<<

Oncogene (2007) 26, 3122–3142 & 2007 Nature Publishing Group All rights reserved 0950-9232/07 $30.00 www.nature.com/onc REVIEW G regulation of MAPK networks

ZG Goldsmith and DN Dhanasekaran

Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA, USA

G provide signal-coupling mechanisms to hepta- the a-subunits has been used as a basis for the helical surface receptors and are criticallyinvolved classification of G proteins into Gs,Gi,Gq and G12 in the regulation of different -activated protein families in which the a-subunits that show more than (MAPK) networks. The four classes of G proteins, 50% homology are grouped together (Simon et al., defined bythe G s,Gi,Gq and G12 families, regulate 1991). In G-protein-coupled receptor (GPCR)-mediated ERK1/2, JNK, p38MAPK, ERK5 and ERK6 modules by signaling pathways, ligand-activated receptors catalyse different mechanisms. The a- as well as bc-subunits are the exchange of the bound GDP to GTP in the a-subunit involved in the regulation of these MAPK modules in a following which the GTP-bound a-subunit disassociate context-specific manner. While the a- and bc-subunits from the receptor as well as the bg-subunit. The GTP- primarilyregulate the MAPK pathwaysvia their respec- bound a-subunit and the bg-subunit stimulate distinct tive effector-mediated signaling pathways, recent studies downstream effectors including , ion channels have unraveled several novel signaling intermediates and small GTPase, thus regulating multiple signaling including receptor and small pathways including those involved in the activation of through which these G-protein subunits positivelyas well mitogen-activated (MAPK) modules as negativelyregulate specific MAPK modules. Multiple (Johnson and Dhanasekaran, 1989; Hepler and Gilman, mechanisms together with specific scaffold proteins that 1992; Dhanasekaran et al., 1995; Dhanasekaran and can link G-protein-coupled receptors or G proteins to Prasad, 1998; Rozengurt, 1998). distinct MAPK modules contribute to the context-specific MAPK modules regulate many critical signaling and spatio-temporal regulation of mitogen-activated pathways including cell proliferation, differentiation protein signaling networks byG proteins. and (Fanger et al., 1997; Minden and Karin, Oncogene (2007) 26, 3122–3142. doi:10.1038/sj.onc.1210407 1997; Dhanasekaran and Premkumar Reddy, 1998; Gutkind, 1998; Garrington and Johnson, 1999; Davis, Keywords: GPCR; ; ; 2000; Chang and Karin, 2001). Each of these modules MAPK; oncogene; cancer consists of at least three distinct kinases, namely an upstream mitogen-activated protein (MAP) kinase kinase kinase (MAP3K), a MAP kinase kinase (MAP2K) and a downstream MAP kinase (MAPK). In a typical signaling pathway, growth factor stimulated Introduction small GTPases such as Ras, CDC42, and Rac activate an upstream MAP kinase kinase kinase kinase Heterotrimericguanine nucleotide-binding proteins, (MAP4K), following which the MAP kinase cascades or G proteins, consisting of a-, b-andg-subunits, proceed through the sequential phosphorylation of transduce signals from heptahelical cell surface receptors constituent MAP3K, MAP2K and MAPK (Minden to intracellular effectors (Simon et al., 1991; Hepler and Karin, 1997; Dhanasekaran and Premkumar and Gilman, 1992; Conklin and Bourne, 1993). The Reddy, 1998; Davis, 2000; Chang and Karin, 2001). a-subunit is a 37–42 kDa protein, which contains Activated MAP kinases translocate from the to the guanine nucleotide-binding pocket along with the the nucleus where they regulate the activities of various intrinsicGTPase activity.The 35-kDa b-subunit and the transcription factors through phosphorylation (Khokh- 8–11-kDa g-subunit are tightly associated with each latchev et al., 1998; Brunet et al., 1999). GPCRs, other and are often referred to as a single entity, the primarily through their cognate G proteins, regulate bg-subunit (Simon et al., 1991; Hepler and Gilman, these three-tier MAPK phospho-relay systems to 1992; Conklin and Bourne, 1993). To date, 17 different modulate the expression of specific primary as well as a-subunits, five b-subunits and 12 g-subunits have secondary response involved in cell proliferation, been identified. However, the identity of differentiation and apoptosis (Gutkind, 2000; Kranen- burg and Moolenaar, 2001; Werry et al., 2005). G proteins have been shown to regulate diverse transcrip- Correspondence: Dr DN Dhanasekaran, Fels Institute for Cancer Research and Molecular Biology, Temple University School of tion factors such as AP-1 (Araki et al., 1997), NF-kB, Medicine, Philadelphia, PA 19140, USA. CRE, SRE (Takeuchi and Fukunaga, 2003), ATF1 E-mail: [email protected] (Ghosh et al., 2002), STAT3 (Sellers et al., 1999), ELK1 G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3123 (Todisco et al., 1997), Egr-1 (Gerasimovskaya et al., studies focused on defining the oncogenic pathways 2002), HIF-1a (Sodhi et al., 2000), MEF2 (Fukuhara regulated by the gsp oncogene that encodes the et al., 2000a, b) via ERK-, JNK-, p38MAPKs- or ERK5 constitutively activated mutants of Gas (Landis et al., modules. However, only recently the dynamic and 1989; Lyons et al., 1990). Functional characterization of multilayered mechanisms by which G proteins regulate these activating indicated that the ectopic these MAPK networks have begun to be unraveled. This expression of Gas could stimulate ERK1/2 in different review is focused on analysing the mechanisms by which cell types (Faure et al., 1994). Using mutant S49 G proteins regulate different MAPK networks in a lymphoma cell lines that lack Gas or PKA, it was context-specific manner. further established that Gas and PKA are required to couple b2-AR signaling to ERK1/2 module (Wan and Huang, 1998). Analysis of such Gas-mediated stimula- tion of ERK1/2 indicated that it was mediated by a Regulation of MAPKs byG s pathway involving Rap-1 and its activation of B-Raf, one of the isoforms of Raf (Vossler et al., 1997; Wan G , defined by the a-subunit Ga , transduces signals s s and Huang, 1998; Schmitt and Stork, 2000; Weissman from G -coupled heptahelical receptors to adenylyl s et al., 2002; Dumaz and Marais, 2005; Houslay, 2006; cyclase that converts ATP to cAMP and subsequently Wang et al., 2006b). One of the mechanisms through to cAMP-mediated activation of which Ga activates Rap-1 involves cAMP-mediated (PKA) (Gilman, 1987; Hepler and Gilman, 1992). s direct activation of EPAC (exchange protein directly Consequently, as depicted in Figure 1, cAMP as well activated by cAMP), a guanine nucleotide exchange as cAMP-activated PKA have been shown to play a factor (GEF) specific for Rap-1 (de Rooij et al., 1998; major role in Ga -mediated stimulation as well as inhi- s Quilliam et al., 2002; Bos, 2006). In rat kidney cortical bition of MAPK modules such as those of ERK1/2, collecting duct cells, it has been shown that the ERK5 and p38MAPK (Cook and McCormick, 1993; stimulation of Ga and subsequent generation of cAMP Graves et al., 1993; Wu et al., 1993; Wan and s leads to the activation of EPAC, which stimulates Huang, 1998; Zheng et al., 2000; Laroche-Joubert GDP–GTP exchange in Rap-1. The GTP-bound Rap-1, et al., 2002; Stork and Schmitt, 2002; Dumaz and thus stimulated, activates B-Raf and the downstream Marais, 2005; Houslay, 2006; Pearson et al., 2006). ERK1/2 modules (Laroche-Joubert et al., 2002). The observation that the introduction of antibodies to Epac-1, Gas stimulation of ERK1/2 Rap-1 and B-Raf into these cells inhibited Gs-mediated The initial evidence that Gas is involved in the activation of ERK1/2 module established the linear regulation of ERK1/2 signaling module consisting pathway leading from cAMP to the activation of of a MAP3K–MAP2K–MAPK module defined by ERK1/2 module involving Epac-1, Rap-1 and B-Raf Raf, MEK1/2 and ERK1/2, respectively, came from (Laroche-Joubert et al., 2002).

Adenylyl  Cylclase P s P cAMP

Ras EPAC PKA Src C-Raf C3G MEK-1/2 ERK-1/2 MEKK2/3 MAP3K MEK5 MKK3/6 ERK5 B-Raf p38MAPK MEK-1/2 ERK-1/2

Figure 1 Gs regulation of ERK1/2. Gas stimulates the B-Raf-mediated activation of ERKs via Rap-1 or Ras and inhibits C-Raf- mediated activation of ERKs by phosphorylating C-Raf through PKA. Gas has also been shown to stimulate p38MAPK and inhibit ERK-5 modules via PKA-dependent mechanism. The dashed lines indicate the pathways that have not been fully resolved.

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3124 Although further studies confirmed the role of Rap-1 ERK pathway involves PKA-mediated phosphorylation in Gas-stimulated activation of ERK1/2 module, they of a specific isoform of Raf, known as Raf-1 or C-Raf also indicated that Gas stimulation of ERK1/2 module (Wu et al., 1993; Dhillon et al., 2002). C-Raf contains via Rap-1 involves a Rap-1 GEF other than EPAC in three PKA-target , namely Ser 43, 259 and 621 many cell types (Stork and Schmitt, 2002). The cAMP (Morrison et al., 1993), all of which can be phosphory- analog, 8-(4-chloro-phenylthio)-20-O-methyladenosine- lated in vitro by PKA (Wu et al., 1993; Hafner et al., 30,50-cyclic monophosphate (8CPT-2Me-cAMP) that 1994; Mischak et al., 1996). Mutational analyses of can activate EPAC, but not PKA, proved useful in C-Raf indicated that the Gas-mediated cAMP–PKA identifying the alternate pathway through which Gas signaling axis inhibits C-Raf through the phosphoryla- activated ERK1/2 via Rap-1 (Enserink et al., 2002). The tion of Ser-259 (Dhillon et al., 2002). The PKA observations that 8CPT-2Me-cAMP stimulated EPAC phosphorylation of Ser-259 of C-Raf attenuates the but failed to activate Rap-1-dependent activation of phosphorylation of Ser-338, which is involved in the ERK suggested that EPAC might not be involved in activation of C-Raf (Dhillon et al., 2002), thus providing Rap-1-mediated activation of ERK1/2 module in all a molecular basis for Gas-mediated inhibition of C-Raf– the cells (Enserink et al., 2002). Further studies have MEK1/3–ERK1/2 module. In addition, it has been indicated that in many cell types, Gas-mediated activa- shown that Gas-cAMP–PKA-mediated activation of tion of ERK1/2 signaling via Rap-1 involves novel Rap- Rap-1 is also involved in the inhibition of C-Raf 1-GEF known as Crk SH3 domain-binding guanine containing ERK module in fibroblast cell lines such as nucleotide-releasing factor, or C3G (Tanaka et al., 1994; NIH3T3 cells (Schmitt and Stork, 2001, 2002b). This Gotoh et al., 1995). It should be noted here that unlike has been primarily attributed to the close sequence and the direct activation of EPAC by cAMP (de Rooij et al., structural similarities of the effector domain of Rap-1 1998; Quilliam et al., 2002; Bos, 2006), the activation with that of Ras, through which Rap-1 binds and of C3G requires the PKA-mediated phosphorylation sequesters C-Raf from being activated by Ras (Bos, of Ser-17 of Src(Schmitt and Stork, 2002a; Obara 1998; Schmitt and Stork, 2001, 2002b). et al., 2004; Weissman et al., 2002; Wang et al., In summary, it can be surmised that Gas stimulates 2006b), subsequent Src-mediated activation of Cbl, B-Raf–MEK–ERK1/2 module via Rap-1 pathway in Cbl-mediated recruitment of Crk–C3G complex and cells that express B-Ras. However, Gas stimulates or Crk-SH3 domain-mediated recruitment as well as inhibits C-Raf–MEK1/2–ERK1/2 modules dependent activation of C3G (Ichiba et al., 1999; Schmitt and on the cell type in which C-Raf is expressed. Whereas Stork, 2000, 2002a). Results from these studies establish Gas-mediated stimulation of C-Raf–MEK1/2–ERK1/2 a paradigm in which Gas stimulates ERK1/2 via a module involves PKA-dependent or -independent pathway involving cAMP–PKA–Src-mediated activa- activation of Ras, Gas-mediated inhibition involves tion of C3G, C3G stimulation of Rap-1 and, finally, PKA-mediated phosphorylation or Rap-1-mediated Rap-1-mediated activation of B-Raf-MEK–ERK sequestration of C-Raf. In light of recent findings that module (Schmitt and Stork, 2000; Weissman et al., as well as PKA are functionally and/or 2002; Obara et al., 2004; Wang et al., 2006b). physically compartmentalized through their interactions Finally, it has also been noted that Gas-cAMP- with proteins such as ERKs (Gros et al., 2006) or mediated activation of Ras is involved in the stimulation AKAPs (Dumaz and Marais, 2005; Houslay, 2006), it is of ERK1/2 modules, as demonstrated in cortical quite possible that the Gas-mediated differential regula- (Ambrosini et al., 2000), thyrocytes (Tsygankova tion of ERK modules by Gas involves different pools of et al., 2000), melanocytes (Busca et al., 2000) and adenylyl cyclase and/or PKA in a single cell. cell lines (Busca et al., 2000; Amsen et al., 2006), in addition to COS-7 as well as HEK293 cells (Norum et al., 2003, 2005). In these cells, Gas stimulates Gas regulation of p38MAPK and ERK5 Ras and the subsequent activation of ERK1/2 modules In addition to its role in the regulation of ERK1/2 via PKA-dependent or -independent mechanism in a cell- modules, Gas is involved in the regulation p38MAPK as type-specific manner. Thus, Gas-mediated PKA-depen- well as ERK5 modules through the cAMP–PKA dent stimulation of Ras signaling to ERK1/2 module has signaling axis. In cardiomyocytes, it has been demon- been shown to involve a PKA-dependent Ras-GEF strated that the b2-AR stimulates the activity of p38 known as Ras-GRF1 in COS-7 and HEK293 cells MAPK involving the classical Gas-cAMP–PKA me- (Norum et al., 2005, 2007), whereas PKA-independent chanism (Zheng et al., 2000). Although the mechanism signaling involves a cAMP-responsive but PKA-indepen- through which Gas couples to the p38MAPK module is dent Ras-GEF known as CNrasGEF in melanoma cell far from clear, the findings that Rap-1 can stimulate lines (Amsen et al., 2006). p38MAPK in diverse cell types (Ahn et al., 2005, 2006) point to the possible role of Rap-1 in this pathway. However, such a role for Rap-1 in Gas-mediated Gas inhibition of ERK1/2 activation of p38MAPK remains to be established. Gas-mediated cAMP–PKA signaling pathway has also In the case of ERK5, it has been established that been shown to potently inhibit ERK pathways (Cook the cAMP–PKA pathway is involved in inhibition of and McCormick, 1993; Graves et al., 1993; Wu et al., the ERK5 module consisting of MEKK2, MEK5 and 1993). A primary mechanism by which Gas inhibits the ERK5 (Pearson et al., 2006). Interestingly, it has been

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3125

PI3K Adenylyl α i Cylclase P ββγ γ P cAMP Rap1 PLC β PKA TK ? GAPII Ras Ca++-CaM Pyk2 Rho/ CDC42 Rap1 Src Shc C-Raf

SOS ? MEK-1/2 ? ERK-1/2

JNK Ras Rac

C-Raf MEK-1/2 MAP3K ERK-1/2 MKK4/7 JNK

Figure 2 Gi regulation of MAPK network. Gi stimulates the activities of ERK, JNK and p38MAPK through a- as well as bg-subunits-dependent mechanisms as depicted. See text for details.

