432
Views
1
CrossRef citations to date
0
Altmetric
Review

GPCRs that Rhoar the Guanine nucleotide exchange factors

ORCID Icon & ORCID Icon
Pages 84-99 | Received 01 Jan 2021, Accepted 24 Feb 2021, Published online: 14 Apr 2021

ABSTRACT

Cell migration, a crucial step in numerous biological processes, is tightly regulated in space and time. Cells employ Rho GTPases, primarily Rho, Rac, and Cdc42, to regulate their motility. Like other small G proteins, Rho GTPases function as biomolecular switches in regulating cell migration by operating between GDP bound ‘OFF’ and GTP bound ‘ON’ states. Guanine nucleotide exchange factors (GEFs) catalyse the shuttling of GTPases from OFF to ON state. G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors that are involved in many signalling phenomena including cell survival and cell migration events. In this review, we summarize signalling mechanisms, involving GPCRs, leading to the activation of RhoGEFs. GPCRs exhibit diverse GEF activation modes that include the interaction of heterotrimeric G protein subunits with different domains of GEFs, phosphorylation, protein–protein interaction, protein–lipid interaction, and/or a combination of these processes.

Introduction

Cell migration is a fundamental event in various biological processes such as embryogenesis, immune surveillance, homoeostasis, wound repair, and tissue formation[Citation1]. Cells can migrate as individuals or as a collective group of cells. Collective cell migration contributes to angiogenesis, lumen formation, and tumour cell invasion[Citation2]. On the other hand, individual cell migration is exhibited by cells like fibroblasts (during wound healing) and leukocytes (during immune response)[Citation1]. To initiate migration, cells receive a variety of migratory and invasive cues from their surroundings, which could be in the form of chemicals such as chemokines[Citation3], growth factors (EGF, IGF-1) [Citation4] or sometimes physical signals such as temperature and pressure [Citation5,Citation6]. The migration cycle of an individual cell includes polarization of the front and rear end of the cell, protrusive structure formation at the front end, adherence of the protrusions with the extracellular matrix (ECM), and retraction of the rear end due to contractile force generated by the cytoskeleton[Citation7]. As compared to individual cell migration, collective cell migration is much more complex in terms of its execution and signalling[Citation2]. Similar to individual cell migration, cells moving in a collective fashion also establish front and rear polarity. However, in the collective cell migration, cells at the front end polarize and produce protrusions, whereas, the ones present at the lateral and rear end maintain cell-cell adhesion to generate the required traction force[Citation2]. Thus, the initial steps in all types of cell migration include polarization and formation of protrusions like lamellipodia and filopodia, which require coordinated cytoskeletal dynamics. As cell migration is a vital biological step, it is tightly regulated in space and time by employing a complex signalling network involving the Rho family of small G proteins[Citation8]. These G proteins control cytoskeletal dynamics by regulating actin polymerization. Amongst the known 22 Rho GTPases family members, RhoA, Rac and Cdc42 are the members, most extensively studied for their role in cell migration[Citation8]. RhoA is mostly involved in stress fibre formation and cell contraction [Citation9,Citation10] whereas, Rac controls the formation of lamellipodia, cell spreading, and membrane ruffling [Citation10–13]. Similarly, Cdc42 has been shown to have important roles in filopodia formation and cell reshaping [Citation11,Citation13]. Thus, this Rho GTPase trio lies at the central stage of signal transduction events that control cell migration. Like all canonical G proteins, Rho GTPases exert signalling events by being biomolecular switches. They are active (ON) when bound to GTP and inactive (OFF) when bound to GDP. Two different classes of proteins, Guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs) help to shuttle the GTPases between ON and OFF states. GEFs activate GTPases by exchanging their bound GDP with GTP, whereas, GAPs bring them to OFF state by promoting their intrinsic GTP hydrolysis [Citation14] (). For Rho GTPases, there exist another level of regulation in terms of Guanine nucleotide dissociation factors (GDIs), which sequester GDP bound GTPases in the cytosol and replenish them back when required by the cell [Citation15] (). Many reviews excellently summarize the regulation of Rho GTPases [Citation8,Citation14,Citation16–18] by GEFs, GAPs, and GDIs. However, how exactly the migratory cues propagate from various sources to the respective GEFs, GAPs, and GDIs is not well understood. In many instances, the migratory information propagates from the plasma membrane which is communicated largely through membrane-bound proteins such as G protein-coupled receptors (GPCR), Receptor tyrosine kinase (RTK), integrin, and cytokine receptors [Citation16,Citation19,Citation20]. Amongst these, GPCRs are known to be the largest class of membrane receptors that play a key role in the regulation of GEFs. These seven transmembrane receptors are associated with heterotrimeric G proteins, comprising Gαβγ subunits and upon receiving the appropriate extracellular cues, they exert the signalling process by releasing the Gα and Gβγ subunits into the cytosol which in turn regulate their downstream effectors[Citation21]. Aberration in these signalling events is implicated in various pathological conditions such as metabolic disorders, immunological disorders, neurodegenerative diseases, infectious diseases, and cancer [Citation22–24]. A broad variety of signalling cascades initiated by GPCRs during cell migration is discussed earlier by Cotton et al. [Citation25]. However, in this review, we will try to summarize how migratory signals from GPCRs specifically activate RhoGEFs, in the context of cell migration. The mode of propagation of these signals includes, binding of GPCR activated heterotrimeric G protein subunits with different domains of GEFs, phosphorylation of GEFs by various kinases downstream of GPCRs, protein–protein interactions, and protein–lipid interactions.

Figure 1. Schematic representation of some of the GEFs discussed in this review showing intra-domain interaction rendering them in autoinhibitory states. (a) GTPase switching mechanism between ON and OFF state. Canonical GEFs such as (b) p115-RhoGEF and (c) PDZ-RhoGEF are proposed to exhibit autoinhibition due to intra-domain interaction involving their RH domain. Similarly, autoinhibition of (d) AKAP-Lbc and (e) p114-RhoGEF is due to intra-domain interaction involving its non-RH, C terminal region. (f) In p63RhoGEF and (g) Vav GEF, the PH domain is suggested to be responsible for its autoinhibition. (h) In P-Rex GEF, the C terminal domains like IPI4, PDZ1, and PDZ2 are responsible for intra-domain interactions. (i) In TIAM1, the initial 50 amino acid domain (N50), N terminal PH domain (PHn) and the coiled coil extension (CC-Ex) domain of TIAM1 interact with each other to render the GEF to its autoinhibitory state. (j) In non-canonical GEFs like DOCK proteins interaction between N terminal SH3 domain and C terminal domains and/or DHR2 domain is responsible for their autoinhibition.

