial Nerves
since muscle cells synthesize NRG and ErbBs, and local- Because NRGs and their direct receptors are widely
ize both ligands and receptors to the synaptic site, NRG- expressed by neurons of the central and peripheral ner-
ErbB signaling at the synapse may function, at least vous systems, paracrine and autocrine regulation of
in part, by an autocrine mechanism (Burden), involving nerves by NRG is possible. Disruption of all NRG iso-
activation of a Ras/ERK2-dependent pathway (Altiok et forms, or only Ig-containing isoforms, resulted, however,
al., 1997). in neural defects that were limited to cranial nerves at
Agrin is an important signal for neuromuscular syn- the time of death of mutant animals. Specifically, the
apse formation, since mice lacking agrin or MuSK, a neural crest–derived component of cranial ganglia V (tri-
component of its receptor complex, lack neuromuscular geminal), and to a lesser extent that of ganglia VII–IX,
synapses. Because mice lacking agrin (Gautam et al., were severely reduced in size. In addition, these ganglia
1996) or MuSK (DeChiara et al., 1996) fail to cluster were disconnected, either completely or partially, from
ErbBs, NRG-ErbB signaling is likely to depend on agrin/ the hindbrain. Apparently, NRG controls survival, rather
MuSK signaling. Agrin is sufficient to cluster NRG and than migration, of cells originating in the NRG-express-
ErbBs in skeletal muscle, since forced ectopic expres- ing hindbrain rhombomeres 2, 4, and 6 (Meyer and Birch-
sion of neural agrin at nonsynaptic sites in skeletal mus- meier, 1995). These neural crest–derived cells are al-
cle cells caused not only AChRs but also ErbBs and ready committed to a neural fate, and their survival
pro-NRG to cluster at the ectopic site (Rimer et al., 1996, specifically depends on an Ig-containing NRG (C. Birch-
Soc. Neurosci. abstract). Thus, agrin/MuSK signaling meier). Survival is mediated by ErbB-2 and ErbB-3, but
regulates the distribution of muscle-derived NRG and not an active ErbB-4, as ErbB-22/2 and ErbB-32/2mice
ErbBs. shared a similar cranial nerve phenotype with NRG2/2
Identification of an ErbB-binding protein, which, like mice (Lee; Erickson), but both ganglia size and hindbrain
the ErbBs, is concentrated at neuromuscular synapses, connections were not affected in ErbB-42/ 2 mice (M.
was described by K. Carraway (Harvard). This heregulin Gassman, Salk Institute). Instead, in the latter animals,
receptor binding (HRB) protein, identified in a yeast two- ganglia V, VII, and VIII are almost fused, and their con-
hybrid screen using the cytoplasmic domain of ErbB-3 nections are mistargeted: the trigeminal and facial gan-
as bait, is expressed in heart, skeletal muscle, and brain. glia are each connected to both rhombomeres 2 and 4,
The protein is myristolated, has an imperfect ring finger instead of their normal single links to rhombomeres 2
domain, and can bind both ErbB-3 and ErbB-4 in vitro, and 4, respectively. The reason for mistargeting is cur-
although the binding does not appear to depend on rently unknown, but it probably relates to the localized
ErbB phosphorylation. Like rapsyn-induced clustering expression of ErbB-4 in the intervening odd numbered
of AChRs in nonmuscle cells, HRB appears to regulate rhombomeres 3 and 5 (Lemke, 1996). Alternatively, mis-
the clustering of ErbB-3 in transfected COS cells. targeting is not seen in NRG2/2 embryos as a result of the
The role of NRG in regulating the nonmyelinating ter- extensive cell loss in their cranial ganglia (Gassmann).
minal Schwann cells at the neuromuscular synapse was Another possible explanation for the discordant NRG
addressed by W. Thompson. In mammalian muscle, syn- and ErbB-4 phenotypes is the existence of additional
aptic sites are usually organized in an endplate band ErbB-4 ligands, such as betacellulin (Riese et al., 1996),
adjacent to the main intramuscular nerve. Schwann cells HB-EGF, or NRG2. Alternatively, reciprocal mechanisms
within skeletal muscle are restricted to these intramus- involving other neurotrophic factors may be involved.