observed that pretreatment of cells with cAMP inhibited that Gai2 can activate the ERK module through a (RTK)-mediated activation diverse set of mechanisms involving either direct or of ERK5 in HeLa, C2C12 and NIH3T3 cells. Analysis indirect activation of Ras (Pace et al., 1995; Edamatsu of the underlying mechanism has indicated that et al., 1998). In addition to its activation of ERK PKA phosphorylates MEKK2 at Ser 153, the phos- modules through these mechanisms, Gai also appears to phorylation of which inhibits its coupling to transmit signals to ERK as well as other MAPK upstream activators, presumably the candidate modules via its erstwhile associated bg-subunit (Figure 2; MAP4Ks. Thus, Gas-cAMP–PKA-mediated phosphor- Crespo et al., 1994; Faure et al., 1994; Koch et al., 1994; ylation of MEKK2 potentially leads to the overall Pace et al., 1995). inhibition of signaling by MEKK2–MEK5–ERK5 module (Pearson et al., 2006). Gai stimulation of ERK1/2 Gai-dependent activation of ERK primarily involves the suppression of two inhibitory pathways. One mechan- Regulation of MAPKs byG i ism involves its inhibitory effect on adenylyl cyclase, whereas the other involves its inhibitory effect on Rap-1 The Gi family of G proteins is defined by the a-subunits via Rap-1-GAP protein. The well-characterized signal Gai1,Gai2,Gai3,Gaz,GaoA and GaoB. The identifica- transduction role of Gai is to couple specific receptors to tion of the activated mutants of Gai2 as the gip2 the inhibition of adenylyl cyclase (Johnson and Dhana- oncogene in pituitary adenomas as well as tumors of the sekaran, 1989; Tang and Gilman, 1992). Consistent with ovary and the adrenal cortex (Lyons et al., 1990) this prototypicsignaling role of G ai, expression of gip2, indicated the mitogenic activity of Gai. Studies focused the activated mutant of Gai, resulted in a decrease in the on defining the molecular basis for such mitogenic accumulation of cAMP in cultured cells (Lowndes et al., activity identified the role of Gai in the regulation of the 1991; Wong et al., 1991). Such a Gai-mediated decrease ERK module (L’Allemain et al., 1991; Pouyssegur and in cAMP levels and subsequent decrease in PKA activity Seuwen, 1992b). Expression of the GTPase deficient, relieves the inhibitory effect of PKA on C-Raf, thus constitutively activated mutant of Gai2 resulted in the potentiating Ras–C-Raf signaling to ERK module oncogenic transformation of Rat1 fibroblasts (Pace (Radhika and Dhanasekaran, 2001). The observation et al., 1991; Gupta et al., 1992b). Functional characteri- that the expression of Gai2 slightly enhances the zation of the transformed indicated that activation of C-Raf (Pace et al., 1995) is consistent with expression of the activated Gai2 was associated this view. with constitutive activation of p44-ERK activity However, Gai has also been shown to activate (Gupta et al., 1992a). Subsequent studies have shown the ERK pathway via an alternate Ras-dependent

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3126 mechanism (Pace et al., 1995; Mochizuki et al., 1999). the bg-subunit is involved in activation of ERK by a Using a strategy wherein the expression of a pertussis number of Gi-coupled receptors including m2-muscarci- toxin-resistant mutant of Gai2 in cells treated with PTX nic, a2-adrenergic, D2 dopamine, A1 adenosine and could be used to monitor the effect of A1-adenosine LPA receptors (Crespo et al., 1994; Koch et al., 1994). receptors specifically coupled to the transfected PTX- Consistent with these observations, expression of bg- resistant Gai2; it was demonstrated that the expression subunit has been shown to increase the basal activity of of Gai2 and subsequent activation by A1 adenosine ERK in COS-7 cell (Faure et al., 1994). Further analyses receptor led to the stimulation of ERK activity (Pace using a dominant negative mutant of Ras have indicated et al., 1995). In addition, the coexpression of dominant that the bg-mediated activation of ERK1/2 module negative mutant of Ras inhibited Gai2-mediated activa- involves Ras (Crespo et al., 1994; Faure et al., 1994; tion of MEK and the downstream ERK, indicating a Koch et al., 1994). Although bg-subunit-mediated role for Ras in this pathway (Pace et al., 1995). activation of ERK module has been shown to involve However, an intriguing observation was that there was Ras (Crespo et al., 1994; Koch et al., 1994), the no increase in the GTP-bound active form of Ras upon mechanism through which the bg-subunit couples to the stimulation of Gai2 by A1-adenosine receptors. Ras appears to be cell-type specific. However, the Thus, although these studies indicated that the activa- consensus is that the activation of Ras by bg-subunit tion of ERK by Gai2 required functional Ras, the involves a tyrosine kinase, the identity of which is molecular basis for such requirement with little or no context specific. The immediate step involved in bg- modulation of the activation Ras was not clear. mediated activation of ERK involves - However, the later observation that Gai2 can interact C-b (PLC) and/or phosphoinositide-3-kinase (PI3K), with Rap-1-GAP provided a molecular basis for a both of which can be activated by the bg-subunit mechanism in which Gai2 can activate ERK through a (Camps et al., 1992; Katz et al., 1992; Stephens et al., Ras-sensitive pathway without directly activating Ras 1994; Thomason et al., 1994). Transfection studies using (Mochizuki et al., 1999). Rap-1 was initially identified as COS-7 or HEK293 cells have established a model in a Ras-like GTPase that can act as a tumor suppressor which Gi-coupled a2A-adrenergicreceptors stimulate by attenuating Ras-mediated cellular transformation ERK1/2 modules via the released bg-subunits (Lev (Pizon et al., 1988; Kitayama et al., 1989). Rap-1-mediated et al., 1995; Dikic et al., 1996; Della Rocca et al., 1997). suppression of transformation is associated with the In this model, bg-subunit released from Gi-heterotrimer ability of activated Rap-1 to attenuate Ras-dependent stimulates PLCb leading to phosphatidylinositol ERK activation by EGF as well as LPA (Cook et al., trisphosphate (IP3)-mediated increase in intracellular 1993). As described earlier, activated Rap-1 through Ca2 þ and Ca2 þ –calmodulin-mediated activation of Pyk2 its Ras-homologous effector domain sequesters C-Raf kinase. The Pyk2 kinase thus stimulated, activates Src, (Bos, 1998; Schmitt and Stork, 2001, 2002b), thus which in turn stimulates Shc adaptor protein leading to disrupting Ras-mediated signaling to the C-Raf– the activation of Ras via the Ras-GEF, mSOS MEK–ERK-signaling module. Gai2 attenuates the (Lev et al., 1995; Dikic et al., 1996; Luttrell et al., Ras-antagonistic inhibitory activity of Rap-1 by recruit- 1996; Della Rocca et al., 1997). ing a specific Rap-1 GTPase activating protein II In some other cellular contexts, it has been observed, (Rap1GAPII), which can downregulate Rap-1 activity bg-subunit transmits signals through an Src- and Shc- (Mochizuki et al., 1999). Thus, mutational or receptor- independent mechanism involving the phosphorylation mediated activation of Gai2 facilitates the translocation of II and its association with Grb2 (Kranen- of Rap1GAPII to the plasma membrane where Rap1- burg et al., 1997, 1999a, b). In Rat1a and COS-7 cells, it GAPII accelerates the GTPase activity of Rap-1, there- has been demonstrated that LPA- or thrombin receptor by rendering it inactive. Subsequent release of stimulated release of bg-subunit activates ERK1/2 sequestered C-Raf leads to Ras–C-Raf reassociation module via a pathway involving PI3K and PI3K- and the activation of the downstream ERK1/2 module mediated activation of a tyrosine kinase that promotes (Mochizuki et al., 1999). It is significant to note here dynamin II–Grb2 complex formation with the resultant that the pathway deduced from this signaling paradigm activation of Ras and the ERK1/2 module (Kranenburg supports a view that Gai2 stimulates Ras-dependent et al., 1997, 1999a, b). Since dynamin can link Ras-GEF ERK signaling without directly involving the activation SOS to Ras (Wunderlich et al., 1999), it is possible that of Ras by inhibiting the activity of Rap-1. the dynamin facilitates bg-mediated activation of Ras in this complex. As dynamin has been shown to play a critical role in vesicular endocytosis (Hinshaw and Gbgi stimulation of ERK1/2 Schmid, 1995; Praefcke and McMahon, 2004), and Gi has also been shown to stimulate the ERK1/2 vesicular endocytosis has been shown to be required for signaling module by mechanisms that involve the H-Ras-mediated activation of ERK1/2 module (Roy disassociated bg-subunits (Crespo et al., 1994; Koch et al., 2002), it is likely that dynamin II-mediated et al., 1994). Expressing the bg-subunit interacting endocytosis plays a critical role in bg-mediated activa- COOH domain of the b-adrenergicreceptor kinase tion of ERK1/2. As seen in the case of b-arrestin- (bARK-CT) or Gat, both of which can function as mediated activation of ERK1/2 in endocytic vesicles antagonists of bg signaling by virtue of their ability to (Lefkowitz and Shenoy, 2005), it is possible that sequester endogenous bg, it has been demonstrated that bg-mediated ERK1/2 signaling is not involved in the

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3127 transmission of signals to the nucleus. However, such a et al., 1999, 2000), presumably involving tyrosine kinase differential role for bg-dynamin-mediated activation of responsive-specific GEFs (Shou et al., 1995; van Biesen ERK1/2 modules remains to be clarified. et al., 1995; Miyamoto et al., 2003). Similar to the activation of the ERK module by Gbgi-subunit, the activation of JNK by Gbgi appears to require a PI3K Gi stimulation of JNK pathway activity in certain cellular contexts, such as the activa- In addition to the activation of ERKs, Gai as well as the tion of Gi-coupled m-opioid receptor (Kam et al., 2004). bg-associated with Gai has been shown to activate the JNK module (Coso et al., 1995, 1996; Edamatsu et al., 1998; Yamauchi et al., 2000). Stimulation of ectopically Regulation of MAPKs byG q expressed m2-muscharinic receptors that couple to Gi- family of G proteins have been shown to activate JNK The G family of G proteins is defined by the via bg-subunits in COS-7 cells (Coso et al., 1995, 1996). q Similar analysis using mastoparan, which mimics GPCR ubiquitously expressed Gaq,Ga11 and hematopoetic cell-specific Ga , and Ga subunits (Simon et al., activation by directly stimulating G proteins (Higashi- 14 15/16 1991; Hubbard and Hepler, 2006). All of the family jima et al., 1990), indicated that JNK could be activated members have been shown to activate ERK1/2, JNK by both the a- and the bg-subunits of G in HEK293 cells i and p38MAPK modules. The a-subunits of G family (Yamauchi et al., 2000). The use of bg-quenching q of G proteins transduce signals from their cognate bARK-CT established that the mastoparan activation receptors to specific cellular responses via the activation of JNK involved both Ga and Gbg in these cells i i of the effector PLCb (Johnson and Dhanasekaran, (Yamauchi et al., 2000). Consistent with this observa- 1989; Simon et al., 1991). Activated PLCb hydrolyses tion, transient expression of the constitutively activated phosphatidylinositol 4,5-bisphosphate (PIP2) to pro- mutants of Gai1,Gai2 and Gai3 in HEK293 cells increased JNK activity. Interestingly, similar constitu- duce inositol triphosphate (IP3) and diacylglycerol (DAG), both of which can activate tively activated mutants of Ga and Ga , which belong o z (PKC), either directly or indirectly via the release of to Gi family, failed to stimulate such JNK activation 2 þ (Yamauchi et al., 2000). Thus, the activation of JNK internally stored Ca . In addition to these a-subunit- mediated pathways, the bg-subunit released from G can appears to be restricted to some G isoforms but not to q i also activate PLCb (Camps et al., 1992: Katz et al., other G family members such as Ga or Ga . The i o z 1992) and transmit signals through both PLC–DAG– expression of dominant negative Rho and Cdc42, but PKC- and PLC–IP3-Ca2 þ -mediated signaling pathways not dominant negative Rac, inhibited Ga -mediated i (Figure 3). activation of JNK, suggesting a pathway involving Rho and CDC42 in the activation of JNK by Gai (Yamauchi et al., 2000). Interestingly, Gai-mediated activation of Gq stimulation of ERK1/2 JNK was not inhibited by the dominant negative Gaq can stimulate ERK1/2 module via PLC–DAG– mutants of MKK4 and MKK7, the upstream MAP2Ks PKC as well as PLC–IP3-Ca2 þ signaling mechanisms. that are typically involved in the activation of JNKs Gaq-activated PKC can stimulate ERK1/2 module by (Yamauchi et al., 2000). Thus, although, Rho and directly phosphorylating and stimulating C-Raf (Kolch Cdc42 have been shown to stimulate JNK via MKK4 in et al., 1993; Ueda et al., 1996; Schonwasser et al., 1998). 2 þ HEK293 T cells (Teramoto et al., 1996), Gai-mediated An alternate mechanism involving Ca –calmodulin- activation of JNK proceeds through a novel pathway mediated activation of Pyk2 leading to the activation of involving Rho and/or CDCD42 and a hitherto unchar- Ras and subsequently ERK1/2 module can also be acterized tyrosine kinase via an MKK4/7-independent envisaged (Lev et al., 1995; Dikic et al., 1996; Della mechanism (Yamauchi et al., 2000). In contrast, the Rocca et al., 1997). However, whether Gaq/11 activates 2 þ pathways involving bg-subunit dissociated from Gai ERK1/2 via the PKC–Raf signaling axis or IP3-Ca – have been demonstrated to involve Ras and Rac(Coso calmodulin–Pyk2–Src–Ras pathway appears to be cell et al., 1996). In addition, bg-mediated stimulation of type dependent. It has been shown that m1-muscarinic JNK involves both MKK4 and MKK7, as bg-activated receptor-mediated activation of Gaq stimulates the JNK can be inhibited by the dominant negative mutants ERK1/2 module via PKC–C-Raf signaling axis in Cos-7 of MKK4 as well as MKK7 (Yamauchi et al., 1999). and CHO cells (Hawes et al., 1995). In contrast, Although bg-activation of MKK4 proceeds through Gq-coupled LPA- or bradykinin receptors activate Rho and CDC42, its activation of MKK7 involves Rac ERK1/2 via calcium–calmodulin-dependent pathway (Yamauchi et al., 1999). Although the physiological involving Pyk2, Srcand Ras (Dikic et al., 1996). In significance is not known, it has been shown that HEK293 cells, a1-adrenergic receptor-mediated activa- bg-preferentially activates MKK4 in HEK293 cells. tion of ERK1/2 appears to involve PKC- as well as The signaling network that is being unraveled by these Ca2 þ -mediated pathways (Della Rocca et al., 1997). studies indicates that both Gai and Gbgi activate JNK In addition to these typical Gaq-mediated pathways, it module via distinct subunit-specific small GTPases has also been shown that Gaq can activate ERK through 2 þ (Figure 2). However, in both Gai-andGbgi-mediated a novel mechanism involving a DAG- as well as Ca - activation of JNK, a tyrosine kinase is involved in the dependent Rap-1-GEF (Guo et al., 2001). Using a activation of the respective small GTPases (Yamauchi variant of PC12 cells, PC12D, it has been shown that

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3128

PI3K α q βγβγ

PLCβ

PLCβ DAG PKC Ca++ Ca++-CAM

TK ? CaDAG- Pyk2 GEFI C-Raf

MEK-1/2 Src ERK-1/2 MEKK1 Shc p130CAS Rap1 MKK4/7 CRKII

SO JNK S MEKK2/3 DOCK B-Raf MEK5 180 C3G ERK5 MEK-1/2 Ras ERK-1/2 R Rac -Ras C-Raf