Figure 1. Schematic representation of some of the GEFs discussed in this review showing intra-domain interaction rendering them in autoinhibitory states. (a) GTPase switching mechanism between ON and OFF state. Canonical GEFs such as (b) p115-RhoGEF and (c) PDZ-RhoGEF are proposed to exhibit autoinhibition due to intra-domain interaction involving their RH domain. Similarly, autoinhibition of (d) AKAP-Lbc and (e) p114-RhoGEF is due to intra-domain interaction involving its non-RH, C terminal region. (f) In p63RhoGEF and (g) Vav GEF, the PH domain is suggested to be responsible for its autoinhibition. (h) In P-Rex GEF, the C terminal domains like IPI4, PDZ1, and PDZ2 are responsible for intra-domain interactions. (i) In TIAM1, the initial 50 amino acid domain (N50), N terminal PH domain (PHn) and the coiled coil extension (CC-Ex) domain of TIAM1 interact with each other to render the GEF to its autoinhibitory state. (j) In non-canonical GEFs like DOCK proteins interaction between N terminal SH3 domain and C terminal domains and/or DHR2 domain is responsible for their autoinhibition.

GPCR mediated heterotrimeric G protein subunit interaction with RH domains of GEFs

Cell polarization, protrusions, and focal adhesions at the site of leading edge formation in both, the individual cell migration as well as collective cell migration (in leader cells) are regulated by RhoA, Rac as well as Cdc42 GTPases [Citation2,Citation7]. Membrane proteins such as chemokine, cytokine, GPCRs, and adhesion receptors activate these GTPases through their GEFs[Citation19]. RhoGEFs can be classified into two categories as canonical and non-canonical (Docker GEFs). Almost all canonical GEFs comprise catalytic Dbl homology domain (DH) and at least one pleckstrin homology (PH) domain[Citation26]. PH domain is primarily involved in the regulation of catalytic activity and also promotes membrane localization of GEF by interacting with membrane lipids like Phosphotidylinositides (PIPs) [Citation27–29]. In a few GEFs, other domains like Ras binding domain (RBD), additional PH domains, regulatory G protein signalling homology (RH) domain, and PDZ domain, are also known to regulate their catalytic activity [Citation30–32]. Most of the canonical GEFs are known to exist in an autoinhibited state due to the interaction between domains at their N and C terminus (), making the catalytical domain inaccessible for the GTPase binding. Unlike canonical GEFs, in non-canonical GEFs the PH and DH domains are replaced by DHR-1 and DHR-2 domains (also known as Docker domains), which perform functions similar to their canonical counterparts. Some of the Dedicator of cytokinesis (DOCK) family members are also known to possess additional domains like SH3 or PH domains [Citation33–35]. Similar to the canonical GEFs, DOCK family GEFs are known to exist in an autoinhibitory state[Citation36]. For example, N terminal SH3 domain of DOCK180/DOCK1 is known to interact with its DHR2 domain, to render it inactive[Citation37].

In both types of GEFs, activation happens upon release of their autoinhibitory state either through protein–protein interactions or structural changes on account of post-translation modifications like phosphorylation, which disrupt the intra-domain interactions in them [Citation29,Citation38,Citation39]. However, for many GEFs, the activation mechanism is still not clear. In few instances, GPCRs are known to activate GEFs either by promoting interaction between the heterotrimeric G protein subunits with them or indirectly through pathways regulated by these subunits [Citation40–42]. In some examples, GEFs are known to be activated due to their direct interactions with different domains of GPCRs[Citation43]. In this section, we will discuss ligand-mediated signalling through GPCRs that activate canonical GEFs like leukaemia-associated Rho GEF (LARG), p115-RhoGEF, and PDZ-RhoGEF through heterotrimeric G protein subunit interaction with the regulator of G protein signalling homology (RH) domain of these GEFs.

Leukaemia-associated Rho GEF (LARG) is a canonical GEF, shown to be expressed in most of the cells including peripheral blood leukocytes, spleen, prostate, testis, ovary, small intestine, colon with a minimal expression in thymus [Citation44] and specifically, activates RhoA (). In mouse embryonic fibroblasts (NIH-3T3), GPCRs, M1 muscarinic cholinergic receptor (M1-mAChR) and Mas related receptor activate RhoA specific GEF LARG, through direct binding of Gαq subunit (). Similarly, G protein-coupled receptor 132 (G2AR), when triggered with its lipid ligand, activates LARG through Gα13[Citation40]. From co-immunoprecipitation studies, it has been shown that the RH domain of LARG interacts with Gαq and Gα13 subunits of GPCRs complex to quell its autoinhibitory state[Citation40]. Similarly, in mouse smooth muscle cells, activation of Sphingosine 1 phosphate receptor 2 (S1P2R) is shown to promote their motility, which is induced by the interaction between LARG and Gα12/13 subunit [Citation45] (). Analogous LARG activation mechanism is seen in human prostate cancer cells (PC-3) and Human embryo kidney (HEK293T) cells where the Thrombin receptor (PAR) operates through PAR/Gα13/LARG pathway. In mouse embryonic fibroblasts LARG is activated through Lysophosphatidic acid receptor (LPAR)/Gα12/13/LARG/RhoA pathway involving GPCR, LPAR [Citation46,Citation47] (). LARG is also found to localize with Microtubule organizing centres (MTOC), indicating that the extracellular signals through GPCRs might be involved in maintaining microtubule dynamics and cell polarity through the same Gα12/13/LARG/RhoA pathway[Citation47].

Table 1. Overview of canonical and non-canonical GEFs discussed in this review, their downstream Rho GTPases, tissue-specific expression and activating GPCRs

Figure 2. Schematic representation of signalling pathways which leads to activation of GEFs through interaction between heterotrimeric G protein subunits and RH domains of GEFs. (a) GPCRs, Mas receptor and M1-mAChR activate the GEF, LARG, through Gαq binding to its RH domain. (b) Activation of LARG through GPCRs, LPAR & S1P2R occur through direct interaction with Gα12/13 whereas, GPCRs, G2A & PAR activate LARG through Gα13 subunit. (c) PDZ-RhoGEF mediated RhoA activation is established by GRPR, LPAR, and CXCR4 triggered Gα13 heterotrimeric subunit binding to the RH domain of PDZ-RhoGEF. (d) GPCRs, LPAR, CXCR4, and S1P2R activate p115-RhoGEF through interaction with Gα13 heterotrimeric subunit. (e) Upon activation of β2AR, p115-RhoGEF is triggered by β-arrestin activation through Gαiβγ signalling.