cular nerves and to synaptic sites. Injection of NRG into For example, sympathetic neuroblast-derived NRG can
skeletal muscle of neonatal mice, however, resulted in induce synthesis of nerotrophin-3 (NT-3) by nonneuronal
the proliferation and redistribution of Schwann cells cells, thereby affecting in an indirect way the survival
throughout the muscle. Motor axons in NRG-injected of neurons (Verdi et al., 1996). Potentially, this or a similar
muscle are likewise found throughout the muscle. Since mechanism may underlie the observation that NRG pro-
prior studies by Thompson indicated that Schwann cells motes survival and neurite outgrowth of cultured embry-
are likely to have an important role in guiding motor onic retinal cells (O. Bermingham-McDonogh, University
axons to denervated synaptic sites (Son and Thompson, of Washington, Seattle). Neurons isolated from neonates
1995), these new results raise the possibility that NRG, (P4) lost responsiveness to NRG, in parallel to a de-
possibly provided by skeletal muscle, indirectly regu- creased expression of ErbB-2 (Bermingham-McDonogh
lates the position of motor axons by regulating the num- et al., 1996). Thus, neurite outgrowth in retinal cells, as
ber and position of Schwann cells in muscle. well as in the more extensively studied PC-12 cell line
Although NRG2, like NRG1, binds to ErbB-3 and to (Gamett et al., 1995), correlates with ErbB-2 expression,
ErbB-4 and concomitantly stimulates their association reemphasizing a receptor-coded specificity biological
with other ErbB family members, resulting in ligand- effect of NRGs.
stimulated tyrosine phosphorylation, NRG1, but not Reciprocal interactions involving NRG also play a role
NRG2, activates expression of AChR genes in muscle in neural migration. E. Anton (Yale) and G. Corfas (Har-
cell lines (Lai, Yang, and Burden; A. Goodearl, Millenium vard; Rio et al., 1997) studied migration in the cerebral
Pharmaceuticals). Similarly, NRG1 but not NRG2 stimu- cortex and in the cerebellum, respectively, and con-
lates Schwann cell proliferation. These unexpected re- cluded that neuron-derived NRG instructs the underly-
sults raise the possibility that the extent or duration of ing glia, either radial glia or the cerebellar Bergmann
signaling by NRG1 and NRG2 may differ and account glia, to promote directed neuron movement. In the cortex,
for the different biological responses observed in mus- glia-expressed ErbB-2 is involved, perhaps in collabora-
cle and in Schwann cells. tion with ErbB-3 and ErbB-4; in Bergmann glia, NRG
Meeting Review
853

induced CREB phosphorylation. Migration could be during puberty, and also promoted mammary tumors
blocked either by an anti-NRG antibody added to ex- (Krane and Leder, 1996). Thus, NRGs appear to induce
plants of the cerebral cortex or by a dominant-negative growth and inhibit differentiation of the primitive epithe-
mutant of ErbB-4 ectopically expressed in glia cells. In lial duct, but later in development these factors enhance
both the developing cerebral cortex and cerebellum, mammary differentiation.
NRG promoted the neuron-primed elongation of glial The reason that epithelial defects are not prominently
cells, but not their adhesion to neuronal cells, indicating detectable in NRG-, ErbB-2-, and ErbB-4-targeted mice
that NRG directly induces radial morphology of the glia, is probably due to the early embryonic death of the
and this cell reciprocally promotes neuron migration mutant mice, and the relatively late onset of epithelial
through an unknown mechanism. organogenesis. Indeed, major defects in epithelia of
Acoustic trauma (exposure to excessive noise for a skin, lung, hair, placenta, and other organs were de-
long time, or to certain antibiotics and diuretics) may tected in ErbB-1-defective mice (which live longer than
lead to loss of mechanosensory hair cells from the co- NRG2/2 mice). Likewise, ErbB-3-defective mice (which
chlea, resulting in irreversible deficits in hearing. In cer- live 3 days longer than NRG2/2 mice) already display
tain species, these damaged epithelial cells can be re- some abnormalities in stomach and in pancreas (Erick-
placed by division of supporting cells, which share some son). Another reflection of the role of ErbB-3 in epithelial
features with Schwann cells. Jeff Corwin (University of differentiation was derived from an in vivo model of
Virginia, Charlottesville) reported that postnatal rat sup- wound healing, where NRG was found to mediate both
porting cells undergo mitosis in response to NRG, and epidermal migration and up-regulation of keratinocyte
to a lesser extent to one of the ErbB-1 ligands (TGFa), differentiation markers (Danilenko et al., 1995). As ex-
suggesting yet another clinical application of these pected from other inductive actions of NRG, the ligand
growth factors. is secreted by skin fibroblasts and affects nearby kera-
tinocytes, whose expression of ErbB proteins changes
during reepithelialization.