MEK-1/2 MAP3K ERK-1/2 MAP3K MKK3/6 MKK 4/7 p38MAPK JNK

Figure 3 Gq regulation of MAPK network. Gaq stimulates ERK, JNK, p38MAPK and ERK5 modules via PKC- as well as Ca2 þ -dependent pathways as illustrated (see text for details). In addition, the bg-subunits can initiate similar pathways through their ability to activate PLCb and/or PI3K. The dashed lines indicate the pathways that need to be further characterized.

m1-muscarinic receptor stimulation of ERK1/2 modules Gq stimulation of JNK is independent of Ras but dependent on Gaq-stimulated In contrast to the major role of the a-subunits of the Gq PLCb. More interestingly, it has been shown that family in activating the ERK module, the a- as well as 2 þ Gq-mediated release of DAG and Ca leads to the the bg-subunits of Gq plays determinant roles in the activation of a calcium- and diacylglycerol-regulated activation of JNK module (Coso et al., 1995, 1996; guanine nucleotide exchange factor, CalDAG-GEFI Mitchell et al., 1995; Zohn et al., 1995; Nagao et al., that can stimulate Rap-1, which in turn, can activate 1998). Receptors that couple to Gq such as the m1- B-Raf and the downstream ERK1/2 module (Guo et al., muscarinic (Coso et al., 1995), m3-muscarinic (Wylie 2001). Thus, Gaq-mediated signaling to ERK1/2 module et al., 1999), angiotensin II (Zohn et al., 1995), a1 appears to involve three distinct mechanisms: (1) adrenergic(Ramirez et al., 1997) thrombin (Shapiro PKC-mediated activation of C-Raf; (2) Ca2 þ -Pyk2- et al., 1996) and endothelin-1 (Shapiro et al., 1996) Src-dependent activation of Ras; and (3) Ca2 þ – receptors have been shown to potently stimulate the DAG-stimulated Rap1-mediated activation of B-Raf. JNK module. The findings that Gaq activates MEKK1 As Gaq/11 activation results in the generation of both and MEKK1 deficiency disrupts Gaq-mediated activa- DAG and IP3, all of these mechanisms need not be tion of JNK suggest that MEKK1 is a primary signaling mutually exclusive. It should be noted here that since bg- hub for the activation of JNK by Gaq (Minamino et al., subunits disassociated from Gq/11-coupled receptors can 2002). However, the molecular mechanism(s) through also stimulate PLCb, it is possible that they also which Gq communicates to the JNK module appears to contribute to Gq/11 signaling to ERK1/2 modules in a be cell-type specific. Using ectopic expression of Gq- similar manner. However, it has been noted that coupled m1-muscarinic receptors in NIH3T3 (Coso Gq/11-coupled receptors primarily use the DAG/ et al., 1995), Rat1a (Mitchell et al., 1995), COS-7 (Coso PKC- and/or IP3/Ca2 þ signaling pathways, rather than et al., 1996) or HEK293 cell (Nagao et al., 1998), it has bg-subunits, to activate ERK1/2 modules (Blaukat been shown that the stimulation of these receptors lead et al., 2000). A footnote here is that in some cellular to the potent activation of JNK module. While, Gq- contexts, as in the case of the activation of ERK1/2 coupled m1-muscarinic receptor activation of JNK does by Gq-coupled oxytocin receptors, the bg-disassociated not involve the PLC-PKC axis in NIH3T3 cells (Coso from Gq activates ERK1/2 through the transactivation et al., 1995), studies with COS-7 cells indicated that the of receptor tyrosine kinase (Zhong et al., 2003). activation of JNK by m1-muscarinic receptor involves a

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3129 pathway initiated by the Gbgq-involving Ras and Rac-1 Gq stimulation of p38MAPK (Coso et al., 1996). In contrast, studies using Rat1a cells Similar to its role in the activation of the JNK module, have shown that m1-muscarinic receptor activation of different Gq-coupled receptors have been shown to 2 þ JNK by Gaq involves a Ca -dependent mechanism activate the p38MAPK module (Kramer et al., 1995; (Mitchell et al., 1995), thereby suggesting a role for the Krump et al., 1997; Yamauchi et al., 1997). Further Gaq-PLC-IP3 signaling axis. Likewise, it has been studies have established that Gq can activate p38MAPK shown that Gaq-coupled angiotensin II receptor activa- module through a- as well as the bg-subunits (Yamauchi tion of JNK proceeds through such Ca2 þ -dependent et al., 1997, 2001; Nagao et al., 1998; Huang et al., signaling, possibly involving a PLC-IP3-based mechan- 2004). The basic mechanism through which Gaq ism (Zohn et al., 1995). Interestingly, such Ca2 þ - activates p38MAPK is not different from the one dependent signaling has also been shown to be sensitive involved in the activation of the JNK module. The 2 þ to genistein, suggesting a role for Ca -dependent PLC–DAG–PKC signaling arm of Gaq along with the tyrosine kinase phosphorylation (ibid). In addition, a activation of Src or an Src-like tyrosine kinase has been direct role for PKC in the activation of JNK has also shown to be involved in Gaq-mediated activation of been established. In HEK293 cells, it has been shown p38MAPK (Nagao et al., 1998; Yamauchi et al., 2001). that the expression of the constitutively activated Thus, the signaling pathways mediated by Gaq involve mutant of Ga11 results in the activation of JNK through PLC-PKC-dependent activation of Src followed by Src a pathway involving PKC as well as Srcfamily of activation of the Rho family of GTPases leading to the tyrosine kinases (Nagao et al., 1998). activation of p38MAPK module. Consistent with this The general mechanism emerging from these studies is view, it has been shown that Gaq activates MKK3 or that Gq can use both the a- as well as bg-subunits MKK6, the MAP2Ks of p38MAPK module, via involving the PLC–DAG–PKC as well as the PLC-IP3- CDC42, Racand Rho (Yamauchi et al., 2001). The Ca2 þ signaling arms to activate JNK. Src- or an Src-like observations that the Rac-specific GEFs Tiam-1 and kinase appears to be involved in the activation of JNK Ras-GRF, both of which can be activated by Src, are in in both the pathways (Zohn et al., 1995; Nagao et al., physical complex with MKK3 and p38MAPK (Buchs- 1998). Therefore, PKC activation of Src and subsequent baum et al., 2002) provides partial evidence for such a Src-mediated activation of Rac through a specific GEF pathway that can be activated by Gaq. appears to be involved in Gaq-mediated activation of In contrast to the role of PLC-PKC in Gaq-mediated JNK via the pathway involving PKC, Srcand Rac pathways, the bg-pathways derived from the Gq hetero- (Servitja et al., 2003). Although such a linear pathway trimer involves a PLC-independent mechanism(s) invol- has not been established in a specific single cellular ving Rho family of GTPases and a tyrosine kinase that context, the previous findings that PKC can readily remains to be identified (Yamauchi et al., 2001). In activate Src through direct phosphorylation (Gould addition, it has been shown that Gq-derived bg-subunit et al., 1985; Moyers et al., 1993), Src can activate Rac- can activate p38MAPK via a pathways involving Rap-1 specific GEFs (Servitja et al., 2003) and such activation (Huang et al., 2004). It is quite likely that the activation of Raccanstimulate the JNK module (Crespo et al., of Rap-1 by bg-subunits involves a Pyk2-Src-C3G 1996; Michiels et al., 1997) strongly points to such a pathway similar to the one involved in the activation pathway. of JNK (Mochizuki et al., 2000). Although the mechan- Activation of JNK module by the PLC-IP3-Ca2 þ arm ism by which Rap-1 activates p38MAPK module 2 þ of Gaq signaling has been shown to involve a Ca - is not fully elucidated, the findings that bg-pathway dependent activation of Pyk2 (Tokiwa et al., 1996; can stimulate C3G (Mochizuki et al., 2000), and C3G Blaukat et al., 1999), Pyk2-induced activation of Src, can stimulate Rap-1 (Gotoh et al., 1995) as well as Src-mediated phosphorylation of p130CAS and the MLK3/DLK family of kinases (Tanaka and Hanafusa, subsequent association of p130CAS and the adaptor 1998) and MLK3 can activate p38MAPK (Chadee and protein CrkII (Blaukat et al., 1999; Kodama et al., Kyriakis, 2004; Kim et al., 2004) points to MLK family 2003). Interestingly, the p130CAS–CRKII complex has of MAP3Ks as a possible link between Rap-1 and been shown to stimulate the JNK module via Rac-1 p38MAPK modules. (Dolfi et al., 1998; Girardin and Yaniv, 2001) presum- ably through the activation of Rac-1 by the Rac-GEF, DOCK180 (Kiyokawa et al., 1998) or R-Ras by the Gaq stimulation of p38MAPKg/ERK6 module R-Ras/Rap-1-GEF, C3G (Mochizuki et al., 2000). p38MAPKg, an isoform of p38MAPK that shows 2 þ Therefore, the signaling from Gaq via PLC-IP3-Ca distinct expression and activation profiles, is character- is likely to involve Rac-1- or R-Ras-dependent mechanisms, ized as ERK6 (Lechner et al., 1996; Kumar et al., 1997; both of which can lead to the activation of JNK module Wang et al., 2000). Similar to the p38MAPK module, by stimulating JNK-specific MAP4Ks such as MLK3 p38MAPKg/ERK6 is activated by the MAP2Ks, (Teramoto et al., 1996; Tanaka and Hanafusa, 1998). MKK3 and MKK6 (Kumar et al., 1997; Wang et al., Since bg-subunits can stimulate PLC-b and subse- 2000). It has also been shown that the small GTPase quent IP3-mediated Ca2 þ release/influx, it should be Rho activates p38MAPKg/ERK6 via its stimulatory noted here that the bg-signaling, leading to the activa- interaction with PKN, which can activate the MLK-like tion of JNK via Ras and Rho family of GTPases, kinases that act as MAP3Ks of this module (Wang et al., involves a pathway similar to the one described for Gaq. 2000; Gross et al., 2002; Takahashi et al., 2003). Initial

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3130 evidence that Gq-mediated pathways are involved in the neoplastictransformation of fibroblast celllines (Chan activation of p38MAPKg/ERK6 came from the obser- et al., 1993; Jiang et al., 1993; Xu et al., 1993; Vara vation that m1-receptor-expressing NIH3T3 cells Prasad et al., 1994; Voyno-Yasenetskaya et al., 1994; demonstrated the activation of ERK6/p38MAPKg Dhanasekaran and Dermott, 1996; Liu et al., 1997). In in response to carbachol (Marinissen et al., 1999). In view of the findings that constitutively active Ga12 was analysing the role of PAR-1 receptors in stimulating isolated as the transforming ‘oncogene’ from the Ewings c-Jun expression and oncogenic transformation, the sarcoma cell line (Chan et al., 1993) and that Ga12 ability of Gaq to activate p38MAPKg/ERK6 was potently stimulates the neoplastictransformation of characterized. It appears that the activation of ERK6 fibroblast cell lines (Chan et al., 1993; Jiang et al., 1993; is mediated primarily by the a-subunit rather than the Xu et al., 1993; Vara Prasad et al., 1994; Voyno- bg-subunit (Marinissen et al., 2003). Since Gaq can Yasenetskaya et al., 1994), it has been designated as stimulate Rho-GEF proteins such as leukemia- ‘gep’ oncogene (Xu et al., 1994; Gutkind et al., 1998). In associated Rho-GEF (LARG) (Booden et al., 2002), it addition, the expression of these a-subunits has been is very likely that Gaq-mediated activation of the shown to stimulate mitogenicresponses in many p38MAPg/ERK6 module involves Rho-dependent different cell lines (Aragay et al., 1995; Denis-Henriot activation of PKN and subsequent stimulation of et al., 1998; Coffield et al., 2004). Further analyses have MLK-family of MAP3Ks followed by MKK3/6 and revealed that these a-subunits can regulate cell prolife- p38MAPKg/ERK6 (Wang et al., 2000; Marinissen et al., ration (Aragay et al., 1995; Denis-Henriot et al., 1998; 2001, 2003; Gross et al., 2002; Takahashi et al., 2003). In Radhika and Dhanasekaran, 2001; Coffield et al., 2004; this context, it should be noted that since p38MAPKg/ Kumar et al., 2006), differentiation (Jho and Malbon, ERK6 is activated by MKK3 and MKK6, which are 1997), apoptosis (Althoefer et al., 1997; Berestetskaya also involved in the activation of other p38MAPK- et al., 1998), as well as (Offermanns et al., isoforms, it could also be regulated by the mechanisms 1997; Radhika et al., 2004; Dhanasekaran, 2006; Shan outlined in the previous section. et al., 2006) in a context-specific manner. A search for the underlying signaling mechanism(s) identified that G stimulation of ERK5 G12 proteins strongly stimulate JNK activity in a variety q of cells (Prasad et al., 1995; Collins et al., 1996; Voyno- G -mediated signaling has also been shown to regulate q Yasenetskaya et al., 1996; Jho et al., 1997; Nagao et al., the ERK-5 module via MEK5 (Marinissen et al., 1999). 1999; Arai et al., 2003; Marinissen et al., 2003), and the Studies using the transient expression of G -coupled q inhibition of which could abrogate Ga -mediated G1 muscarinic receptors have identified that the G - 12/13 q to progression (Mitsui et al., 1997). mediated signaling pathway is involved in the regulation of the MEK5–ERK-5 signaling module and subsequent transactivation of c-Jun via MEF2 family of transcrip- G signaling to JNK module tion factors (Marinissen et al., 1999). Further studies 12/13 Having 67% shared amino acid identity, Ga and Ga using a chimeric mutant of Ga and Ga , through which 12 13 q i stimulate similar responses including the activation of Ga -mediated signaling can be activated by Ga -coupled q i JNK in many cellular contexts (Radhika and Dhanase- muscarinic receptors, have established that Gaq and not Gbg is involved in the activation of ERK5 (Fukuhara karan, 2001). However, the mechanism through which q these a-subunits couple to the JNK module appears to et al., 2000b). Although the mechanism by which Ga is q be cell-type or physiological context dependent. In these coupled to the MEK5–ERK5 module is not fully different contexts, Ga and Ga activate the JNK understood, initial studies have identified that it is not 12 13 module via the small GTPases Ras, CDC42, Racor Rho through Ras, CDC42, Racor Rho (Fukuhara et al., (Figure 4). Although the activated mutant of Ga 2000b). However, the observation that the MAP3Ks 12 stimulates JNK via Ras in NIH3T3 cells, it involves MEKK2 and MEKK3 can interact and activate MEK5 Rac1 in 1321N astrocytoma and HEK293 cells (Collins (Sun et al., 2001) suggests the possible role of these et al., 1996). In Cos-7 cells, it has been shown to involve MAP3Ks in this pathway. In conjunction with the CDC42 (Voyno-Yasenetskaya et al., 1996). All of these findings that Rap-1 is involved in the activation of ERK5 via MEKK2 (Wang et al., 2006a) and G can small GTPases appear to transmit signals to the JNK q module via MEKK1, as the expression of dominant activate Rap-1 via the Rap-1-GEF CG3 or CalDAG- negative MEKK1 has been shown to inhibit Ga - GEFI as discussed in the previous sections, it is likely 12 mediated activation of JNK in all of these instances that Ga , via Rap-1, activates the MEKK2–MEK5– q (Collins et al., 1996; Voyno-Yasenetskaya et al., 1996). ERK5 module. However, these earlier events involved Thus, Ga as well as Ga communicates to MEKK1, in Ga -mediated activation of ERK5 remain to be 12 13 q the upstream MAP3K of the module, via the Ras- or established. Rho family of small GTPases, presumably through specific GEFs. The identity of the specific GEFs that couple Ga12 or Ga13 to JNK module has been identified Regulation of MAPKs byG 12 only in a few instances (Lee et al., 2004). In the case of the Ras-GEF linking Ga12 to Ras, it The G12 family of G proteins, defined by the a-subunits appears to involve Shc-mediated recruitment of SOS Ga12 and Ga13, is extremely potent in inducing (Collins et al., 1997). It has been shown that Ga12