Figure 2. Schematic representation of signalling pathways which leads to activation of GEFs through interaction between heterotrimeric G protein subunits and RH domains of GEFs. (a) GPCRs, Mas receptor and M1-mAChR activate the GEF, LARG, through Gαq binding to its RH domain. (b) Activation of LARG through GPCRs, LPAR & S1P2R occur through direct interaction with Gα12/13 whereas, GPCRs, G2A & PAR activate LARG through Gα13 subunit. (c) PDZ-RhoGEF mediated RhoA activation is established by GRPR, LPAR, and CXCR4 triggered Gα13 heterotrimeric subunit binding to the RH domain of PDZ-RhoGEF. (d) GPCRs, LPAR, CXCR4, and S1P2R activate p115-RhoGEF through interaction with Gα13 heterotrimeric subunit. (e) Upon activation of β2AR, p115-RhoGEF is triggered by β-arrestin activation through Gαiβγ signalling.

PDZ-RhoGEF and p115-RhoGEF provide examples for Gα13 subunit induced activation of GEF, where the heterotrimeric G subunit interacts with their RH domain to release their autoinhibitory state [Citation38,Citation48]. However, GPCRs that provide the heterotrimeric G subunit (Gα13) for GEF activation differ from cell to cell. C-X-C chemokine receptor type 4 (CXCR4), LPAR and Gastrin-releasing peptide receptor (GRPR) activate RhoA in breast cancer cell lines (MCF7, MDA-MB-231), PC-3 cells and colon cancer cells (Caco-2), respectively, through Gα13/PDZ-RhoGEF/RhoA pathway [Citation38,Citation46,Citation49] (). As seen in PDZ-RhoGEF, p115-RhoGEF also activates RhoA signalling in lymphocytes through Gα13/p115-RhoGEF/RhoA upon CXCR4, LPAR, and S1PR activation [Citation50] (). These are the examples where G protein subunits, Gα12/13, bind directly to the RH domain containing GEFs to activate them. An exception is seen for p115-RhoGEF in renal cell carcinoma 7 (RCC7) where activated GPCR, β2-adrenergic receptor (β2AR), triggers the GEF indirectly through βArrestin2Citation55 (). Furthermore, there are examples where PAR and LPA receptors in PC-3 cells are known to specifically activate LARG and PDZ-RhoGEF, respectively. However, the mechanism underlining their specificity is yet to be determined[Citation46]. Recently it has been shown that prostaglandin receptor activated Gαs binds to the PH and DH domains as well as their linker region of PDZ-RhoGEF. This interaction of Gαs with PDZ-RhoGEF renders the GEF active towards Cdc42 [Citation51,Citation52] ().

Figure 3. Schematic representation of signalling pathways which leads to activation of GEFs through interaction between heterotrimeric G protein subunits and non-RH domains of GEFs. (a) Stimulation of prostaglandin receptor activates Gαs, which in turn binds to PDZ-RhoGEF through DH, PH domains, and their linker region of GEF, thus making it active towards Cdc42. (b) AKAP-Lbc dependent activation of RhoA, by LPAR and AT1R, happens through the interaction of Gα12 with the C terminus of AKAP-Lbc. (c) and (d) Activation of Gβγ (through M3R and LPAR) and Gα12 triggers p114-RhoGEF, where Gβγ interacts with the C terminus of DH-PH domain of p114-RhoGEF. (e) p63RhoGEF mediated triggering of RhoA upon activation of M3R and H1R receptors. This involves the interaction of Gαq/11 towards the PH domain of p63RhoGEF. (f) Stimulated GPCRs, LPAR, and CXCR4 activate P-Rex1 through the interaction of the Gβγ subunit with the IP4P domain and the tandem PDZ domains of P-Rex1. This may further lead to the activation of ELMO/DOCK2 through P-Rex activated RhoG. (g) Activation of ET1R triggers βPIX mediated Cdc42 through the interaction of Gαq with βPIX.

Figure 3. Schematic representation of signalling pathways which leads to activation of GEFs through interaction between heterotrimeric G protein subunits and non-RH domains of GEFs. (a) Stimulation of prostaglandin receptor activates Gαs, which in turn binds to PDZ-RhoGEF through DH, PH domains, and their linker region of GEF, thus making it active towards Cdc42. (b) AKAP-Lbc dependent activation of RhoA, by LPAR and AT1R, happens through the interaction of Gα12 with the C terminus of AKAP-Lbc. (c) and (d) Activation of Gβγ (through M3R and LPAR) and Gα12 triggers p114-RhoGEF, where Gβγ interacts with the C terminus of DH-PH domain of p114-RhoGEF. (e) p63RhoGEF mediated triggering of RhoA upon activation of M3R and H1R receptors. This involves the interaction of Gαq/11 towards the PH domain of p63RhoGEF. (f) Stimulated GPCRs, LPAR, and CXCR4 activate P-Rex1 through the interaction of the Gβγ subunit with the IP4P domain and the tandem PDZ domains of P-Rex1. This may further lead to the activation of ELMO/DOCK2 through P-Rex activated RhoG. (g) Activation of ET1R triggers βPIX mediated Cdc42 through the interaction of Gαq with βPIX.

Activation of GEFs involving interaction between heterotrimeric G protein subunits and non-RH domains of GEFs

G proteins of certain GPCRs are also known to activate GEFs in RH domain independent manner. Examples of these GEFs include A-kinase anchoring protein (AKAP)-Lbc, p114-RhoGEF, p63RhoGEF, Trio, and P-Rex. Details of activation mechanisms of these GEFs by GPCRs will be discussed further. Tissue-specific expression of aforementioned GEFs is given in . Activation of LPA receptor and Angiotensin 1 receptor (AT1R) promotes RhoA mediated signalling in HEK293 cells and adult ventricular fibroblasts (AVFs) through Gα12/AKAP-Lbc/RhoA pathway [Citation72,Citation73] (). Here, the interaction of AKAP-Lbc with Gα12 is independent of the RH domain, as this protein lacks this domain[Citation74]. However, the binding of Gα12 to the GEF depends on a 106 amino acid residue region, stretching from residues 2567–2672 towards the C terminus of AKAP-Lbc[Citation41]. Studies in breast cancer cell lines (MDA-MB-435) show that, through a feedback loop signalling, α6β4 integrin regulates LPA induced AKAP-Lbc/Gα12/RhoA pathway to promote activation of Rac GTPase[Citation75]. AKAP-Lbc is predominantly localized in the cytoplasm where it is known to regulate spatial distribution of Protein kinase A (PKA) activity during cell migration [Citation72,Citation76].