NRGs in Epithelial Morphogenesis The dynamic pattern of ErbB expression during cell
The wealth of information on NRGs as primary mediators fate determination and morphogenic processes appar-
of nerve communication with neighboring nonneuronal ently dictates the nature of cellular response. This ap-
cells may be paralleled by initial reports of similar inter- pears to be true in all in vitro models of NRG actions,
actions in other epithelial organs, whose cellular hetero- including Schwann cells. As oligodendrocytes differenti-
geneity and complex anatomical connections are reminis- ate, ErbB-4 is down-regulated, and ErbB-2 expression
cent of the nervous system. Morphogenesis of epithelial is increased (Canoll et al., 1996), whereas during skeletal
organs is controlled, in part, by multiple polypeptide muscle differentiation, ErbB-3 expression is strongly in-
factors secreted by the underlying stromal mesen- duced while ErbB-2 expression remains unchanged (Jo
chyme. With the exception of ErbB-4, whose expression et al., 1995). Dramatic examples of how the repertoire
is limited to certain epithelia, the three other receptors of ErbB proteins might affect cell function are provided
and coreceptors of NRGs are present in most epithelial by several types of epithelial cancers (carcinomas):
cell layers. Moreover, mesenchymal cells are the richest some tumors overexpress ErbB-2 at levels 100-fold
source, outside of the nervous system, of the corre- greater than normal, and in some types of cancers, the
sponding ligands, both NRGs and ErbB-1-specific li- extent of overexpression predicts disease outcome and
gands. An example of NRG action in mesenchyme– response to hormone- and chemotherapy. Apparently,
epithelial interactions is provided by the mammary gland ErbB-2 overexpression in epithelial cells biases forma-
(C., and W. Birchmeier, Max Delbrück Center, Berlin). tion of its more potent heterodimers, leading to en-
The gland is one of a few organs in which major morpho- hanced proliferation at the expense of a differentiated
genesis takes place after birth. During puberty, it in- phenotype. On the other hand, blocking ErbB-2 action
volves branching and elongation that result in a ductal by using anti-ErbB-2 monoclonal antibodies results in
tree. A second budding phase, during pregnancy, gener- retardation of cell proliferation, an effect that has been
ates the milk-producing lobuloalveolar structures. Anal- recently used in breast cancer patients to inhibit mam-
yses of organ cultures of the mouse mammary gland mary tumor growth (Baselga et al., 1996).
suggest that NRG, whose expression peaks at preg-
nancy, regulates the second morphogenic phase by Conclusions
stimulating lobuloalveolar budding and milk production. Results obtained in the few years since the NRG family
Of note, synthesis of milk components (e.g., casein and was discovered already make it clear that this signaling
neutral lipids) in response to NRG is retained in vitro by module is involved in a myriad of physiological and de-
certain mammary cell lines. velopmental processes, as well as in pathological
Interestingly, when pellets releasing NRG were im- states. This multiplicity of biological functions likely re-
planted within the mammary glands of prepubescent flects the enormous biochemical potential of the NRG-
female mice, NRG-a, but not NRG-b, could induce ductal ErbB network to diversify signals. The available NRG2
branching and migration in the absence of exogenous isoforms, together with the possible existence of addi-
steroid hormones, but both isoforms were active in in- tional ErbB receptors and a direct ErbB-2 ligand, may
duction of secretory lobuloalveoli (Jones et al., 1996). extend the ensuing complexity even further. Results
Mice engineered to constitutively overexpress NRG-b2 from mice with targeted mutations support the possibil-
in the mammary gland displayed persistence of the ity that this signaling complexity, which has been estab-
mammary end bud structures, which normally disappear lished by expression studies and in vitro experiments, is
Neuron
854

utilized in vivo. Thus, in the near future, we will probably Gautam, M., Noakes, P.G., Moscoso, L., Rupp, F., Scheller, R., Mer-
lie, J.P., and Sanes, J.R. (1996). Defective neuromuscular synapto-
witness parallel biochemical and physiological decod-
genesis in agrin-deficient mutant mice. Cell 85, 525–535.
ing of the many distinct signaling pathways employed
Graus-Porta, D., Beerli, R.R., and Hynes, N.E. (1995). Single-chain
by ErbB proteins. Conceivably, lessons learned in this
antibody-mediated intracellular retention of ErbB-2 impairs Neu dif-
subfamily of growth factor receptors will be applicable ferentiation factor and epidermal growth factor signaling. Mol. Cell.
to other subtypes of receptor tyrosine kinases. Biol. 15, 1182–1191.