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3131

Ras α 12/13 Rac

CDC42 α α 12 13 α Rho 12

Raf MAP3K MEKK2/3 MAP3K MAP3K Raf MEK-1/2 MKK4/6 MEK5 MKK3/6 MKK3/6 MEK-1/2 ERK-1/2 JNK ERK5 p38MAPK p38MAPK ERK-1/2

Figure 4 G12/13 regulation of MAPK networks. Ga12 as well as Ga13 can potently activate JNK module by stimulating Ras as well as Rho family of GTPases. In some cells, Ga12/13 can weakly stimulate ERK1/2 in a Ras-dependent manner, whereas in others they attenuate ERK1/2 activation at the levels of MEKs. In addition, both Ga12 and Ga13 can activate ERK5 through hitherto uncharacterized mechanism. In addition, Ga12 and Ga13 can regulate different MAPKs through pathways unique to them (dashed lines). While Ga12 inhibits p38MAPKs in some cell types at the levels of MKKs, Ga13 stimulates p38MAPKs including p38MAPKg/ ERK6 via MKK3/6 in some cells.

stimulates Shc-phosphorylation in 1321N1 cells, in Rho-GEF involved in linking Ga12 to Rho in an Src- which it stimulates JNK via Ras-dependent mechanism dependent manner and subsequent activation of JNK (Collins et al., 1996, 1997). Therefore, it has been module. However, it is also possible that Ga12 stimulates suggested that Shc-Grb2-recruited SOS is involved in Rho-mediated activation of JNK module via other Src- stimulating Ras and subsequently JNK in these cells responsive Rho-GEFs. In the case of Ga13, it has been (Collins et al., 1997). Although the kinase involved in shown that it can physically interact with p115RhoGEF stimulating the phosphorylation of Shchas not been and stimulate its GEF activity toward RhoA (Hart identified, considering the findings that Ga12 can et al., 1998). Consistent with this observation, Rho- stimulate several tyrosine kinases such as Src (Nagao dependent activation of JNK has been shown to involve et al., 1999), Fak (Needham and Rozengurt, 1998; p115RhoGEF in P19 cells (Lee et al., 2004). Notwith- Chikumi et al., 2002) and Tecfamily of kinases (Mao standing the small GTPase utilized, the MAP3K et al., 1998; Jiang et al., 1998), it possible that one of involved in Ga12/13-mediated signaling appears to these kinases may be involved in stimulating the involve MEKK1 (Collins et al., 1996; Voyno-Yasenets- phosphorylation of Shc. In this context, the observa- kaya et al., 1996; Wang et al., 2002). In some contexts, tions that Ga12 can stimulate BTK, a member of Tec the JNK module activated by Ga12/13 involves ASK1 family of kinases (Jiang et al., 1998) and BTK can (Berestetskaya et al., 1998), MEKK-2 or MEKK4 as activate JNK via Shc-Grb2-mediated activation of Ras well (Wang et al., 2002; Lee et al., 2004). Generally, the (Deng et al., 1998), make BTK or one of its family MAP3Ks activate MKK4 as well as MKK7, the JNK- members an attractive candidate in this pathway. The specific MAP2Ks. However, it has been observed that identities of the GEFs that link Ga12/13 to downstream Ga12-mediated activation of JNK preferentially involves Racand CD42 are largely unknown. MKK7 (Dermott et al., 2004), whereas Ga13- as well as In addition to Ras, Racand CDC42, it has also been bg-mediated activation appears to involve MKK4 shown that Ga12 stimulates JNK via RhoA (Nagao (Yamauchi et al., 1999; Wang et al., 2002; Lee et al., et al., 1999; Arai et al., 2003). In HEK293 cells, it has 2004). Although, the functional significance of these also been shown that Src-dependent stimulation of differential preferences for specific MKKs is largely not RhoA is involved in Ga12-mediated activation of JNK known, it should be noted here that the dual specificity (Nagao et al., 1999). The findings that inhibition of Src kinases MKK4 and MKK7 activate JNK by phosphory- by treating the cells with PP2 or expressing the lating two different amino acid residues. MKK4 has dominant negative mutant of Srcabrogated the ability been shown to preferentially phosphorylate Tyr185, of Ga12 to stimulate such activation suggested that an whereas MKK7 phosphorylates Thr187 of JNK1 Src-mediated GEF activity is involved in linking Ga12 to (Fleming et al., 2000). It has been proposed that both Rho-mediated activation of JNK module (Nagao et al., MKK4 and MKK7 are required for the maximal 1999). Although the mechanism by which Ga12 signals activation of JNK1 (Yang et al., 1997; Lawler et al., to Srcis not fully understood, the ability of G a12 to 1998; Lisnock et al., 2000). Since Ga12 appears to stimulate the GEF activity of tyrosine-phosphorylated preferentially stimulate MKK7 (Dermott et al., 2004), LARG is quite significant (Fukuhara et al., 2000a; whereas Ga13 stimulates MKK4 (Wang et al., 2002; Lee Suzuki et al., 2003). LARG may thus be a candidate et al., 2004), it is likely that the Ga12- and Ga13-mediated

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3132 response requires only a partial activation of JNK from directly stimulate PP5 (Yamaguchi et al., the respective MAP2K signaling nodes. It is likely that 2002) as well as PP2A (Zhu et al., 2004), it is likely that such partial activation of JNK along with other either or both of these play a role in signaling inputs, such as the activation of ERK module attenuating p38MAPK module. However, such a role and attenuation of p38MAPK module, are prerequisites for these proteins in Ga12-mediated attenuation of for Ga12-mediated cell proliferation of NIH3T3 cells or p38MAPK module remains to be clarified. Likewise, Ga13-mediated P19 cell differentiation in which the whether the downregulation of p38MAPK by Ga12 is differential recruitment of MAP2Ks have been observed seen in other cell types remain to be established. (Dermott et al., 2004; Wang et al., 2002; Lee et al., However, such differential regulation of JNK and 2004). p38MAPK modules is likely to have a significant role in the potent mitogenicactivity of G a12 seen in diverse cell types (Chan et al., 1993; Jiang et al., 1993; Xu et al., Ga attenuation of ERK1/2 12/13 1993; Vara Prasad et al., 1994; Voyno-Yasenetskaya In contrast to their potent activation of the JNK et al., 1994; Aragay et al., 1995; Dhanasekaran and module, Ga and Ga attenuate the activation of 12 13 Dermott, 1996; Liu et al., 1997; Denis-Henriot et al., ERK1/2 module in a cell-type-dependent manner 1998; Coffield et al., 2004). It is likely that the weak (Voyno-Yasenetskaya et al., 1996). Ras-mediated albeit stimulation of ERK activity and the decisive down- weaker stimulation of ERK1/2 has been shown to be regulation of p38MAPK in addition to the potent critically required – along with Ras-/Rac-mediated stimulation of JNK module provide the multiple potent stimulation of JNK activity – for Ga -induced 12 signaling inputs required for Ga -mediated cell pro- G1–S phase cell cycle progression of NIH3T3 cells 12 liferation. (Mitsui et al., 1997). However, the ectopic expression of Ga12/13 stimulated JNK activity with a concomitant inhibition of ERK1/2 pathway (Voyno-Yasenetskaya Ga13 stimulation of p38MAPg/ERK6 et al., 1996). Further analysis has identified MEK as the In contrast to Ga12, but similar to Gaq,Ga13 has been at which Ga12/13 exerts its inhibitory effects on shown to stimulate the p38MAPKg/ERK6 module ERK module. Although the underlying mechanism as (Marinissen et al., 2003). Considering the inhibitory well as the physiological significance of such regulation effect of Ga12 on MKK3/MKK6, it is more likely that is not fully known, recent studies have demonstrated Ga12 exerts similar attenuation of p38MAPKg/ERK6, that Ga12/13 can interact with PP5 to stimulate its at least in fibroblasts such as NIH3T3 cells. However, activity (Yamaguchi et al., 2002) and PP5 is involved in the transient expression of Ga13 has been shown to the downregulation of ERK module by dephosphory- stimulate p38MAPKg/ERK6 activity in NIH3T3 as well lating Raf-1 at Ser-338 (von Kriegsheim et al., 2006). as HEK293 cells. It has been suggested that ERK6 Therefore, although it remains to be established, it is stimulation of ATF2 and MEF2 is required for the possible that Ga12/13-mediated activation of PP5 is complete transactivation of c-Jun and the resulting involved in the inhibition of ERK1/2 modules. cellular transformation mediated by Ga13-coupled PAR-1 receptors (Marinissen et al., 2003). It has been identified that Rho is involved in the upstream stimula- Inhibition of p38MAPK by Ga 12 tion of the ERK6 module via PKN family of protein The expression of Ga has also been shown to attenuate 12 kinases (Marinissen et al., 2001, 2003). As Ga can the activation of the p38MAPK module (Dermott et al., 13 stimulate Rho via different GEFs, the mechanism by 2004). The inhibition was found to be at the level of the which Ga transmits signals to p38MAPKg/ERK6 is three MAP2Ks involved in the activation of p38MAPK, 13 likely to involve Rho and Rho-activated PKN. Since the namely MKK4, MKK3 and MKK6. It is interesting MAP3Ks such as MLK-like MAP triple kinase (Taka- that Ga stimulates JNK-specific MKK7 while inhibit- 12 hashi et al., 2003) or MLK-related kinase (Gross et al., ing MKK4, which is common to both JNK and 2002) can regulate ERK6 via MKK3/6 (Wang et al., p38MAPK. Thus, Ga appears to block decisively all 12 2000; Marinissen et al., 2001, 2003; Gross et al., 2002; the possible signaling routes that lead to the activation Takahashi et al., 2003), it is likely that Ga -activation of p38MAPK. However, it should be noted here that the 13 involves a phospho-relay module involving these inhibition of p38MAPK appears to be specific for Ga 12 upstream kinases. as the expression of Ga13 in NIH3T3 cells stimulates both JNK as well as p38MAPK activities (Marinissen et al., 2003). Although the molecular basis for such Ga12/13 stimulation of ERK5 selective inhibition of an MAPK at the levels of multiple In analysing the mechanism by which PAR-1 receptors, MAP2Ks by distinct a-subunits of the G12 family is not which can couple to both Gq and G12-family of G clear at present, at least two possible mechanisms can be proteins, activate ERK5, it was identified that the speculated: one involving the inhibition of p38MAPK- a-subunits of G12 as well as G13 are involved in the specific MKKs, presumably through the activation of a regulation of ERK5. Since activation of ERK5 by specific phosphatase, and the other involving a specific constitutively active Ga12 and Ga13 was insensitive to scaffolding protein that distinctly couples Ga12 to the the coexpression of the dominant negative mutants of MKK7–JNK module, while attenuating the MKK3/6/4- Ras, CDC42, Racor Rho, the activation appears to p38MAPK module. Since Ga12 has been shown to involve an alternate GTPase or mechanism (Fukuhara

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3133 et al., 2000b). An attractive candidate is Rap-1, which a G-protein-independent as well as G-protein-dependent can stimulate MEKK2 and hence the MEKK2–MEK5– pathways (Conway et al., 1999). In the G-protein ERK5 module (Wang et al., 2006a). However this independent, classical pathway, signaling to ERK possibility as well as the mechanism by which Ga12/13 involves PDGF-activated PDGFR autophosphoryla- communicates to Rap-1 needs to be experimentally tion on tyrosine residues, recruitment of adaptor verified. proteins and activation of p42/p44 MAPK. However, the G-protein-dependent pathway appears to involve Gi and b-arrestin-mediated endocytosis. In this pathway, endocytitic internalization of Gi-activated GPCRs along Receptor tyrosine kinases in G-protein-mediated with Gi-activated c-Src, PDGFR, ERK module and activation of MAPKs Grb-2 is facilitated by arrestin and dynamin II. It has been speculated that the close proximity and the possible In addition to the signaling pathways leading to the activation of Raf-1 by Src promotes the activation of regulation of MAPK modules by different G proteins, ERK module in endosomes (Waters et al., 2005). GPCRs have been shown to regulate ERK1/2 modules Further studies should define the role of such network- through the transactivation of RTKs. Such a novel role ing between RTKs and G proteins in specific cellular for EGFR in the activation of ERK by GPCRs was responses. identified with the observation that the stimulation of Rat1a cells with GPCR agonists including endothelin, LPA and thrombin stimulated the phosphorylation of EGFR and Her2 along with the activation of ERK2 Scaffold proteins in G-protein signaling to MAPKs (Daub et al., 1996). The findings that the treatment of As discussed in previous sections, diverse GPCRs, these cells with the EGFR inhibitor tyrphostin AG1478 or the expression of a dominant negative EGFR mutant through the a- as well as the bg-subunits of the four classes of G proteins, modulate the activities of different inhibited such ERK activation indicated the critical role MAPK modules. Interestingly, as can be seen in the case of EGFR in the activation of ERK by GPCRs (Daub et al., 1996). Further analyses revealed that EGFR plays of PAR- or thrombin receptors, a specific GPCR may recruit a-orbg-subunits from different classes of G a determinant role in GPCR-mediated activation of the proteins to activate an MAPK module in a cell type- or ERK module in a variety of cell types (Daub et al., 1997; context-specific manner. Even the individual a-orbg- Zwick et al., 1997; Eguchi et al., 1998). Primarily, G - i subunit may activate a specific MAPK module through and G -coupled receptors transactivate EGFR through q more than one mechanism in a context-specific manner. a novel mechanism involving ‘ecto-domain shedding’ In addition, the signaling molecules providing these and subsequent generation of soluble EGF-like growth mechanisms such as the small GTPase, their cognate factors that can stimulate EGFRs (Prenzel et al., 1999). GEFs, and the downstream kinases, are often shared by Recently, the molecular basis for GPCR-mediated different Ga-orbg-subunits activating distinct MAPK transactivation of EGFR has been summarized (Ohtsu et al., 2006). The transactivation of EGFR by GPCRs modules. So the major question is how do these distinct pathways from different G proteins elicit distinct involves both intracellular as well as extracellular functional responses involving specific MAPKs with mechanisms. Thus, EGFR transactivation by G -or i little or no signaling crosstalk with each other. Although G -coupled receptors has been shown to require the q studies from yeast have pointed out that specific scaffold intracellular generation of Ca2 þ (Zwick et al., 1997; proteins such as Ste5 can facilitate such context-specific Eguchi et al., 1998; Murasawa et al., 1998; Soltoff, 1998; linking of G-protein signaling to downstream MAPK Iwasaki et al., 1999), and activation of PKC, Src and modules (Herskowitz, 1995; Elion, 2001; Elion et al., Pyk2 (Gschwind et al., 2001). These pathways appear to 2005), the evidence for such scaffold proteins in converge on the activation of a family of a disintegrin mammalian G-protein signaling to MAPKs emerged and metalloprotease (ADAM) and/or matrix metallo- protease (MMP) proteins through phosphorylation as only recently. Of these scaffold proteins, some of them such as the b-arrestins play a catalytic as well as well as protein–protein interactions (Higashiyama and anchoring role in linking different GPCRs to MAPK Nanba, 2005; Ohtsu et al., 2006). The activated single transmembrane ADAMs, specifically ADAM-10, -12, signaling, whereas others such as MAP kinase organizer (MORG) and JLP play an anchoring role by tethering -15, -17, cleave the ectodomains of membrane-bound specific G proteins and MAPK modules in a context- EGFR ligands such as HB-EGF to generate soluble specific manner (Garrington and Johnson, 1999; Morrison EGF ligands that can activate EGFRs (Gschwind et al., and Davis, 2003; Kolch, 2005). 2001; Schafer et al., 2004; Higashiyama and Nanba, 2005; Ohtsu et al., 2006). Although these studies have shown the role of EGFR Catalytic role of b-arrestin in GPCR signaling to ERKs in G-protein signaling, it has also been shown that The arrestin family of proteins was initially identified signaling by RTKs to the ERK module can be enhanced to be involved in the desensitization of GPCRs by G proteins through a mechanism involving by facilitating their internalization (Freedman and b-arrestin. For example, it has been shown that the Lefkowitz, 1996; Kohout and Lefkowitz, 2003). Signaling activation of the ERK module by RTK ligands involves by agonist-activated GPCRs is terminated by the