The next example involving non-RH domain GEF interacting with heterotrimeric G protein includes p114-RhoGEF, activated by GPCRs, M3-muscarinic receptor (M3R), and LPA receptor operating in HEK293 and NIH3T3 cells, respectively. In these pathways, the Gβγ subunit of G protein interacts with the DH-PH domain of p114-RhoGEF to bring about its activation, eventually leading to RhoA and Rac signalling [Citation77,Citation78] (). Apart from Gβγ, the Gα12 subunit is also found to interact with p114-RhoGEF (), however, it binds to a region spanning residues from 686 to 791, towards the C terminus of the DH-PH domain [Citation41,Citation77,Citation79]. Furthermore, some evidence suggest that p114-RhoGEF gets triggered both in GPCR dependent and independent manner. For example, it has been shown that tissue-specific apical cell constriction in invertebrates is brought about by non-conserved GPCRs such as Mist (mesoderm-invagination signal transducer) and Smog through the activation of p114-RhoGEF[Citation80]. However, in vertebrates the apical cell constriction process, involving the same p114-RhoGEF, takes place in a GPCR independent manner. In this instance, another GEF DAPLE, which possess Gα-binding-and-activating (GBA) motif could directly activate Gαiβγ resulting in stimulation of p114-RhoGEF through the Gβγ subunit[Citation81].

Another non-RH domain containing GEF, p63RhoGEF, preferentially expressed in heart and brain tissues, is known to interact with the Gαq/11 subunit. Here the agonist-stimulated GPCRs like M3-cholinoceptor (M3R) and the histamine H1 receptor (H1R), triggers the GEF to activate Gαq/11/p63RhoGEF/RhoA pathway [Citation82,Citation83] (). Crystal structure of Gαi/q -p63RhoGEF-RhoA shows an amphipathic helical extension of PH domain (residues 471–485) of the GEF interacting with Gαi/q [Citation83,Citation84]. Thus, it can be proposed that the autoinhibitory state of the GEF due to the inter-domain interaction invloving PH domain [Citation85,Citation86] is released upon its engagement with Gαi/q. Furthermore, in fibroblast and COS-7 cells, it is shown that M3R activated p63RhoGEF competes with phospholipase C (PLCβ) for binding Gαq[Citation83]. Similar to p63RhoGEF, the Triple function domain protein (Trio) also contains an inhibitory C terminal extension beyond its PH domain[Citation85]. Trio exhibits dual Rac/RhoA GEF activity[Citation87]. Gαq subunit renders Trio active towards RhoA by binding the inhibitory PH domains and hence relieving its autoinhibition[Citation85]. However, the GPCR which is involved in this pathway has not yet been identified.

In MCF-7 breast cancer cells, chemotactic GPCRs, CXCR4 and LPAR, activate Rac through the Gβγ/PIP3-dependent Rac exchanger1 (P-Rex1)/Rac pathway (). Here the Gαq or Gα13 but not Gαs or Gαi form stable complex with Gβγ. Therefore, the activation of P-Rex1 could be through CXCR4 and LPAR, preferably via Gαiβγ dependent fashion [Citation88–90]. From cryo-electron microscopy structure it has been suggested that Gβγ interaction with P-Rex1 is a multidomain assembly involving IP4P and tandem PDZ domains of the GEF[Citation91]. Operating under the same pathway, P-Rex1 is also known to activate RhoG leading to the RhoG/ELMO/DOCK2 dependent activation of Rac [Citation92] ().

Activation of GEFs through Phosphorylation, protein–lipid, and protein–protein interactions mediated by GPCRs

Phosphorylation is another means through which GPCRs activate GEFs, wherein these receptors activate downstream kinases that in turn trigger GEFs. One such example is p21-activated kinases interacting exchange proteins (PIX) which, exist in two isoforms αPIX and βPIX and are known to activate Cdc42 and Rac1 GTPases[Citation13]. αPIX is predominantly expressed in haematopoietic and muscle cells whereas, βPIX is expressed ubiquitously. Splice variants of these proteins are majorly found in the brain [Citation13,Citation93]. It has been shown that activation of βPIX involves Gαi of LPA receptor in HeyA8 ovarian cancer cell lines, where Gαi recruits Src kinase, which phosphorylates Tyr-442 residue of βPIX to activate the GEF [Citation94–96] (). Apart from Src, other kinases like p21-activated kinases (PAK) are also known to phosphorylate numerous serine and tyrosine residues of βPIX, especially the residues at its PH, GITI-binding, N-terminal spacer region (amino acid 61–99), and C terminal coiled-coil domains. Other than GEF activation, phosphorylation at many of these sites could have important roles in regulating βPIX interaction with other signal transduction proteins[Citation97]. Another kinase, PKA, is shown to phosphorylate βPIX primarily at Ser-516 and Thr-526, when triggered through Endothelin 1 receptor (ET1R) induced Gαs subunit[Citation98]. Furthermore, βPIX is also known to interact with Endothelin 1 receptor (ET1R) activated Gαq subunits to trigger Cdc42Citation99 (). Though these studies suggest that the direct binding of the Gαq subunit is required for βPIX activation[Citation99]. But, the role of phosphorylation in this case is not clear.

Figure 4. Activation of GEFs through Phosphorylation, protein-protein, and lipid-mediated interactions involving downstream of GPCR signalling. (a) Upon LPAR stimulation, activation of βPIX takes place through, Gαi mediated Src kinase activation, which further phosphorylates βPIX. (b) Activation of fMLPR triggers Cdc42 through Gβγ-PAK1-αPIX and Rac1 through Gβγ-PAK1-αPIX-GIT2 sequential complex formation, where activation of αPIX takes place through PAK1 binding. Cdc42 and Rac1 are involved in feedforward mechanism where αPIX and Rac1 enhance PAK1 activity. Here activation of Cdc42 enhances GEF activity of αPIX towards Rac1. (c) CXCR4 activates Vav1 and Vav2 through JAK signalling. (d) Activation of Rac through fMLPR stimulation involves concerted signalling between Vav1/3 and P-Rex GEFs. (e) LPAR triggers Rac1 through PKC and CamKII mediated phosphorylation of TIAM1. (f) Stimulation of GPCRs, LPAR and S1P1R, activate TIAM through Gαiβγ/PI3K/TIAM1/Rac1 pathway, where TIAM1 interacts with a PI3K lipid product, PIP3. (g) Protein–protein interaction mediated triggering of RhoA when activated Frizzled7 recruits Dishevelled (Dvl) and Daam-1 proteins which activate the Rho WGEF.

Figure 4. Activation of GEFs through Phosphorylation, protein-protein, and lipid-mediated interactions involving downstream of GPCR signalling. (a) Upon LPAR stimulation, activation of βPIX takes place through, Gαi mediated Src kinase activation, which further phosphorylates βPIX. (b) Activation of fMLPR triggers Cdc42 through Gβγ-PAK1-αPIX and Rac1 through Gβγ-PAK1-αPIX-GIT2 sequential complex formation, where activation of αPIX takes place through PAK1 binding. Cdc42 and Rac1 are involved in feedforward mechanism where αPIX and Rac1 enhance PAK1 activity. Here activation of Cdc42 enhances GEF activity of αPIX towards Rac1. (c) CXCR4 activates Vav1 and Vav2 through JAK signalling. (d) Activation of Rac through fMLPR stimulation involves concerted signalling between Vav1/3 and P-Rex GEFs. (e) LPAR triggers Rac1 through PKC and CamKII mediated phosphorylation of TIAM1. (f) Stimulation of GPCRs, LPAR and S1P1R, activate TIAM through Gαiβγ/PI3K/TIAM1/Rac1 pathway, where TIAM1 interacts with a PI3K lipid product, PIP3. (g) Protein–protein interaction mediated triggering of RhoA when activated Frizzled7 recruits Dishevelled (Dvl) and Daam-1 proteins which activate the Rho WGEF.