Grimm, S., and Leder, P. (1997). An apoptosis-inducing isoform of
Neu differentiation factor (NDF) identified using a novel screen for
Acknowledgments
dominant, apoptosis-inducing genes. J. Exp. Med. 185, 1137–1142.
Grinspan, J.B., Marchionni, M.A., Reeves, M., Coulaloglou, M., and
We thank the organizers of the workshop and many participants for
Scherer, S.S. (1996). Axonal interactions regulate Schwann cell
helpful comments, and Iris Alroy for help with the figure.
apoptosis in developing peripheral nerve: neuregulin receptors and
the role of neureguins. J. Neurosci. 16, 6107–6118.
References Ho, W.H., Armanini, M.P., Nuijens, A., Phillips, H.S., and Osheroff,
P.L. (1995). Sensory and motor neuron-derived factor. A novel here-
Altiok, N., Altiok, X., and Changeux, J.P. (1997). Heregulin-stimulated gulin variant highly expressed in sensory and motor neurons. J. Biol.
acetylcholine receptor gene expression in muscle: requirement for Chem. 270, 14523–14532.
MAP kinase and evidence for a parallel inhibitory pathway indepen- Jacobsen, N.E., Abadi, N., Sliwkowski, M.X., Reilly, D., Skelton, N.J.,
dent of electrical activity. EMBO J. 16, 717–725. and Fairbrother, W.J. (1996). High-resolution solution structure of
Barbacci, E.G., Guarino, B.C., Stroh, J.G., Singleton, D.H., Rosnack, the EGF-like domain of heregulin-alpha. Biochemistry 35, 3402–
K.J., Moyer, J.D., and Andrews, G.C. (1995). The structural basis for 3417.
the specificity of epidermal growth factor and heregulin binding. J. Jo, S.A., Zhu, X., Marchionni, M.A., and Burden, S.J. (1995). Neuregu-
Biol. Chem. 270, 9585–9589. lins are concentrated in nerve-muscle synapses and activate Ach-
Baselga, J., Tripathy, D., Mendelsohn, J., Baughman, S., Benz, C.C., receptor gene expression. Nature 373, 158–161.
Dantis, L., Sklarin, N.T., Seidman, A.D., Hudis, C.A., Moore, J., Ro- Jones, F.E., Jerry, D.J., Guarino, B.C., Andrews, G.C., and Stern,
sen, P.P., Twaddell, T., Henderson, I.C., and Norton, L. (1996). Phase D.F. (1996). Heregulin induces in vivo proliferation and differentiation
II study of weekly intravenous recombinant humanized anti-p185 of mammary epithelium into secretory lobuloalveoli. Cell Growth
HER2 monoclonal antibody in patients with HER2/neu-overexpress- Diff. 7, 1031–1038.
ing metastatic breast cancer. J. Clin. Oncol. 14, 737–744. Karunagaran, D., Tzahar, E., Beerli, R.R., Chen, X., Graus-Porta, D.,
Baulida, J., Kraus, M.H., Alimandi, M., Di Fiore, P.P., and Carpenter, Ratzkin, B.J., Seger, R., Hynes, N.E., and Yarden, Y. (1996). ErbB-2 is
G. (1996). All ErbB receptors other than the epidermal growth factor a common auxiliary subunit of NDF and EGF receptors: implications
receptor are endocytosis impaired. J. Biol. Chem. 271, 5251–5257. for breast cancer. EMBO J. 15, 254–264.
Bermingham-McDonogh, O., McCabe, K.L., and Reh, T.A. (1996). Kornfeld, K. (1997). Vulval development in Caenorbaditis elegans.
Effects of GGF/neuregulins on neuronal survival and neurite out- Trends Genetics 13, 55–61.
growth correlate with erbB2/neu expression in developing rat retina. Kramer, R., Bucay, N., Kane, D.J., Martin, L.E., Tarpley, J.E., and
Development 122, 1427–1438. Theill, L.E. (1996). Neuregulins with an Ig-like domain are essential
Canoll, P.D., Musacchio, J.M., Hardy, R., Reynolds, R., Marchionni, for mouse myocardial and neuronal development. Proc. Natl. Acad.