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3134 GRK-family of / kinases that phosphory- MORG-1 suppressed ERK-1 activation by FBS (which late the receptors at their C-termini (Freedman and contains several ligands including LPA that can activate Lefkowitz, 1996). b-arrestins bind to such phospho- the respective GPCRs), it did not have any effect on rylated receptors and facilitate their internalization EGF-stimulated ERK response. Thus, MORG-1 that by endocytosis via -coated pits (Shenoy and can associate with the MAP3K, MAP2K, as well as the Lefkowitz, 2003). Interestingly, it has been shown that MAPK components of ERK1/2 module, appears to b-arrestin-1 can bind to both GRK-phosphorylated provide a scaffolding function specifically for GPCRs receptors, such b2-AR, as well as c-Src, targeting them such as LPA receptors. At present, it is not known to clathrin-coated pits where the complex acts as whether such a role of MORG1 involves its physical signaling hub for the activation of ERK1 and ERK2 interaction with specific GPCRs, Ga-orGbg-subunits. (Luttrell et al., 1999). In addition, it has been shown that Further studies should define the molecular mechanism b-arrestin-2 can sequester components of the ERK by which MORG-1 links GPCR signaling to ERK1/2. module such as Raf-1, MEK1/2 and ERK1/2 via its interaction with Raf-1 (Luttrell et al., 2001). The b- Role of JLPin linking G a12/13 to JNK module arrestin scaffold containing the components of the ERK JLP was initially identified as a Leucine Zipper domain module and the receptor are internalized via clathrin- containing protein that can interact with Max (Lee coated pits into endosomal vesicles where the receptor et al., 2002). In addition, it has been observed that can activate the ERK module (Lefkowitz and Shenoy, JLP can interact with JNK, p38MAPK, MKK4 2005). Although the mechanism by which the inter- and MEKK3. Furthermore, it was observed that the nalized GPCR can activate the ERK module is yet to expression of JLP potentiated cytotoxic stress-mediated be elucidated fully, it is possible that the other activation of JNK (Lee et al., 2002). Recent studies have co-internalized intermediary molecules such as Src play shown that JLP can interact with Ga as well as Ga a role in providing such a link. Interestingly, it has been 12 13 and provide a scaffolding role for JNK activation by noted that the ERK module activated by b-arrestin has these a-subunits (Kashef et al., 2005). It has been slower kinetics of activation than that mediated by G demonstrated that JLP interacts with the a-subunits of proteins (Lefkowitz and Shenoy, 2005). In addition, it the G family through the C-terminal region of JLP, has been observed that the ERK module activated by 12 spanning amino acids 1165–1307. Using the expression b-arrestin is not involved in nuclear signaling (DeFea of this Ga interacting domain or siRNA-mediated et al., 2000; Tohgo et al., 2002). Thus, b-arrerestin-2- 12/13 silencing of JLP, it was demonstrated that the disruption mediated activation of ERK module by different of the JLP–Ga interaction leads to the attenuation of GPCRs appears to be involved in the cytosolic responses 13 LPA- as well as Ga -mediated activation of JNK. that require slower but persistent activation of ERKs 13 Furthermore, it has been observed that silencing JLP (DeFea et al., 2000; Tohgo et al., 2002; Lefkowitz and inhibits retinoicacid-mediated endodermal differentia- Shenoy, 2005). tion of P19 cells (Kashef et al., 2006), which is critically In addition to the activation of the ERK module, dependent on Ga -mediated activation of JNK (Jho b-arrestin-2 also appears to be involved in regulating 13 et al., 1997; Wang et al., 2002; Lee et al., 2004). In this the JNK module. Using yeast two hybrid screening, it context, it is significant to note that JLP is the first was identified that b-arrestin-2 can interact with JNK3 known mammalian , albeit similar to (McDonald et al., 2000). Further analyses indicated that Ste5, in tethering all the signaling components from the b-arrestin-2 interacts with the MAP3K, ASK1. In proximal Ga-subunit to the downstream MAPK. Since addition, MKK-4 also can be colocalized along with the Ga12/13-interacting C-terminus of JLP is conserved b-arrestin, ASK1 and JNK3 in a complex. It has also among all the JIP family of scaffold proteins, it is been observed that angiotensin II type IA receptor possible that JIPs may also be involved in linking Ga stimulated the colocalization of b-arrestin-2 and JNK3 12/ 13 or other a-subunits to JNK or p38MAPK modules in to intracellular vesicles along with the activation of a context-specific manner. This, as well as other finer et al JNK3 (McDonald ., 2000). However, it is not clear details of the JLP scaffold, such as its interactions with whether b-arrestin plays a catalytic role in stimulating specific small GTPases and their GEFs, remains to be JNK3 in this context. established. Similarly, further studies should define the role of such scaffold proteins in Ga-orGbg-mediated activation of other MAPK modules. Role of MORG-1 in linking GPCRs to ERK1/2 pathway MORG-1 was identified as a 35 kDa protein that interacts with MEK-partner 1 proteins in a yeast two hybrid screen (Vomastek et al., 2004). Further charac- Conclusion terization of this WD-40 motif containing protein indicated that it can associate with Raf-1, B-Raf, Recent studies have contributed to a greater under- MEK1, MEK2, ERK1 and ERK2. Although the ectopic standing of the mechanisms by which the GPCR family expression of MORG1 enhanced LPA or PMA- of cell surface receptors critically regulate different mediated activation of ERK, it failed to have such aspects of via the MAPK signaling modules. stimulatory effects on PDGFR- or EGFR-mediated As the signaling nexus between different G proteins activation of ERK. In addition, although silencing and distinct MAPK modules are being realized, the

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3135 pathophysiological roles of aberrant- or asynchronous cancers (Mills and Moolenaar, 2003; Booden et al., signaling by these pathways are becoming increasingly 2004; Daaka, 2004; Boire et al., 2005). The findings that clear. However, as presented in this review, several all of these GPCRs, their cognate G proteins and the G- aspects of these pathways involved in G-protein- protein oncogenes gsp, gip2 and gep converge on the mediated MAPK activation remain to be resolved. activation of distinct MAPK modules, taken together For example, the molecular basis for context-specific with the observation that GPCR-mediated signaling signaling, the spatio-temporal organization of G-protein– pathways are well-validated drug targets, underscore MAPK signaling complexes, the effects of these path- the critical need for defining these pathways further so ways in relation to receptor tyrosine kinase- or cytokine that better therapeutictargets can be identified. receptor-mediated activation of MAPKs, and most importantly, the potential role of G-protein-regulated MAPK networks in tumorigenesis and progression Acknowledgements remain to be fully characterized. A case to the point is the identification of the potential role of LPA- and The study was supported by a grant from NIH (GM 49897). PAR-receptor-mediated signaling pathways in the - Critical reading of the paper by Dr Rashmi Kumar and sis and progression of ovarian, breast and prostate Mr Jake Gardner are gratefully acknowledged.

References

Ahn YH, Han JH, Hong SH. (2006). Rap1 and p38 MAPK Booden MA, Eckert LB, Der CJ, Trejo J. (2004). Persistent mediate 8-chloro-cAMP-induced growth inhibition in signaling by dysregulated thrombin receptor trafficking mouse fibroblast DT cells. J Cell Physiol 209: 1039–1045. promotes breast carcinoma cell invasion. Mol Cell Biol 24: Ahn YH, Jung JM, Hong SH. (2005). 8-Chloro-cyclic AMP- 1990–1999. induced growth inhibition and apoptosis is mediated by p38 Booden MA, Siderovski DP, Der CJ. (2002). Leukemia- mitogen-activated protein kinase activation in HL60 cells. associated Rho guanine nucleotide exchange factor pro- Cancer Res 65: 4896–4901. motes Gaq-coupled activation of RhoA. Mol Cell Biol 22: Althoefer H, Eversole-Cire P, Simon MI. (1997). Constitu- 4053–4061. tively active Gaq and Ga13 trigger apoptosis through Bos JL. (1998). All in the family? New insights and questions different pathways. J Biol Chem 272: 24380–24386. regarding interconnectivity of Ras, Rap1 and Ral. EMBO J Ambrosini A, Tininini S, Barassi A, Racagni G, Sturani E, 17: 6776–6782. Zippel R. (2000). cAMP cascade leads to Ras activation in Bos JL. (2006). Epacproteins: multi-purpose cAMPtargets. cortical neurons. Mol Brain Res 75: 54–60. Trends Biochem Sci 31: 680–686. Amsen EM, Pham N, Pak Y, Rotin D. (2006). The guanine Brunet A, Roux D, Lenormand P, Dowd S, Keyse S, nucleotide exchange factor CNrasGEF regulates melanogenesis Pouyssegur J. (1999). Nuclear translocation of p42/p44 and cell survival in melanoma cells. JBiolChem281: 121–128. mitogen-activated protein kinase is required for growth Aragay AM, Collins LR, Post GR, Watson AJ, Feramisco JR, factor-induced and cell cycle entry. EMBO J Brown JH et al. (1995). G12 requirement for thrombin- 18: 664–674. stimulated gene expression and DNA synthesis in 1321N1 Buchsbaum RJ, Connolly BA, Feig LA. (2002). Interaction of astrocytoma cells. J Biol Chem 270: 20073–20077. Rac exchange factors Tiam1 and Ras-GRF1 with a scaffold Arai K, Maruyama Y, Nishida M, Tanabe S, Takagahara S, for the p38 mitogen-activated protein kinase cascade. Mol Kozasa T et al. (2003). Differential requirement of Ga12, Cell Biol 22: 4073–4085. Ga13,Gaq, and Gbg for endothelin-1-induced c-Jun NH2- Busca R, Abbe P, Mantoux F, Aberdam E, Peyssonnaux C, terminal kinase and extracellular signal-regulated kinase Eychene A et al. (2000). Ras mediates the cAMP-dependent activation. Mol Pharmacol 63: 478–488. activation of extracellular signal-regulated kinases (ERKs) Araki S, Haneda M, Togawa M, Kikkawa R. (1997). in melanocytes. EMBO J 19: 2900–2910. Endothelin-1 activates c-Jun NH2-terminal kinase in Camps M, Carozzi A, Schnabel P, Scheer A, Parker PJ, mesangial cells. Kidney Int 51: 631–639. Gierschik P. (1992). Isozyme-selective stimulation of Berestetskaya YV, Faure MP, Ichijo H, Voyno-Yasenetskaya -b2 by G protein bg-subunits. Nature 360: TA. (1998). Regulation of apoptosis by a-subunits of G12 684–686. and G13 proteins via apoptosis signal-regulating kinase-1. Chadee DN, Kyriakis JM. (2004). MLK3 is required for J Biol Chem 273: 27816–27823. mitogen activation of B-Raf, ERK and cell proliferation. Blaukat A, BaracA, Cross MJ, Offermanns S, DikicI. (2000). Nat Cell Biol 6: 770–776. G protein-coupled receptor-mediated mitogen-activated Chan AM, Fleming TP, McGovern ES, Chedid M, Miki T, protein kinase activation through cooperation of Gaq and Aaronson SA. (1993). Expression cDNA cloning of a Gai signals. Mol Cell Biol 20: 6837–6848. transforming gene encoding the wild-type Ga12 gene Blaukat A, Ivankovic-Dikic I, Gronroos E, Dolfi F, Tokiwa product. Mol Cell Biol 13: 762–768. G, Vuori K et al. (1999). Adaptor proteins Grb2 and Crk Chang L, Karin M. (2001). Mammalian MAP kinase couple Pyk2 with activation of specific mitogen-activated signalling cascades. Nature 410: 37–40. protein kinase cascades. J Biol Chem 274: 14893–14901. Chikumi H, Fukuhara S, Gutkind JS. (2002). Regulation of G Boire A, CovicL, Agarwal A, JacquesS, Sherifi S, Kuliopulos protein-linked guanine nucleotide exchange factors for Rho, A. (2005). PAR1 is a matrix metalloprotease-1 receptor that PDZ-RhoGEF, and LARG by tyrosine phosphorylation: promotes invasion and tumorigenesis of breast cancer cells. evidence of a role for kinase. J Biol Chem Cell 120: 303–313. 277: 12463–12473.