Although relatively little is known about phosphorylation induced activation of αPIX by PAK, it has been suggested that PAK triggers this GEF by releasing it from its inactive dimer state [Citation13,Citation100]. This pathway involves activation of Gβγ through formyl-methionyl-leucyl-phenylalanine receptor (fMLPR), which further triggers a cascade of signalling events, propagating through p21-activated kinase 1 (PAK1). Further, PAK1 interacts with the SH3 domain of the GEF to release αPIX from its dimeric state to its active monomeric state which further triggers Cdc42 [Citation13,Citation100]. Activated Cdc42 is also known to enhance the affinity of this GEF towards Rac1 (). Thus, it appears that there is a mutual feedforward mechanism between these two GTPases. On one hand, binding of Cdc42 to one of the DH domains of the dimeric αPIX introduces conformational changes in the GEF to enhance its activity towards Rac1Citation100[Citation101], whereas, on the other hand, in a feedforward mechanism, αPIX enhances kinase activity of PAK1 to further activate Cdc42[Citation102]. Furthermore, αPIX is also known to trigger Rac1 in fMLPR controlled pathway. Here, fMLPR triggers to form a quaternary complex Gβγ-PAK1-αPIX-GIT2 in a sequential manner to recast its specificity for Rac1 (). During this pathway, the role of phosphorylation in the activation of αPIX is not clear, but it has been shown that activation of αPIX could take place through binding to phosphatidylinositol 3, 4, 5-trisphosphate (PIP3), a lipid product of PI3-kinase[Citation103]. Similar to Cdc42, Rac1 is also suggested to operate in the feedforward cycle to further enhance the activity of PAK1 towards the GEF[Citation104].

Vav family of GEFs provide another example for activation of GEFs, through phosphorylation relay triggered by GPCRs like CXCR4 and fMLPR [Citation42,Citation105]. This family of GEF consists of 3 members, namely, Vav1 (specific to Rac1/2, RhoG, and Cdc42), Vav2 (specific to Rac1, RhoA, RhoG, and Cdc42), and Vav3 (specific to Rac1, RhoG, and RhoA) [Citation106,Citation107] and their tissue-specific expression is shown in . Out of these GEFs, Vav1 and Vav2 are activated by pathways involving Janus kinase2 (JAK2), Janus kinase3 (JAK3) and the receptor involved in this particular signalling is CXCR4[Citation42]. It is interesting to know that activation of this GEF through JAK is independent of GPCR-Gαi pathway [Citation42,Citation108] (). In P1V1 neutrophils, through an unknown mechanism, fMLPR leads to activation of P-Rex1 and Vav1/Vav3Citation105 GEFs (). However, the role of GPCRs, CXCR4 and fMLPR, in the activation of Vav proteins is not clear. For Vav1 it has been surmised that fMLPR could activate Src or Syk kinases through downstream signalling to trigger this RacGEF [Citation109,Citation110].

The T-cell lymphoma invasion and metastasis inducing factor 1 (TIAM1) provides a good example of phosphorylation mediated activation of GEF. Through Small-angle X-ray scattering (SAXS) based model it was suggested that the initial 50 amino acid domain (N50), N terminal PH domain (PHn), and the coiled-coil extension (CC-Ex) domain of TIAM1 interact with each other to render the GEF inactive[Citation32]. Release of this autoinhibition involves multiple steps including phosphorylation of TIAM1 by protein kinase C (PKCα) and Ca2+/calmodulin-dependent protein kinase II (CamKII) (). In Swiss 3T3 fibroblast cells, this phosphorylation pathway is triggered by the LPA receptor[Citation111]. Based on various experimental studies it has been proposed that sequential phosphorylation of Ser29 and Ser33 of TIAM1 by atypical C kinase (aPKCγ)[Citation32], followed by phosphorylation of Tyr829 by TrkB kinase[Citation112], disrupt the intra-domain interactions of the GEF to make it active [Citation32,Citation113]. Apart from phosphorylation, there exist other modes of TIAM1 activation including protein–protein interactions involving PHn-CC-Ex domain of TIAM1 with Par3 in the Par polarity complex (Par3, Par6 and aPKC) [Citation32,Citation114,Citation115].

It has also been proposed that lipid-mediated interactions too can activate TIAM1. For example GPCRs, LPA1R and S1P1R, induced Gαiβγ/PI3K/TIAM1/Rac1 pathway promote binding of PI3K lipid product, phosphatidylinositol 3, 4, 5-trisphosphate (PIP3)[Citation116], to the PH domain of the GEF to make it active [Citation12,Citation117] (). However, complete activation of TIAM1 requires additional signals from Gαiβγ[Citation12]. Although TIAM2/STEF is relatively less characterized compared to TIAM1, it has been suggested that the activation of this GEF is also a complex multistep process. Forster resonance energy transfer based biosensor studies on dibutyryl cAMP (dbcAMP) treated PC12D cells has revealed that the phosphorylation of Thr-749 by dbcAMP activated PKA at the plasma membrane enhances the GEF activity of TIAM2 towards Rac1[Citation118].

Activation of GEFs through direct interaction of GPCR domains

Few reports suggest that GPCRs could activate GEFs by directly interacting with them, instead of utilizing the heterotrimeric G proteins. For example, PDZ-RhoGEF and LARG interact with the C terminus of LPA1R and LPA2R receptors through their PDZ domains () to get activated[Citation43]. Similarly, activation of Vav2 involves interaction of the C terminus of the Parathyroid hormone receptor (PTHR) with its DH and PH domains. It is interesting to note that Vav2 competes with PTHR to bind Gαq subunit and also interferes with the PTHR signalling[Citation119] (). Other than these examples, there is not much information on how GPCRs could directly activate GEFs.

Figure 5. Schematic representation of activation of GEFs upon direct interaction with GPCR domains. (a) GEFs, LARG, and PDZ-RhoGEF activate RhoA signalling upon interacting with the C terminus of LPAR through their PDZ domains. (b) Similarly, Vav2 interacts with the C terminus of PTHR through its DH and PH domains.

Figure 5. Schematic representation of activation of GEFs upon direct interaction with GPCR domains. (a) GEFs, LARG, and PDZ-RhoGEF activate RhoA signalling upon interacting with the C terminus of LPAR through their PDZ domains. (b) Similarly, Vav2 interacts with the C terminus of PTHR through its DH and PH domains.