M.A., and Salzer, J.L. (1996). GGF/neuregulin is a neuronal signal Sci. USA 93, 4833–4838.
that promotes the proliferation and survival and inhibits the differen- Krane, I.M., and Leder, P. (1996). NDF/heregulin induces persistence
tiation of oligodendrocyte progenitors. Neuron 17, 229–243. of terminal end buds and adenocarcinomas in the mammary glands
Carroll, S.L., Miller, M.L., Fohnert, P.W., Kim, S.S., and Corbett, of transgenic mice. Oncogene 12, 1781–1788.
J.A. (1997). Expression of neuregulins and their putative receptors, Lee, K.F., Simon, H., Chen, H., Bates, B., Hung, M.C., and Hauser,
ErbB2 and ErbB3, is induced during Wallerian degeneration. J. Neu- C. (1995). Requirement for neuregulin receptor erbB2 in neural and
rosc. 17, 1642–1659. cardiac development. Nature 378, 394–398.
Cheng, L., and Mudge, A.W. (1996). Cultured Schwann cells consti- Lemke, G. (1996). Neuregulins in development. Mol. Cell. Neurosci.
tutively express the myelin protein P0. Neuron 16, 309–319. 7, 247–262.
Cohen, B.D., Kiener, P.K., Green, J.M., Foy, L., Fell, H.P., and Zhang, Lemmon, M.A., Bu, Z., Ladbury, J.E., Zhou, M., Pinchasi, D., Lax,
K. (1996). The relationship between human epidermal growth-like I., Engelman, D.E., and Schlessinger, J. (1997). Two EGF molecules
factor receptor expression and cellular transformation in NIH-3T3 contribute additively to stabilization of the EGFR dimer. EMBO J.
cells. J. Biol. Chem. 271, 30897–30903. 16, 281–294.
Danilenko, D.M., Ring, B.D., Lu, J.Z., Tarpley, J.E., Chang, D., Liu, Levi, A.D., Bunge, R.P., Lofgren, J.A., Meima, L., Hefti, F., Nikolics,
N., Wen, D., and Pierce, G.F. (1995). Neu differentiation factor upreg- K., and Sliwkowski, M.X. (1995). The influence of heregulins on hu-
ulates epidermal migration and integrin expression in excisional man Schwann cell proliferation. J. Neurosci. 15, 1329–1340.
wounds. J. Clin. Invest. 95, 842–851. Levkowitz, G., Klapper, L.N., Tzahar, E., Freywald, A., Sela, M., and
DeChiara, T.M., Bowmen, D.C., Valenzuela, D.M., Simmons, M.V., Yarden, Y. (1996). Coupling of the c-Cbl protooncogene product to
Poueymirou, W.T., Thomas, S., Kinetz, E., Copton, D.L.R.E., Park, ErbB-1/EGF-receptor but not to other ErbB proteins. Oncogene 12,
S.J., Smith, S., DiStefano, P.S., Glass, D.J., Burden, S.J., and Yanco- 1117–1125.
poulos, G.D. (1996). The receptor tyrosine kinase MuSK is required Mahanthappa, N.K., Anton, E.S., and Matthew, W.D. (1996). Glial
for neuromuscular junction formation in vivo. Cell 85, 501–512. growth factor 2, a soluble neuregulin, directly increases Schwann
Elenius, K., Paul, S.G.A., Sun, J., and Klagsbrun, M. (1997). Activation cell motility and indirectly promotes neurite outgrowth. J. Neurosci.
of HER4 by heparin-binding EGF-like growth factor stimulates che- 16, 4673–4683.
motaxis but not proliferation. EMBO J. 16, 1268–1278. Meyer, D., and Birchmeier, C. (1995). Multiple essential functions of
Gamett, D.C., Greene, T., Wagreich, A.R., Kim, H.H., Koland, J.G., neuregulin in development. Nature 378, 386–390.
and Cerione, R.A. (1995). Heregulin-stimulated signaling in rat pheo- Moscoso, L.M., Chu, G.C., Gautam, M., Noakes, P.G., Merlie, J.P.,
chromocytoma cells. Evidence for ErbB3 interactions with Neu/ and Sanes, J.R. (1995). Synapse-associated expression of an acetyl-
ErbB2 and p85. J. Biol. Chem. 270, 19022–19027. choline receptor-inducing protein, ARIA/heregulin, and its putative
Gassmann, M., Casagranda, F., Orioli, D., Simon, H., Lai, C., Klein, receptors, ErbB2 and ErbB3, in developing mammalian muscle. Dev.