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3136 Coffield VM, Helms WS, Jiang Q, Su L. (2004). Ga13 mediates receptors. Convergence of Gi- and Gq-mediated pathways a signal that is essential for proliferation and survival of on calcium/calmodulin, Pyk2, and Src kinase. J Biol Chem thymocyte progenitors. J Exp Med 200: 1315–1324. 272: 19125–19132. Collins LR, Minden A, Karin M, Brown JH. (1996). Ga12 Deng J, Kawakami Y, Hartman SE, Satoh T, Kawakami T. stimulates c-Jun NH2-terminal kinase through the small G (1998). Involvement of Ras in Bruton’s tyrosine proteins Ras and Rac. J Biol Chem 271: 17349–17353. kinase-mediated JNK activation. J Biol Chem 273: Collins LR, Ricketts WA, Olefsky JM, Brown JH. (1997). The 16787–16791. G12 coupled thrombin receptor stimulates mitogenesis Denis-Henriot D, de Mazancourt P, Morot M, Giudicelli Y. through the ShcSH2 domain. Oncogene 15: 595–600. (1998). Mutant alpha-subunit of the G protein G12 activates Conklin BR, Bourne HR. (1993). Structural elements of Ga proliferation and inhibits differentiation of 3T3-F442A subunits that interact with Gbg, receptors, and effectors. preadipocytes. Endocrinology 139: 2892–2899. Cell 73: 631–641. Dermott JM, Ha JH, Lee CH, Dhanasekaran N. (2004). Conway AM, Rakhit S, Pyne S, Pyne NJ. (1999). Platelet- Differential regulation of Jun N-terminal kinase and derived-growth-factor stimulation of the p42/p44 mitogen- p38MAP kinase by Ga12. Oncogene 23: 226–232. activated protein kinase pathway in airway smooth muscle: Dhanasekaran DN. (2006). Transducing the signals: a G role of pertussis-toxin-sensitive G-proteins, c-Src tyrosine protein takes a new identity. Sci STKE 347: pe31. kinases and phosphoinositide 3-kinase. Biochem J 337: Dhanasekaran N, Dermott JM. (1996). Signaling by the G12 171–177. class of G proteins. Cell Signal 8: 235–245. Cook SJ, McCormick F. (1993). Inhibition by cAMP of Ras- Dhanasekaran N, Heasley LE, Johnson GL. (1995). G dependent activation of Raf. Science 262: 1069–1072. protein-coupled receptor systems involved in cell growth Cook SJ, Rubinfeld B, Albert I, McCormick F. (1993). and oncogenesis. Endocr Rev 16: 259–270. RapV12 antagonizes Ras-dependent activation of ERK1 Dhanasekaran N, Prasad MV. (1998). G protein subunits and and ERK2 by LPA and EGF in Rat-1 fibroblasts. EMBO J cell proliferation. Biol Signals Recept 7: 109–117. 12: 3475–3485. Dhanasekaran N, Premkumar Reddy E. (1998). Signaling by Coso OA, Chiariello M, KalinecG, Kyriakis JM, Woodgett J, dual specificity kinases. Oncogene 17: 1447–1455. Gutkind JS. (1995). Transforming G protein-coupled Dhillon AS, Pollock C, Steen H, Shaw PE, Mischak H, Kolch receptors potently activate JNK (SAPK). Evidence for a W. (2002). Cyclic AMP-dependent kinase regulates Raf-1 divergence from the tyrosine kinase signaling pathway. kinase mainly by phosphorylation of serine 259. Mol Cell J Biol Chem 270: 5620–5624. Biol 22: 3237–3246. Coso OA, Teramoto H, Simonds WF, Gutkind JS. (1996). DikicI, Tokiwa G, Lev S, Courtneidge SA, SchlessingerJ. Signaling from G protein-coupled receptors to c-Jun kinase (1996). A role for Pyk2 and Srcin linking G-protein-coupled involves beta gamma subunits of heterotrimericG proteins receptors with MAP kinase activation. Nature 383: 547–550. acting on a Ras and Rac1-dependent pathway. J Biol Chem Dolfi F, Garcia-Guzman M, Ojaniemi M, Nakamura H, 271: 3963–3966. Matsuda M, Vuori K. (1998). The adaptor protein Crk Crespo P, Bustelo XR, Aaronson DS, Coso OA, Lopez- connects multiple cellular stimuli to the JNK signaling Barahona M, Barbacid M et al. (1996). Rac-1 dependent pathway. Proc Natl Acad Sci USA 95: 15394–15399. stimulation of the JNK/SAPK signaling pathway by Vav. Dumaz N, Marais R. (2005). Integrating signals between Oncogene 13: 455–460. cAMP and the RAS/RAF/MEK/ERK signalling pathways. Crespo P, Cachero TG, Xu N, Gutkind JS. (1995). Dual effect FEBS J 272: 3491–3504. of b-adrenergic receptors on mitogen-activated protein Edamatsu H, Kaziro Y, Itoh H. (1998). Expression of an kinase. Evidence for a bg-dependent activation and a Gas- oncogenic mutant G alpha i2 activates Ras in Rat-1 cAMP-mediated inhibition. J Biol Chem 270: 25259–25265. fibroblast cells. FEBS Lett 440: 231–234. Crespo P, Xu N, Simonds WF, Gutkind JS. (1994). Ras- Eguchi S, Numaguchi K, Iwasaki H, Matsumoto T, dependent activation of MAP kinase pathway mediated by Yamakawa T, Utsunomiya H et al. (1998). Calcium- G-protein bg subunits. Nature 369: 418–420. dependent epidermal growth factor receptor transactivation Daaka Y. (2004). G proteins in cancer: the prostate cancer mediates the angiotensin II-induced mitogen-activated paradigm. Sci STKE 216: re2. protein kinase activation in vascular smooth muscle cells. Daub H, Wallasch C, Lankenau A, Herrlich A, Ullrich A. J Biol Chem 273: 8890–8896. (1997). Signal characteristics of G protein-transactivated Elion EA, Qi M, Chen W. (2005). Signal transduction. EGF receptor. EMBO J 16: 7032–7044. Signaling specificity in yeast. Science 307: 687–688. Daub H, Weiss FU, Wallasch C, Ullrich A. (1996). Role Elion EA. (2001). The Ste5p scaffold. J Cell Sci 114: of transactivation of the EGF receptor in signalling by 3967–3978. G-protein-coupled receptors. Nature 379: 557–560. Enserink JM, Christensen AE, de Rooji J, van Triest M, Davis RJ. (2000). Signal transduction by the JNK group of Schwede F, Genieser HG et al. (2002). A novel Epac-specific MAP kinases. Cell 103: 239–252. cAMP analogue demonstrates independent regulation of de Rooij J, Zwartkruis FJ, Verheijen MH, Cool RH, Nijman Rap1 and ERK. Nat Cell Biol 4: 901–906. SM, Wittinghofer A et al. (1998). Epacis a Rap1 guanine- Fanger GR, Johnson NL, Johnson GL. (1997). MEK kinases nucleotide-exchange factor directly activated by cyclic are regulated by EGF and selectively interact with Rac/ AMP. Nature 396: 474–477. Cdc42. EMBO J 16: 4961–4972. DeFea KA, Zalevsky J, Thoma MS, Dery O, Mullins RD, Faure M, Voyno-Yasenetskaya TA, Bourne HR. (1994). Bunnett NW. (2000). arrestin-dependent endocytosis of cAMP and bg subunits of heterotrimericG proteins proteinase-activated receptor 2 is required for intracellular stimulate the mitogen-activated protein kinase pathway in targeting of activated ERK1/2. J Cell Biol 148: 1267–1281. COS-7 cells. J Biol Chem 269: 7851–7854. Della Rocca GJ, van Biesen T, Daaka Y, Luttrell DK, Luttrell Fleming Y, Armstrong CG, Morrice N, Paterson A, LM, Lefkowitz RJ. (1997). Ras-dependent mitogen- Goedert M, Cohen P. (2000). Synergisticactivationof activated protein kinase activation by G protein-coupled stress-activated protein kinase 1/c-Jun N-terminal kinase

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3137 (SAPK1/JNK) isoforms by mitogen-activated protein Gupta SK, Gallego C, Lowndes JM, Pleiman CM, Sable C, kinase kinase 4 (MKK4) and MKK7. Biochem J 352: Eisfelder BJ et al. (1992b). Analysis of the fibroblast 145–154. transformation potential of GTPase-deficient gip2 onco- Freedman NJ, Lefkowitz RJ. (1996). Desensitization of genes. Mol Cell Biol 12: 190–197. G protein-coupled receptors. Recent Prog Horm Res 51: Gutkind JS, Coso OA, Xu N. (1998). Ga12- and Ga13-Subunits 319–351. of heterotrimericG proteins: a novel family of oncogenes. Fukuhara S, Chikumi H, Gutkind JS. (2000a). Leukemia- In: Spiegel S (ed). G Proteins, Receptors, and Diseases. associated Rho guanine nucleotide exchange factor (LARG) Humana Press: Totowa, NJ, USA, pp 101–117. links heterotrimericG proteins of the G 12 family to Rho. Gutkind JS. (1998). Cell growth control by G protein-coupled FEBS Lett 485: 183–188. receptors: from signal transduction to signal integration. Fukuhara S, Marinissen MJ, Chiariello M, Gutkind JS. Oncogene 17: 1331–1342. (2000b). Signaling from G protein-coupled receptors to Gutkind JS. (2000). Regulation of mitogen-activated protein ERK5/Big MAPK 1 involves Gaq and Ga12/13 families of kinase signaling networks by G protein-coupled receptors. heterotrimeric G proteins. Evidence for the existence of a Sci STKE 40: re1. novel Ras and Rho-independent pathway. J Biol Chem 275: Hafner S, Adler HS, Mischak H, Janosch P, Heidecker G, 21730–21736. Wolfman A et al. (1994). Mechanism of inhibition of Raf-1 Garrington TP, Johnson GL. (1999). Organization and by protein kinase A. Mol Cell Biol 14: 6696–6703. regulation of mitogen-activated protein kinase signaling Hart MJ, Jiang X, Kozasa T, Roscoe W, Singer WD, Gilman pathways. Curr Opin Cell Biol 11: 211–218. AG et al. (1998). Direct stimulation of the guanine Gerasimovskaya EV, Ahmad S, White CW, Jones PL, nucleotide exchange activity of p115 RhoGEF by Ga13. Carpenter TC, Stenmark KR. (2002). Extracellular ATP is Science 280: 2112–2114. an autocrine/paracrine regulator of hypoxia-induced adven- Hawes BE, van Biesen T, Koch WJ, Luttrell LM, Lefkowitz titial fibroblast growth: signaling through extracellular RJ. (1995). Distinct pathways of Gi- and Gq-mediated signal-regulated kinase-1/2 and the Egr-1 transcription mitogen-activated protein kinase activation. J Biol Chem factor. J Biol Chem 277: 44638–44650. 270: 17148–17153. Ghosh SK, Gadiparthi L, Zeng ZZ, Bhanoori M, Tellez C, Hepler JR, Gilman AG. (1992). G proteins. Trends Biochem Bar-Eli M et al. (2002). ATF-1 mediates protease-activated Sci 17: 383–387. receptor-1 but not receptor tyrosine kinase-induced DNA Herskowitz I. (1995). MAP kinase pathways in yeast: for synthesis in vascular smooth muscle cells. J Biol Chem 277: mating and more. Cell 80: 187–197. 21325–21331. Higashijima T, Burnier J, Ross EM. (1990). Regulation of Gi Gilman AG. (1987). G proteins: transducers of receptor- and Go by mastoparan, related amphiphilicpeptides, and generated signals. Annu Rev Biochem 56: 615–649. hydrophobic amines. Mechanism and structural determi- Girardin SE, Yaniv M. (2001). A direct interaction between nants of activity. J Biol Chem 265: 14176–14186. JNK1 and CrkII is critical for Rac1-induced JNK activa- Higashiyama S, Nanba D. (2005). ADAM-mediated ectodo- tion. EMBO J 20: 3437–3446. main shedding of HB-EGF in receptor cross-talk. Biochim Gotoh T, Hattori S, Nakamura S, Kitayama H, Noda M, Biophys Acta 1751: 110–117. Takai Y et al. (1995). Identification of Rap1 as a target for Hinshaw JE, Schmid SL. (1995). Dynamin self-assembles into the Crk SH3 domain-binding guanine nucleotide-releasing rings suggesting a mechanism for coated vesicle budding. factor C3G. Mol Cell Biol 15: 6746–6753. Nature 374: 190–192. Gould KL, Woodgett JR, Cooper JA, Buss JE, Shalloway D, Houslay MD. (2006). A RSK(y) relationship with promis- Hunter T. (1985). Protein kinase C phosphorylates pp60src cuous PKA. Sci STKE 349: pe32. at a novel site. Cell 42: 849–857. Huang CC, You JL, Wu MY, Hsu KS. (2004). Rap1-induced Graves LM, Bornfeldt KE, Raines EW, Potts BC, Macdonald p38 mitogen-activated protein kinase activation facilitates SG, Ross R et al. (1993). Protein kinase A antagonizes AMPA receptor trafficking via the GDI.Rab5 complex. platelet-derived growth factor-induced signaling by mito- Potential role in (S)-3,5-dihydroxyphenylglycene-induced gen-activated protein kinase in human arterial smooth long term depression. J Biol Chem 279: 12286–12292. muscle cells. Proc Natl Acad Sci USA 90: 10300–10304. Hubbard KB, Hepler JR. (2006). Cell signalling diversity of Gros R, Ding Q, Chorazyczewski J, Pickering JG, Limbird the Gqa family of heterotrimericG proteins. Cell Signal 18: LE, Feldman RD. (2006). Adenylyl cyclase isoform-selective 135–150. regulation of vascular smooth muscle proliferation and Ichiba T, Hashimoto Y, Nakaya M, Kuraishi Y, Tanaka S, cytoskeletal reorganization. Circ Res 99: 845–852. Kurata T et al. (1999). Activation of C3G guanine Gross EA, Callow MG, Waldbaum L, Thomas S, Ruggieri R. nucleotide exchange factor for Rap1 by phosphorylation (2002). MRK, a mixed lineage kinase-related molecule that of tyrosine 504. J Biol Chem 274: 14376–14381. plays a role in gamma-radiation-induced cell cycle arrest. Iwasaki H, Eguchi S, Ueno H, Marumo F, Hirata Y. (1999). J Biol Chem 277: 13873–13882. Endothelin-mediated vascular growth requires p42/p44 Gschwind A, Zwick E, Prenzel N, Leserer M, Ullrich A. mitogen-activated protein kinase and p70 S6 kinase cascades (2001). Cell communication networks: epidermal growth via transactivation of epidermal growth factor receptor. factor receptor transactivation as the paradigm for inter- Endocrinology 140: 4659–4668. receptor signal transmission. Oncogene 20: 1594–1600. Jho EH, Davis RJ, Malbon CC. (1997). c-Jun amino-terminal Guo FF, Kumahara E, Saffen D. (2001). A CalDAG-GEFI/ kinase is regulated by Ga12/Ga13 and obligate for differen- Rap1/B-Raf cassette couples M1 muscarinic acetylcholine tiation of P19 embryonal carcinoma cells by retinoic acid. receptors to the activation of ERK1/2. J Biol Chem 276: J Biol Chem 272: 24468–24474. 25568–25581. Jho EH, Malbon CC. (1997). Ga12 and Ga13 mediate Gupta SK, Gallego C, Johnson GL, Heasley LE. (1992a). differentiation of P19 mouse embryonal carcinoma MAP kinase is constitutively activated in gip2 and src cells in response to retinoic acid. J Biol Chem 272: transformed rat 1a fibroblasts. J Biol Chem 267: 7987–7990. 24461–24467.