Non-canonical GPCRs activate GEFs through the scaffold protein Dishevelled (Dvl)

There are about 11 non-canonical GPCRs identified as Frizzled (FZD) class receptors. Other than smoothened (SMO), all other members of this family are activated by secreted glycoproteins, Wingless/Int1 (Wnt). There are 19 Wnts identified in humans. A combination of interactions between FZDs and Wnts is known to trigger diverse signalling pathways classified as canonical and non-canonical Wnt signalling pathways[Citation120]. However, the exact mapping between specific Wnt-FZD interactions to a particular pathway is not clear. The common feature in the non-canonical FZD GPCR pathway is the recruitment of a scaffold protein dishevelled (Dvl) at the cytosolic end of the receptor upon binding of Wnts at their extracellular domain. It has been shown that Dvl forms higher oligomers to act as a signalling node, known as signalosome to further propagate the signal[Citation121]. With reference to GEF activation, Frizzled 7 (FZD7) is known to activate RhoA through WGEF (Weakly similar to Rho GEF 5). In Xenopus embryos, it has been shown that activation of FZD7 leads to recruitment of Dvl-2, which subsequently interacts with Dishevelled-associated activator of morphogenesis 1 (Daam-1) protein. This signal further propagates to activate WGEF. However, the exact mechanism of activation is not clearCitation122 (). It is interesting to note that WGEF is found to interact directly with the PDZ domain of Dvl-2 and with the N terminal domain of Daam-1 leading to its activation. Thus, it has been suggested that FZD7 could operate through multiple modes to activate WGEF[Citation122].

Activation of non-canonical GEFs through GPCRs

As mentioned previously, the non-canonical Dedicator of cytokinesis (DOCK) family GEFs, also known as Docker GEFs, contain DHR-1 and DHR-2 domains, in place of PH and DH domains of canonical GEFs. Similar to canonical GEFs, Docker GEFs are known to be activated through phosphorylation, protein–protein interactions, and/or lipid-mediated interactions. In HL60 and HeLa cell lines, activation of the chemokine receptor, CXCR4, triggers activation of Rac through Gβγ/ELMO1/DOCK180/Rac1 pathway. Similarly, in MDA-MB-231 cell lines, CXCR4 activates Rac through Gαi/ELMO1/DOCK180/Rac1 pathwayCitation69[Citation123], (). In both of these pathways Gβγ and Gαi subunits were found to interact with ELMO1, wherein Gαi interacts with N terminus of ELMO1,Citation69[Citation123], leading to conformational changes in the ELMO-DOCK complex, resulting in the release of DOCK from its autoinhibitory state. Another mechanism for DOCK180 activation by LPA is through a sequential complexation of Gαi2-src-p130Cas/ELMO/DOCK180Citation94 [Citation124,Citation125] (), in which Src kinase is shown to phosphorylate DOCK180 at Tyr1811, enhancing the interaction of DOCK180 with CrkII and p130Cas proteins, which activates Rac[Citation126].

Figure 6. Schematic representation of activation of non-canonical GEFs by GPCRs. (a) CXCR4 activates ELMO1/DOCK180 through the interaction of ELMO1 with Gβγ and Gαi subunits. (b) Stimulation of LPAR leads to Rac activation through Gαi2-Src-p130Cas/ELMO/DOCK180/Rac1 pathway during which Src is proposed to phosphorylate DOCK180 at Tyr1811. (c) ELMO/DOCK2 mediated Rac activation takes place through binding of DHR-1 domain of DOCK2 to lipid, PIP3 upon fMLPR activation. (d) ELMO1 and ELMO2 are shown to interact with the C terminal of the BAI1 receptor through their N-terminal fragments to trigger the DOCK/Rac pathway.

Figure 6. Schematic representation of activation of non-canonical GEFs by GPCRs. (a) CXCR4 activates ELMO1/DOCK180 through the interaction of ELMO1 with Gβγ and Gαi subunits. (b) Stimulation of LPAR leads to Rac activation through Gαi2-Src-p130Cas/ELMO/DOCK180/Rac1 pathway during which Src is proposed to phosphorylate DOCK180 at Tyr1811. (c) ELMO/DOCK2 mediated Rac activation takes place through binding of DHR-1 domain of DOCK2 to lipid, PIP3 upon fMLPR activation. (d) ELMO1 and ELMO2 are shown to interact with the C terminal of the BAI1 receptor through their N-terminal fragments to trigger the DOCK/Rac pathway.

DOCK2, another member of the DOCK family is shown to be activated through lipid-mediated interaction. This process involves fMLPR induced association of DHR-1 domain of DOCK2 with phosphatidylinositol 3, 4, 5-trisphosphate (PIP3) in a PI3K dependent manner [Citation127,Citation128], wherein interaction with PIP3 triggers GEF activity of DOCK2 towards Rac (). It has been also suggested that activation of chemokine receptors triggers DOCK2 through Gαi subunit, but the exact mechanism is unclear[Citation129]. In apoptotic cells, Brain-specific angiogenesis inhibitor 1 receptor (BAI1) is shown to trigger ELMO/DOCK/Rac signalling (). These signalling events depend on the interaction of cytosolic fragment of BAI1 (aa 1431–1582) with N terminal fragments of ELMO1 (aa 1–558) and ELMO2 (aa 1–520), which subsequently binds to DOCK1/DOCK2 [Citation130,Citation131]. Although the activation of non-canonical GEFs is similar to canonical GEFs, the pathways that operate are markedly different.

Summary

Activation of GTPases like RhoA, Cdc42, and Rac is a crucial step in cell migration, as they are the key players controlling the cytoskeletal rearrangement. This process is highly regulated in space and time by employing GEFs and GAPs. GEFs provide one of the important steps of regulation, wherein they switch the GTPases ‘ON’ by exchanging GDP with GTP. Based on sequence and structure GEFs can be classified as canonical and non-canonical. The common feature of both canonical and non-canonical GEFs is that they exist in an autoinhibitory inactive state () and release of this autoinhibitory state is essential for them to activate their cognate GTPases. The GEF activation process is done through multiple modes like phosphorylation of GEFs or interaction of GEFs with a different scaffold or activating proteins or through lipid interactions. In this review, we have consolidated how GPCRs play a crucial role in activating GEFs by exerting these activation mechanism. These events include the interaction of GEFs (through their RH and non-RH domains) with heterotrimeric G protein subunits activated by GPCRs, activation of kinases that phosphorylate GEFs, and protein/lipid interactions that release GEFs from their autoinhibited state. Complete understanding of these signalling events at the atomic level is crucial to decipher signalling processes, as this would be highly rewarding in addressing pathological conditions like cancer metastasis, immunological disorders, and neurodegenerative diseases.