R., and Lemke, G. (1995). Aberrant neural and cardiac development Biol. 172, 158–169.
in mice lacking the ErbB4 neuregulin receptor. Nature 378, 390–394. Murphy, P., Topilko, P., Schneider-Maunoury, S., Seitanidou, T.,
Meeting Review
855

Evercooren, B.V., and Charnay, P. (1996). The regulation of Krox-


20 expression reveals important steps in the control of peripheral
glial cell development. Development 122, 2847–2857.
Perrimon, N., and Perkins, L.A. (1997). There must be 50 ways to
rule the signal: the case of the Drosophila EGF receptor. Cell 89,
13–16.
Pinkas-Kramarski, R., Shelly, M., Glathe, S., Ratzkin, B.J., and Yar-
den, Y. (1996a). Neu differentiation factor/neuregulin isoforms acti-
vate distinct receptor combinations. J. Biol. Chem. 271, 19029–
19032.
Pinkas-Kramarski, R., Soussan, L., Waterman, H., Levkowitz, G.,
Alroy, I., Klapper, L., Lavi, S., Seger, R., Ratzkin, B., Sela, M., and
Yarden, Y. (1996b). Diversification of Neu differentiation factor and
epidermal growth factor signaling by combinatorial receptor interac-
tions. EMBO J. 15, 2452–2467.
Riese, D.J., van Raaij, T.M., Plowman, G.D., Andrews, G.C., and
Stern, D.F. (1995). The cellular response to neuregulins is governed
by complex interactions of the ErbB receptor family. Mol. Cell. Biol.
15, 5770–5776.
Riese, D.J., Bermingham, Y., van Raaij, T.M., Buckley, S., Plowman,
G.D., and Stern, D.F. (1996). Betacellulin activates the epidermal
growth factor receptor and erbB-4, and induces cellular response
patterns distinct from those stimulated by epidermal growth factor
or neuregulin-beta. Oncogene 12, 345–353.
Rio, C., Rieff, H.I., Qi, P., and Corfas, G. (1997). Neuregulin and erbB
receptors play a critical role in neuronal migration. Neuron 19, in
press.
Rosenbaum, C., Karyala, S., Marchionni, M.A., Kim, H.A., Krasnosel-
sky, A.L., Happel, B., Isaacs, I., Brackenbury, R., and Ratner, N.
(1997). Schwann cells express NDF and SMDF/n-ARIA mRNAs, se-
crete neuregulin, and show constitutive activation of erbB3 recep-
tors: evience for a neuregulin autocrine loop. J. Exp. Neurol., in
press.
Son, Y.J., and Thompson, W.J. (1995). Nerve sprouting in muscle
is induced and guided by processes extended by Schwann cells.
Neuron 14, 133–141.
Summerfield, A.E., Hudnall, A.K., Lukas, T.J., Guyer, C.A., and
Staros, J.V. (1996). Identification of residues of the epidermal growth
factor receptor proximal to residue 45 of bound epidermal growth
factor. J. Biol. Chem. 271, 19656–19659.
Syroid, D.E., Maycox, P.R., Burrola, G.P., Liu, N., We, D., Lee, K.-F.,
Lemke, G., and Kilpatrick, T.J. (1996). Cell death in the Schwann
cell lineage and its regulation by neuregulin. Proc. Natl. Acad. Sci.
USA 93, 9229–9234.
Tzahar, E., Waterman, H., Chen, X., Levkowitz, G., Karunagaran, D.,
Lavi, S., Ratzkin, B.J., and Yarden, Y. (1996). A hierarchical network
of inter-receptor interactions determines signal transduction by
NDF/neuregulin and EGF. Mol. Cell. Biol. 16, 5276–5287.
Verdi, J.M., Groves, A.K., Farinas, I., Jones, K., Marchionni, M.A.,
Reichardt, L.F., and Anderson, D.J. (1996). A reciprocal cell–cell
interaction mediated by NT-3 and neuregulins controls the early
survival and development of sympathetic neuroblasts. Neuron 16,
515–527.
Zhang, K., Sun, J., Liu, N., Wen, D., Chang, D., Thomason, A., and
Yoshinaga, S.K. (1996). Transformation of NIH 3T3 cells by HER3
or HER4 receptors requires the presence of HER1 or HER2. J. Biol.
Chem. 271, 3884–3890.

You might also like