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3138 Jiang H, Wu D, Simon MI. (1993). The transforming activity Kranenburg O, Verlaan I, Moolenaar WH. (1999b). of activated Ga12. FEBS Lett 330: 319–322. Gi-mediated tyrosine phosphorylation of Grb2 (growth- Jiang Y, Ma W, Wan Y, Kozasa T, Hattori S, Huang XY. factor-receptor-bound protein 2)-bound dynamin-II by (1998). The G protein Ga12 stimulates Bruton’s tyrosine lysophosphatidicacid. Biochem J 339: 11–14. kinase and a rasGAP through a conserved PH/BM domain. Kranenburg O, Verlaan I, Hordijk PL, Moolenaar WH. (1997). Nature 395: 808–813. Gi-mediated activation of the Ras/MAP kinase pathway Johnson GL, Dhanasekaran N. (1989). The G-protein involves a 100 kDa tyrosine-phosphorylated Grb2 SH3 family and their interaction with receptors. Endocr Rev 10: binding protein, but not Srcnor Shc. EMBO J 16: 3097–3105. 317–331. Krump E, Sanghera JS, Pelech SL, Furuya W, Grinstein S. Kam AY, Chan AS, Wong YH. (2004). Phosphatidylinositol-3 (1997). Chemotactic peptide N-formyl-met-leu-phe activa- kinase is distinctively required for m-, but not k-opioid tion of p38 mitogen-activated protein kinase (MAPK) and receptor-induced activation of c-Jun N-terminal kinase. MAPK-activated protein kinase-2 in human neutrophils. J Neurochem 89: 391–402. J Biol Chem 272: 937–944. Kashef K, Lee CM, Ha JH, Reddy EP, Dhanasekaran DN. Kumar RN, Ha JH, Radhakrishnan R, Dhanasekaran DN. (2005). JNK-interacting leucine zipper protein is a novel (2006). Transactivation of platelet-derived growth factor scaffolding protein in the Ga13 signaling pathway. Biochem receptor alpha by the GTPase-deficient activated mutant of 44: 14090–14096. Ga12. Mol Cell Biol 26: 50–62. Kashef K, Xu H, Reddy EP, Dhanasekaran DN. (2006). Kumar S, McDonnell PC, Gum RJ, Hand AT, Lee JC, Young Endodermal differentiation of murine embryonic carcinoma PR. (1997). Novel homologues of CSBP/p38 MAP kinase: cells by retinoic acid requires JLP, a JNK-scaffolding activation, specificity and sensitivity to inhibition protein. J Cell Biochem 98: 715–722. by pyridinyl imidazoles. Biochem Biophys Res Commun 235: Katz A, Wu D, Simon MI. (1992). Subunits bg of hetero- 533–538. trimericG protein activate b2 isoform of phospholipase C. L’Allemain G, Pouyssegur J, Weber MJ. (1991). p42/mitogen- Nature 360: 686–689. activated protein kinase as a converging target for different Khokhlatchev AV, Canagarajah B, Wilsbacher J, Robinson growth factor signaling pathways: use of pertussis toxin as a M, Atkinson M, Goldsmith E et al. (1998). Phosphorylation discrimination factor. Cell Regul 2: 675–684. of the MAP kinase ERK2 promotes its homodimerization Landis CA, Masters SB, Spada A, Pace AM, Bourne HR, and nuclear translocation. Cell 93: 605–615. Vallar L. (1989). GTPase inhibiting mutations activate the a Kim KY, Kim BC, Xu Z, Kim SJ. (2004). Mixed lineage chain of Gs and stimulate adenylyl cyclase in human kinase 3 (MLK3)-activated p38 MAP kinase mediates pituitary tumours. Nature 340: 692–696. transforming growth factor-beta-induced apoptosis in he- Laroche-Joubert N, Marsy S, Michelet S, Imbert-Teboul M, patoma cells. J Biol Chem 279: 29478–29484. Doucet A. (2002). Protein kinase A-independent activation Kitayama H, Sugimoto Y, Matsuzaki T, Ikawa Y, Noda M. of ERK and H,K-ATPase by cAMP in native kidney cells: (1989). A ras-related gene with transformation suppressor role of EpacI. J Biol Chem 277: 18598–18604. activity. Cell 56: 77–84. Lawler S, Fleming Y, Goedert M, Cohen P. (1998). Synergistic Kiyokawa E, Hashimoto Y, Kobayashi S, Sugimura H, activation of SAPK1/JNK1 by two MAP kinase kinases in Kurata T, Matsuda M. (1998). Activation of Rac1 vitro. Curr Biol 8: 1387–1390. by a Crk SH3-binding protein, DOCK180. Genes Dev 12: Lechner C, Zahalka MA, Giot JF, Moller NP, Ullrich A. 3331–3336. (1996). ERK6, a mitogen-activated protein kinase involved Koch WJ, Hawes BE, Allen LF, Lefkowitz RJ. (1994). Direct in C2C12 myoblast differentiation. Proc Natl Acad Sci USA evidence that Gi-coupled receptor stimulation of mitogen- 93: 4355–4359. activated protein kinase is mediated by Gbg activation of Lee CL, Onesime D, Reddy CD, Dhanasekaran N, Reddy EP. p21ras. Proc Natl Acad Sci USA 91: 12706–12710. (2002). JLP: A scaffolding protein that tethers JNK/ Kodama H, Fukuda K, Takahashi E, Tahara S, Tomita Y, p38MAPK signaling modules and transcription factors. Ieda M et al. (2003). Selective involvement of p130Cas/Crk/ Proc Natl Acad Sci USA 99: 14189–14194. Pyk2/c-Src in endothelin-1-induced JNK activation. Hyper- Lee YN, Malbon CC, Wang HY. (2004). Ga13 signals via tension 41: 1372–1379. p115RhoGEF cascades regulating JNK1 and primitive Kohout TA, Lefkowitz RJ. (2003). Regulation of G protein- endoderm formation. J Biol Chem 279: 54896–54904. coupled receptor kinases and arrestins during receptor Lefkowitz RJ, Shenoy SK. (2005). Transduction of receptor desensitization. Mol Pharmacol 63: 9–18. signals by b-arrestins. Science 308: 512–517. Kolch W, Heidecker G, Kochs G, Hummel R, Vahidi H, Lev S, Moreno H, Martinez R, Canoll P, Peles E, Musacchio Mischak H et al. (1993). Protein kinase C alpha activates JM et al. (1995). Protein tyrosine kinase PYK2 involved in RAF-1 by direct phosphorylation. Nature 364: 249–252. Ca(2+)-induced regulation of ion channel and MAP kinase Kolch W. (2005). Coordinating ERK/MAPK signalling functions. Nature 376: 737–745. through scaffolds and inhibitors. Nat Rev Mol Cell Biol 6: Lisnock J, Griffin P, Calaycay J, Frantz B, Parsons J, O’Keefe 827–837. SJ et al. (2000). Activation of JNK3a1 requires both MKK4 Kramer RM, Roberts EF, Strifler BA, Johnstone EM. (1995). and MKK7: kinetic characterization of in vitro phosphory- Thrombin induces activation of p38 MAP kinase in human lated JNK3a1. Biochemistry 39: 3141–3148. platelets. J Biol Chem 270: 27395–27398. Liu JL, Blakesley VA, Gutkind JS, LeRoith D. (1997). The Kranenburg O, Moolenaar WH. (2001). Ras-MAP kinase constitutively active mutant Ga13 transforms mouse fibro- signaling by lysophosphatidicacidand other G protein- blast cells deficient in -like growth factor-I receptor. coupled receptor agonists. Oncogene 20: 1540–1546. J Biol Chem 272: 29438–29441. Kranenburg O, Verlaan I, Moolenaar WH. (1999a). Dynamin Lowndes JM, Gupta SK, Osawa S, Johnson GL. (1991). is required for the activation of mitogen-activated protein GTPase-deficient Gai2 oncogene gip2 inhibits adenylylcy- (MAP) kinase by MAP kinase kinase. J Biol Chem 274: clase and attenuates receptor-stimulated 35301–35304. activity. J Biol Chem 266: 14193–14197.

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3139 Luttrell LM, Ferguson SS, Daaka Y, Miller WE, Maudsley S, Miyamoto Y, Yamauchi J, Itoh H. (2003). Src kinase regulates Della Rocca GJ et al. (1999). Arrestin-dependent formation the activation of a novel FGD-1-related Cdc42 guanine of b2 adrenergic receptor-Src protein kinase complexes. nucleotide exchange factor in the signaling pathway Science 283: 655–661. from the endothelin A receptor to JNK. J Biol Chem 278: Luttrell LM, Hawes BE, van Biesen T, Luttrell DK, Lansing 29890–29900. TJ, Lefkowitz RJ. (1996). Role of c-Src tyrosine kinase in G Mochizuki N, Ohba Y, Kiyokawa E, Kurata T, Murakami T, protein-coupled receptor- and Gbg subunit-mediated activa- Ozaki T et al. (1999). Activation of the ERK/MAPK tion of mitogen-activated protein kinases. J Biol Chem 271: pathway by an isoform of rap1GAP associated with Gai. 19443–19450. Nature 400: 891–894. Luttrell LM, Roudabush FL, Choy EW, Miller WE, Field Mochizuki N, Ohba Y, Kobayashi S, Otsuka N, Graybiel AM, ME, Pierce KL et al. (2001). Activation and targeting of Tanaka S et al. (2000). Crk activation of JNK via C3G and extracellular signal-regulated kinases by b-arrestin scaffolds. R-Ras. J Biol Chem 275: 12667–12671. Proc Natl Acad Sci USA 98: 2449–2454. Morrison DK, Davis RJ. (2003). Regulation of MAP kinase Lyons J, Landis CA, Harsh G, Vallar L, Grunewald K, signaling modules by scaffold proteins in mammals. Annu Feichtinger H et al. (1990). Two G protein oncogenes in Rev Cell Dev Biol 19: 91–118. human endocrine tumors. Science 249: 655–659. Morrison DK, Heidecker G, Rapp UR, Copeland TD. (1993). Mao J, Xie W, Yuan H, Simon MI, Mano H, Wu D. (1998). Identification of the major phosphorylation sites of the Tec/Bmx non-receptor tyrosine kinases are involved in Raf-1 kinase. J Biol Chem 268: 17309–17316. regulation of Rho and serum response factor by Ga12/13. Moyers JS, Bouton AH, Parsons SJ. (1993). The sites of EMBO J 17: 5638–5646. phosphorylation by protein kinase C and an intact SH2 Marinissen MJ, Chiariello M, Gutkind JS. (2001). Regulation domain are required for the enhanced response to b- of gene expression by the small GTPase Rho through the adrenergic agonists in cells overexpressing c-src. Mol Cell ERK6 (p38 gamma) MAP kinase pathway. Genes Dev 15: Biol 13: 2391–2400. 535–553. Murasawa S, Mori Y, Nozawa Y, Gotoh N, Shibuya M, Marinissen MJ, Chiariello M, Pallante M, Gutkind JS. (1999). Masaki H et al. (1998). Angiotensin II type 1 receptor- A network of mitogen-activated protein kinases links G induced extracellular signal-regulated protein kinase protein-coupled receptors to the c-jun promoter: a role for activation is mediated by Ca2+/calmodulin-dependent trans- c-Jun NH2-terminal kinase, p38s, and extracellular signal- activation of epidermal growth factor receptor. Circ Res 82: regulated kinase 5. Mol Cell Biol 19: 4289–4301. 1338–1348. Marinissen MJ, Servitja JM, Offermanns S, Simon MI, Nagao M, Kaziro Y, Itoh H. (1999). The Srcfamily tyrosine Gutkind JS. (2003). Thrombin protease-activated receptor- kinase is involved in Rho-dependent activation of c-Jun 1 signals through Gq- and G13-initiated MAPK cascades N-terminal kinase by Ga12. Oncogene 18: 4425–4434. regulating c-Jun expression to induce cell transformation. Nagao M, Yamauchi J, Kaziro Y, Itoh H. (1998). Involvement J Biol Chem 278: 46814–46825. of protein kinase C and Srcfamily tyrosine kinase in G aq/11- McDonald PH, Chow CW, Miller WE, Laporte SA, Field induced activation of c-Jun N-terminal kinase and p38 ME, Lin FT et al. (2000). arrestin 2: a receptor-regulated mitogen-activated protein kinase. J Biol Chem 273: 22892– MAPK scaffold for the activation of JNK3. Science 290: 22898. 1574–1577. Needham LK, Rozengurt E. (1998). Ga12 and Ga13 stimulate Michiels F, Stam JC, Hordijk PL, van der Kammen RA, Rho-dependent tyrosine phosphorylation of focal adhesion Ruuls-Van Stalle L, Feltkamp CA et al. (1997). Regulated kinase, paxillin, and p130 Crk-associated substrate. J Biol membrane localization of Tiam1, mediated by the NH2- Chem 273: 14626–14632. terminal pleckstrin homology domain, is required for Rac- Norum JH, Dawood H, Mattingly RR, Sandnes D, Levy FO. dependent membrane ruffling and C-Jun NH2-terminal (2007). Epac- and Rap-independent ERK1/2 phosphoryla- kinase activation. J Cell Biol 137: 387–398. tion induced by Gs-coupled receptor stimulation in HEK293 Mills GB, Moolenaar WH. (2003). The emerging role cells. FEBS Lett 581: 15–20. of lysophosphatidic acid in cancer. Nat Rev Cancer 3: Norum JH, Hart K, Levy FO. (2003). Ras-dependent ERK 582–591. activation by the human Gs-coupled serotonin receptors Minamino T, Yujiri T, Terada N, Taffet GE, Michael LH, 5-HT4(b) and 5-HT7(a). J Biol Chem 278: 3098–3104. Johnson GL et al. (2002). MEKK1 is essential for cardiac Norum JH, Methi T, Mattingly RR, Levy FO. (2005). hypertrophy and dysfunction induced by Gq. Proc Natl Endogenous expression and protein kinase A-dependent Acad Sci USA 99: 3866–3871. phosphorylation of the guanine nucleotide exchange factor Minden A, Karin M. (1997). Regulation and function of the Ras-GRF1 in human embryonickidney 293 cells. FEBS JNK subgroup of MAP kinases. Biochim Biophys Acta 1333: J 272: 2304–2316. F85–F104. Obara Y, Labudda K, Dillon TJ, Stork PJ. (2004). PKA Mischak H, Seitz T, Janosch P, Eulitz M, Steen H, Schellerer phosphorylation of Srcmediates Rap1 activationin M et al. (1996). Negative regulation of Raf-1 by phosphory- NGF and cAMP signaling in PC12 cells. J Cell Sci 117: lation of serine 621. Mol Cell Biol 16: 5409–5418. 6085–6094. Mitchell FM, Russell M, Johnson GL. (1995). Differential Offermanns S, Mancino V, Revel JP, Simon MI. (1997). calcium dependence in the activation of c-Jun kinase and Vascular system defects and impaired cell chemokinesis as a mitogen-activated protein kinase by muscarinic acetylcho- result of Ga13 deficiency. Science 275: 533–536. line receptors in rat 1a cells. Biochem J 309: 381–384. Ohtsu H, Dempsey PJ, Eguchi S. (2006). ADAMs as mediators Mitsui H, Takuwa N, Kurokawa K, Exton JH, Takuwa Y. of EGF receptor transactivation by G protein-coupled (1997). Dependence of activated Ga12-induced G1 to S receptors. Am J Physiol Cell Physiol 291: C1–C10. phase cell cycle progression on both Ras/mitogen-activated Pace AM, Faure M, Bourne HR. (1995). Gi2-mediated protein kinase and Ras/Rac1/Jun N-terminal kinase cas- activation of the MAP kinase cascade. Mol Biol Cell 6: cades in NIH3T3 fibroblasts. J Biol Chem 272: 4904–4910. 1685–1695.