Although the field of cell signalling is rapidly evolving, there are significant gaps in our understanding of signalling events that control cell migration, especially with reference to the role of GPCRs. Majority of the efforts, to date, are on mapping the key players involved in the signal transduction, which is largely driven by gene knockout/silencing and protein pull-down studies. However, the key questions like how in real-time the signals transverse from GPCRs to the downstream proteins? how concomitantly operating diverse pathways synchronize to exert cell migration? and how few GPCRs autonomously operate to control divergent pathways? remain unaddressed. Perhaps, innovative real-time imaging techniques, involving bioluminescence resonance energy transfer and fluorescence resonance energy transfer would help to dissect the system. Although some efforts have been made in this direction [Citation132–135], given the emerging complexity of the system, unravelling the signalling events that control cell migration remains challenging.

Acknowledgments

KK thank the ICGEB Research Grant [CRP/IND15] for the grant and AO would like to acknowledge the Council of Scientific & Industrial Research, India for fellowship.

Disclosure statement

The authors declare no conflicts of interest.

Additional information

Funding

This work was supported by the International Center for Genetic Engineering and Biotechnology [CRP/IND15].

References

  • Trepat X, Chen Z, Jacobson K. Cell migration. Compr Physiol. 2012;2(4):2369–2392.
  • Zegers MM, Friedl P. Rho GTPases in collective cell migration. Small GTPases 2014; 5:e983869.
  • Paoletti S, Petkovic V, Sebastiani S, et al. A rich chemokine environment strongly enhances leukocyte migration and activities. Blood. 2005;105(9):3405–3412.
  • Peplow PV, Chatterjee MP. A review of the influence of growth factors and cytokines in in vitro human keratinocyte migration. Cytokine. 2013;62(1):1–21.
  • Coombs VA, Nissen BK, Marks R, et al. The influence of temperature on epidermal cell migration in vitro. Br J Exp Pathol. 1973;54(6): 673–677. PMID: 4798773.
  • Su C, Zhang B, Liu W, et al. High extracellular pressure promotes gastric cancer cell adhesion, invasion, migration and suppresses gastric cancer cell differentiation. Oncol Rep. 2016;36(2):1048–1054.
  • Ridley AJ, Schwartz MA, Burridge K, et al. Cell migration: integrating signals from front to back. Science. 2003;302(5651):1704–1709.
  • Martin K, Reimann A, Fritz RD, et al. Spatio-temporal co-ordination of RhoA, Rac1 and Cdc42 activation during prototypical edge protrusion and retraction dynamics. Sci Rep. 2016;6(1):1–14.
  • Ridley AJ, Hall A. The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell. 1992;70(3):389–399.
  • Nobes CD, Hall A. Rho 1. nobes CD, hall A. Rho GTPases control polarity, protrusion, and adhesion during cell movement. J Cell Biol. 1999;144(6):1235–1244.
  • Nobes CD, Rho HA. Rac, and Cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell. 1995;81(1):53–62.
  • Van Leeuwen FN, Olivo C, Grivell S, et al. Rac activation by lysophosphatidic acid LPA1 receptors through the guanine nucleotide exchange factor Tiam1. J Biol Chem. 2003;278(1):400–406.
  • Manser E, Loo TH, Koh CG, et al. PAK kinases are directly coupled to the PIX family of nucleotide exchange factors. Mol Cell. 1998;1(2):183–192.
  • Bos JL, Rehmann H, Wittinghofer A. GEFs and GAPs: critical elements in the control of small G proteins. Cell. 2007;129(5):865–877.
  • Cho HJ, Kim BY, Baek KE, et al. Regulation of Rho GTPases by RhoGDIs in human cancers. Cells. 2019;8(9):1–12.
  • Buchsbaum RJ. Rho activation at a glance. J Cell Sci. 2007;120(7):1149–1152.
  • Dovas A, Couchman JR. RhoGDI: multiple functions in the regulation of Rho family GTPase activities. Biochem J. 2005;390(1):1–9.
  • Goicoechea SM, Awadia S, Garcia-Mata R. I’m coming to GEF you: regulation of RhoGEFs during cell migration. Cell Adhes Migr. 2014;8(6):535–549.
  • Zheng Y, Quilliam LA. Activation of the Ras superfamily of small GTPases.Workshop on exchange factors. EMBO Rep. 2003;4(5):463–468.
  • Hartmann S, Ridley AJ, Lutz S. The function of rho-associated kinases ROCK1 and ROCK2 in the pathogenesis of cardiovascular disease. Front Pharmacol. 2015;6:276.
  • Hilger D, Masureel M, Kobilka BK. Structure and dynamics of GPCR signaling complexes. Nat Struct Mol Biol. 2018;25(1):4–12.
  • Heng BC, Aubel D, Fussenegger M. An overview of the diverse roles of G-protein coupled receptors (GPCRs) in the pathophysiology of various human diseases. Biotechnol Adv. 2013;31:1676–1694.
  • Du Z, Lovly CM. Mechanisms of receptor tyrosine kinase activation in cancer. Mol Cancer. 2018;17:1–13.
  • Hamidi H, Ivaska J. Every step of the way: integrins in cancer progression and metastasis. Nat Rev Cancer. 2018;18(1):1–16.
  • Cotton M, Claing A. G protein-coupled receptors stimulation and the control of cell migration. Cell Signal. 2009;21(7):1045–1053.
  • Rossman KL, Der CJ, Sondek J. GEF means go: turning on Rho GTPases with guanine nucleotide-exchange factors. Nature Reviews Molecular Cell Biology. 2005;6(2):167–180.
  • Viaud J, Lagarrigue F, Ramel D, et al. Phosphatidylinositol 5-phosphate regulates invasion through binding and activation of Tiam1. Nat Commun. 2014;5(1):1–17.
  • Baumeister MA, Martinu L, Rossman KL, et al. Loss of phosphatidylinositol 3-phosphate binding by the C-terminal Tiam-1 pleckstrin homology domain prevents in vivo Rac1 activation without affecting membrane targeting. J Biol Chem. 2003;278(13):11457–11464.
  • Schmidt A, Hall A. Guanine nucleotide exchange factors for Rho GTPases: turning on the switch. Genes Dev. 2002;16(13):1587–1609.