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3140 Pace AM, Wong YH, Bourne HR. (1991). A mutant a subunit cellular signal-regulated kinase pathway by conventional, of Gi2 induces neoplastic transformation of Rat-1 cells. Proc novel, and atypical protein kinase C isotypes. Mol Cell Biol Natl Acad Sci USA 88: 7031–7035. 18: 790–798. Pearson GW, Earnest S, Cobb MH. (2006). Cyclic AMP Sellers LA, Feniuk W, Humphrey PP, Lauder H. (1999). selectively uncouples mitogen-activated protein kinase cas- Activated G protein-coupled receptor induces tyrosine cades from activating signals. Mol Cell Biol 26: 3039–3047. phosphorylation of STAT3 and agonist-selective serine Pizon V, Chardin P, Lerosey I, Olofsson B, Tavitian A. (1988). phosphorylation via sustained stimulation of mitogen- Human cDNAs rap1 and rap2 homologous to the Droso- activated protein kinase. Resultant effects on cell prolifera- phila gene Dras3 encode proteins closely related to ras in the tion. J Biol Chem 274: 16423–16430. ‘effector’ region. Oncogene 3: 201–204. Servitja JM, Marinissen MJ, Sodhi A, Bustelo XR, Gutkind Pouyssegur J, Seuwen K. (1992a). G protein-controlled signal JS. (2003). Rac1 function is required for Src-induced transduction pathways and the regulation of cell prolifera- transformation. Evidence of a role for Tiam1 and Vav2 in tion. Adv Cancer Res 58: 75–94. Rac activation by Src. J Biol Chem 278: 34339–34346. Pouyssegur J, Seuwen K. (1992b). Transmembrane receptors Shan D, Chen L, Wang D, Tan YC, Gu JL, Huang XY. and intracellular pathways that control cell proliferation. (2006). The G protein Ga13 is required for growth factor- Annu Rev Physiol 54: 195–210. induced cell migration. Dev Cell 10: 707–718. Praefcke GJ, McMahon HT. (2004). The dynamin super- Shapiro PS, Evans JN, Davis RJ, Posada JA. (1996). The family: universal membrane tubulation and fission mole- seven-transmembrane-spanning receptors for endothelin cules? Nat Rev Mol Cell Biol 5: 133–147. and thrombin cause proliferation of airway smooth muscle Prasad MV, Dermott JM, Heasley LE, Johnson GL, cells and activation of the extracellular regulated kinase and Dhanasekaran N. (1995). Activation of Jun kinase/stress- c-Jun NH2-terminal kinase groups of mitogen-activated activated protein kinase by GTPase-deficient mutants of protein kinases. J Biol Chem 271: 5750–5754. Ga12 and Ga13. J Biol Chem 270: 18655–18659. Shenoy SK, Lefkowitz RJ. (2003). Multifaceted roles of Prenzel N, Zwick E, Daub H, Leserer M, Abraham R, b-arrestins in the regulation of seven-membrane-spanning Wallasch C et al. (1999). EGF receptor transactivation by receptor trafficking and signalling. Biochem J 375: 503–515. G-protein-coupled receptors requires metalloproteinase Shou C, Wurmser A, Suen KL, Barbacid M, Feig LA, Ling K. cleavage of proHB-EGF. Nature 402: 884–888. (1995). Differential response of the Ras exchange factor, Quilliam LA, Rebhun JF, Castro AF. (2002). A growing Ras-GRF to tyrosine kinase and G protein mediated signals. family of guanine nucleotide exchange factors is responsible Oncogene 10: 1887–1893. for activation of Ras-family GTPases. Prog Nucleic Acid Simon MI, Strathmann MP, Gautam N. (1991). Diversity of G Res Mol Biol 71: 391–444. proteins in signal transduction. Science 252: 802–808. Radhika V, Dhanasekaran N. (2001). Transforming G Sodhi A, Montaner S, Patel V, Zohar M, Bais C, Mesri EA proteins. Oncogene 20: 1607–1614. et al. (2000). The Kaposi’s sarcoma-associated herpes Radhika V, Onesime D, Ha JH, Dhanasekaran N. (2004). virus G protein-coupled receptor up-regulates vascular Ga13 stimulates cell migration through cortactin-interacting endothelial growth factor expression and secretion protein Hax-1. J Biol Chem 279: 49406–49413. through mitogen-activated protein kinase and p38 pathways Ramirez MT, Sah VP, Zhao XL, Hunter JJ, Chien KR, Brown acting on hypoxia-inducible factor 1a. Cancer Res 60: JH. (1997). The MEKK–JNK pathway is stimulated by 4873–4880. alpha1-adrenergic receptor and ras activation and is Soltoff SP. (1998). Related adhesion focal tyrosine kinase and associated with in vitro and in vivo cardiac hypertrophy. the epidermal growth factor receptor mediate the stimula- J Biol Chem 272: 14057–14061. tion of mitogen-activated protein kinase by the G-protein- 2+ Roy S, Wyse B, Hancock JF. (2002). H-Ras signaling and coupled P2Y2 receptor. Phorbol ester or [Ca ]i elevation K-Ras signaling are differentially dependent on endocytosis. can substitute for receptor activation. J Biol Chem 273: Mol Cell Biol 22: 5128–5140. 23110–23117. Rozengurt E. (1998). Signal transduction pathways in the Stephens L, Smrcka A, Cooke FT, Jackson TR, Sternweis PC, mitogenicresponse to G protein-coupledneuropeptide Hawkins PT. (1994). A novel phosphoinositide 3 kinase receptor agonists. J Cell Physiol 177: 507–517. activity in myeloid-derived cells is activated by G protein bg Schafer B, Gschwind A, Ullrich A. (2004). Multiple G-protein- subunits. Cell 77: 83–93. coupled receptor signals converge on the epidermal Stork PJ, Schmitt JM. (2002). Crosstalk between cAMP and growth factor receptor to promote migration and invasion. MAP kinase signaling in the regulation of cell proliferation. Oncogene 23: 991–999. Trends Cell Biol 12: 258–266. Schmitt JM, Stork PJ. (2000). b2-adrenergic receptor activates Sun W, Kesavan K, Schaefer BC, Garrington TP, Ware M, extracellular signal-regulated kinases (ERKs) via the small Johnson NL et al. (2001). MEKK2 associates with the G protein rap1 and the serine/threonine kinase B-Raf. J Biol adapter protein Lad/RIBP and regulates the MEK5– Chem 275: 25342–25350. BMK1/ERK5 pathway. J Biol Chem 276: 5093–5100. Schmitt JM, Stork PJ. (2001). Cyclic AMP-mediated inhibi- Suzuki N, Nakamura S, Mano H, Kozasa T. (2003). Ga12 tion of cell growth requires the small G protein Rap1. Mol activates Rho GTPase through tyrosine-phosphorylated Cell Biol 21: 3671–3683. leukemia-associated RhoGEF. Proc Natl Acad Sci USA Schmitt JM, Stork PJ. (2002a). Ga and Gbg require distinct 100: 733–738. Src-dependent pathways to activate Rap1 and Ras. J Biol Takahashi M, Gotoh Y, Isagawa T, Nishimura T, Goyama E, Chem 277: 43024–43032. Kim HS et al. (2003). Regulation of a mitogen-activated Schmitt JM, Stork PJ. (2002b). PKA phosphorylation of Src protein kinase kinase kinase, MLTK by PKN. J Biochem mediates cAMP’s inhibition of cell growth via Rap1. Mol 133: 181–187. Cell 9: 85–94. Takeuchi Y, Fukunaga K. (2003). Differential regulation of Schonwasser DC, Marais RM, Marshall CJ, Parker PJ. (1998). NF-kB, SRE and CRE by dopamine D1 and D2 receptors in Activation of the mitogen-activated protein kinase/extra- transfected NG108–15 cells. J Neurochem 85: 729–739.

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3141

Tanaka S, Hanafusa H. (1998). Guanine-nucleotide exchange Wan Y, Huang XY. (1998). Analysis of the Gs/mitogen- protein C3G activates JNK1 by a ras-independent mechan- activated protein kinase pathway in mutant S49 cells. J Biol ism. JNK1 activation inhibited by kinase negative forms of Chem 273: 14533–14537. MLK3 and DLK mixed lineage kinases. J Biol Chem 273: Wang HY, Kanungo J, Malbon CC. (2002). Expression of 1281–1284. Ga13 (Q226L) induces P19 stem cells to primitive endoderm Tanaka S, Morishita T, Hashimoto Y, Hattori S, Nakamura via MEKK1, 2, or 4. J Biol Chem 277: 3530–3536. S, Shibuya M et al. (1994). C3G, a guanine nucleotide- Wang X, McGowan CH, Zhao M, He L, Downey JS, Fearns releasing protein expressed ubiquitously, binds to the Src C et al. (2000). Involvement of the MKK6-p38g cascade homology 3 domains of CRK and GRB2/ASH proteins. in g-radiation-induced cell cycle arrest. Mol Cell Biol 20: Proc Natl Acad Sci USA 91: 3443–3447. 4543–4552. Tang WJ, Gilman AG. (1992). Adenylyl cyclases. Cell 70: Wang Y, Su B, Xia Z. (2006a). Brain-derived neurotrophic 869–872. factor activates ERK5 in cortical neurons via a Rap1- Teramoto H, Coso OA, Miyata H, Igishi T, Miki T, Gutkind MEKK2 signaling cascade. J Biol Chem 281: 35965–35974. JS. (1996). Signaling from the small GTP-binding proteins Wang Z, Dillon TJ, Pokala V, Mishra S, Labudda K, Hunter Rac1 and Cdc42 to the c-Jun N-terminal kinase/stress- B et al. (2006b). Rap1-mediated activation of extracellular activated protein kinase pathway. A role for mixed signal-regulated kinases by cyclic AMP is dependent on the lineage kinase 3/protein-tyrosine kinase 1, a novel mode of Rap1 activation. Mol Cell Biol 26: 2130–2145. member of the mixed lineage kinase family. J Biol Chem Waters CM, Connell MC, Pyne S, Pyne NJ. (2005). c-Src is 271: 27225–27228. involved in regulating signal transmission from PDGFbeta Thomason PA, James SR, Casey PJ, Downes CP. (1994). A G- receptor-GPCR(s) complexes in mammalian cells. Cell protein bg-subunit-responsive phosphoinositide 3-kinase Signal 17: 263–277. activity in human platelet cytosol. J Biol Chem 269: Weissman JT, Ma JN, Essex A, Gao Y, Burstein ES. (2002). 16525–16528. G-protein-coupled receptor-mediated activation of rap Todisco A, Takeuchi Y, Yamada J, Sadoshima JI, Yamada T. GTPases: characterization of a novel Gai regulated path- (1997). Molecular mechanisms for somatostatin inhibition way. Oncogene 23: 241–249. of c-fos gene expression. Am J Physiol 272: G721–G726. Werry TD, Sexton PM, Christopoulos A. (2005). Ins and outs Tohgo A, Pierce KL, Choy EW, Lefkowitz RJ, Luttrell LM. of seven-transmembrane receptor signalling to ERK. Trends (2002). Arrestin scaffolding of the ERK cascade enhances Endocrinol Metab 16: 26–33. cytosolic ERK activity but inhibits ERK-mediated tran- Wong YH, Federman A, Pace AM, Zachary I, Evans T, scription following angiotensin AT1a receptor stimulation. Pouyssegur J et al. (1991). Mutant a subunits of Gi2 inhibit J Biol Chem 277: 9429–9436. cyclic AMP accumulation. Nature 351: 63–65. Tokiwa G, Dikic I, Lev S, Schlessinger J. (1996). Activation of Wu J, Dent P, Jelinek T, Wolfman A, Weber MJ, Sturgill TW. Pyk2 by stress signals and coupling with JNK signaling (1993). Inhibition of the EGF-activated MAP kinase pathway. Science 273: 792–794. signaling pathway by adenosine 30,50-monophosphate. Tsygankova OM, Kupperman E, Wen W, Meinkoth JL. Science 262: 1065–1069. (2000). Cyclic AMP activates Ras. Oncogene 19: 3609–3615. Wunderlich L, Farago A, Buday L. (1999). Characterization of Ueda Y, Hirai S, Osada S, Suzuki A, Mizuno K, Ohno S. interactions of Nck with Sos and dynamin. Cell Signal 11: (1996). Protein kinase C activates the MEK–ERK pathway 25–29. in a manner independent of Ras and dependent on Raf. Wylie PG, Challiss RA, Blank JL. (1999). Regulation of J Biol Chem 271: 23512–23519. extracellular-signal regulated kinase and c-Jun N-terminal van Biesen T, Hawes BE, Luttrell DK, Krueger KM, Touhara kinase by G-protein-linked muscarinic acetylcholine recep- K, Porfiri E et al. (1995). Receptor-tyrosine-kinase- and tors. Biochem J 338: 619–628. Gbg-mediated MAP kinase activation by a common Xu N, Bradley L, Ambdukar I, Gutkind JS. (1993). A mutant signalling pathway. Nature 376: 781–784. alpha subunit of G12 potentiates the eicosanoid pathway Vara Prasad MV, Shore SK, Dhanasekaran N. (1994). and is highly oncogenic in NIH 3T3 cells. Proc Natl Acad Activated mutant of Ga13 induces Egr-1, c-fos, and Sci USA 90: 6741–6745. transformation in NIH 3T3 cells. Oncogene 9: 2425–2429. Xu N, Voyno-Yasenetskaya T, Gutkind JS. (1994). Potent Vomastek T, Schaeffer HJ, Tarcsafalvi A, Smolkin ME, transforming activity of the G13 a subunit defines a novel Bissonette EA, Weber MJ. (2004). Modular construction of family of oncogenes. Biochem Biophys Res Commun 201: a signaling scaffold: MORG1 interacts with components of 603–609. the ERK cascade and links ERK signaling to specific Yamaguchi Y, Katoh H, Mori K, Negishi M. (2002). Ga12 and agonists. Proc Natl Acad Sci USA 101: 6981–6986. Ga13 interact with Ser/Thr type 5 and von Kriegsheim A, Pitt A, Grindlay GJ, Kolch W, Dhillon AS. stimulate its phosphatase activity. Curr Biol 12: 1353–1358. (2006). Regulation of the Raf–MEK–ERK pathway by Yamauchi J, Itoh H, Shinoura H, Miyamoto Y, Hirasawa A, protein phosphatase 5. Nat Cell Biol 8: 1011–1016. Kaziro Y et al. (2001). Involvement of c-Jun N-terminal Vossler MR, Yao H, York RD, Pan MG, Rim CS, Stork PJ. kinase and p38 mitogen-activated protein kinase in a1B- (1997). cAMP activates MAP kinase and Elk-1 through a adrenergicreceptor/G aq-induced inhibition of cell proli- B-Raf- and Rap1-dependent pathway. Cell 89: 73–82. feration. Biochem Biophys Res Commun 281: 1019–1023. Voyno-Yasenetskaya TA, Faure MP, Ahn NG, Bourne HR. Yamauchi J, Kawano T, Nagao M, Kaziro Y, Itoh H. (2000). (1996). Ga12 and Ga13 regulate extracellular signal-regulated Gi-dependent activation of c-Jun N-terminal kinase in kinase and c-Jun kinase pathways by different mechanisms human embryonal kidney 293 cells. J Biol Chem 275: in COS-7 cells. J Biol Chem 271: 21081–21087. 7633–7640. Voyno-Yasenetskaya TA, Pace AM, Bourne HR. (1994). Yamauchi J, Kaziro Y, Itoh H. (1999). Differential regulation Mutant a subunits of G12 and G13 proteins induce of mitogen-activated protein kinase kinase 4 (MKK4) and 7 neoplastictransformation of Rat-1 fibroblasts. Oncogene 9: (MKK7) by signaling from G protein bg subunit in human 2559–2565. embryonal kidney 293 cells. J Biol Chem 274: 1957–1965.

Oncogene G Proteins and MAPKs ZG Goldsmith and DN Dhanasekaran 3142

Yamauchi J, Nagao M, Kaziro Y, Itoh H. (1997). Activation receptor involves GaqGbg and epidermal growth factor of p38 mitogen-activated protein kinase by signaling receptor tyrosine kinase activation. Endocrinology 144: through G protein-coupled receptors. Involvement of Gbg 2947–2956. and Gaq/11 subunits. J Biol Chem 272: 27771–27777. Zhu D, Kosik KS, Meigs TE, Yanamadala V, Denker BM. Yang D, Tournier C, Wysk M, Lu HT, Xu J, Davis RJ et al. (2004). Ga12 directly interacts with PP2A: evidence FOR (1997). Targeted disruption of the MKK4 gene causes Ga12-stimulated PP2A phosphatase activity and dephos- embryonicdeath, inhibition of c-JunNH2-terminal kinase phorylation of microtubule-associated protein, tau. J Biol activation, and defects in AP-1 transcriptional activity. Proc Chem 279: 54983–54986. Natl Acad Sci USA 94: 3004–3009. Zohn IE, Yu H, Li X, Cox AD, Earp HS. (1995). Angiotensin Zheng M, Zhang SJ, Zhu WZ, Ziman B, Kobilka BK, Xiao II stimulates calcium-dependent activation of c-Jun RP. (2000). 2-adrenergic receptor-induced p38 MAPK N-terminal kinase. Mol Cell Biol 15: 6160–6168. activation is mediated by protein kinase A rather than by Zwick E, Daub H, Aoki N, Yamaguchi-Aoki Y, Tinhofer I, Gi or Gbg in adult mouse cardiomyocytes. J Biol Chem 275: Maly K et al. (1997). Critical role of calcium- dependent 40635–40640. epidermal growth factor receptor transactivation in PC12 Zhong M, Yang M, Sanborn BM. (2003). Extracellular signal- cell membrane depolarization and bradykinin signaling. regulated kinase 1/2 activation by myometrial oxytocin J Biol Chem 272: 24767–24770.

Oncogene