  • Consonni SV, Brouwer PM, Van Slobbe ES, et al. The PDZ domain of the guanine nucleotide exchange factor PDZGEF directs binding to phosphatidic acid during brush border formation. PLoS One. 2014;9(5):e98253.
  • Fukuharaa S, Chikumi H, Silvio Gutkind J. RGS-containing RhoGEFs: the missing link between transforming G proteins and Rho? Oncogene. 2001;20(13):1661–1668.
  • Xu Z, Gakhar L, Bain FE, et al. The Tiam1 guanine nucleotide exchange factor is autoinhibited by its pleckstrin homology coiled-coil extension domain. J Biol Chem. 2017;292(43):17777–17793.
  • Premkumar L, Bobkov AA, Patel M, et al. Structural basis of membrane targeting by the Dock180 family of Rho family guanine exchange factors (Rho-GEFs). J Biol Chem. 2010;285(17):13211–13222.
  • Brugnera E, Haney L, Grimsley C, et al. Unconventional Rac-GEF activity is mediated through the Dock180-ELMO complex. Nat Cell Biol. 2002;4(8):574–582.
  • Kulkarni K, Yang J, Zhang Z, et al. Multiple factors confer specific Cdc42 and Rac protein activation by dedicator of cytokinesis (DOCK) nucleotide exchange factors. J Biol Chem. 2011;286(28):25341–25351.
  • Chang L, Yang J, Jo CH, et al. Structure of the DOCK2−ELMO1 complex provides insights into regulation of the auto-inhibited state. Nat Commun. 2020;11(1):1–17.
  • Lu M, Kinchen JM, Rossman KL, et al. A steric-inhibition model for regulation of nucleotide exchange via the Dock180 family of GEFs. Curr Biol. 2005;15(4):371–377.
  • Chen Z, Guo L, Hadas J, et al. Activation of p115-RhoGEF requires direct association of Gα13 and the Dbl homology domain. J Biol Chem. 2012;287(30):25490–25500.
  • Patel M, Karginov AV. Phosphorylation-mediated regulation of GEFs for RhoA. Cell Adhes Migr. 2014;8(1):11–18.
  • Booden MA, Siderovski DP, Der CJ. Leukemia-associated Rho Guanine nucleotide exchange factor promotes Gαq-coupled activation of RhoA. Mol Cell Biol. 2002;22(12):4053–4061.
  • Martin JW, Cavagnini KS, Brawley DN, et al. A Gα12-specific binding domain in AKAP-Lbc and p114RhoGEF. J Mol Signal. 2016;11:1–17.
  • Bartolomé RA, Molina-Ortiz I, Samaniego R, et al. Activation of Vav/Rho GTPase signaling by CXCL12 controls membrane-type matrix metalloproteinase-dependent melanoma cell invasion. Cancer Res. 2006;66(1):248–258.
  • Yamada T, Ohoka Y, Kogo M, et al. Physical and functional interactions of the lysophosphatidic acid receptors with PDZ domain-containing Rho guanine nucleotide exchange factors (RhoGEFs). J Biol Chem. 2005;280(19):19358–19363.
  • Kourlas PJ, Strout MP, Becknell B, et al. Identification of a gene at 11q23 encoding a guanine nucleotide exchange factor: evidence for its fusion with MLL in acute myeloid leukemia. Proc Natl Acad Sci U S A. 2000;97(5):2145–2150.
  • Medlin MD, Staus DP, Dubash AD, et al. Sphingosine 1-phosphate receptor 2 signals through leukemia-associated RhoGEF (LARG), to promote smooth muscle cell differentiation. Arterioscler Thromb Vasc Biol. 2010;30(9):1779–1786.
  • Wang Q, Liu M, Kozasa T, et al. Thrombin and lysophosphatidic acid receptors utilize distinct rhoGEFs in prostate cancer cells. J Biol Chem. 2004;279(28):28831–28834.
  • Goulimari P, Knieling H, Engel U, et al. LARG and mDia1 Link Gα 12/13 to Cell Polarity and Microtubule Dynamics. Mol Biol Cell. 2008;19(1):30–40.
  • Patel M, Kawano T, Suzuki N, et al. Gα13/PDZ-RhoGEF/RhoA signaling is essential for gastrin- releasing peptide receptor – mediated colon cancer cell migration. Molecular Pharmacology. 2014;86(3):252–262.
  • Struckhoff AP, Rana MK, Kher SS, et al. PDZ-RhoGEF is essential for CXCR4-driven breast tumor cell motility through spatial regulation of RhoA. J Cell Sci. 2013;126(19):4514–4526.
  • Mathew D, Kremer KN, Torres RM. ARHGEF1 deficiency reveals Gα13-associated GPCRs are critical regulators of human lymphocyte function. J Clin Invest. 2019;129(3):965–968.
  • Castillo-Kauil A, García-Jiménez I, Cervantes-Villagrana RD, et al. Gαsdirectly drives PDZ-RhoGEF signaling to Cdc42. J Biol Chem. 2020;295(50):16920–16928.
  • Slepak VZ, Pronin A. A Gs-RhoGEF interaction: an old G protein finds a new job. J Biol Chem. 2020;295(50):16929–16930.
  • Fukuhara S, Murga C, Zohar M, et al. A novel PDZ domain containing guanine nucleotide exchange factor links heterotrimeric G proteins to Rho. J Biol Chem. 1999;274(9):5868–5879.
  • Scarlett KA, White ESZ, Coke CJ, et al. Agonist-induced CXCR4 and CB2 heterodimerization inhibits Gα13/RhoA-mediated migration. Mol Cancer Res. 2018;16(4):728–739.
  • Ma X, Zhao Y, Daaka Y, et al. Acute activation of β2-adrenergic receptor regulates focal adhesions through βArrestin2- and p115RhoGEF protein-mediated activation of RhoA. J Biol Chem. 2012;287(23):18925–18936.
  • Chen Z, Guo L, Sprang SR, et al. Modulation of a GEF switch: autoinhibition of the intrinsic guanine nucleotide exchange activity of p115-RhoGEF. Protein Sci. 2011;20:(1):107–117.
  • Appert-Collin A, Cotecchia S, Nenniger-Tosato M, et al. The A-kinase anchoring protein (AKAP)-Lbc-signaling complex mediates α1 adrenergic receptor-induced cardiomyocyte hypertrophy. Proc Natl Acad Sci U S A. 2007;104(24):10140–10145.
  • Turton KB, Wilkerson EM, Hebert AS, et al. Expression of novel “LOCGEF” isoforms of ARHGEF18 in eosinophils. J Leukoc Biol. 2018;104(1):135–145.
  • Cervantes-Villagrana RD, Color-Aparicio VM, Reyes-Cruz G, et al. Protumoral bone marrow-derived cells migrate via Gβγ-dependent signaling pathways and exhibit a complex repertoire of RhoGEFs. J Cell Commun Signal. 2019;13(2):179–191.
  • Jaiswal M, Dvorsky R, Ahmadian MR. Deciphering the molecular and functional basis of Dbl family proteins: a novel systematic approach toward classification of selective activation of the Rho family proteins. J Biol Chem. 2013;288(6):4486–4500.
  • Welch HCE Regulation and function of P-Rex family Rac-GEFs. 2015; 6:49–70.
  • Niggli V. Signaling to migration in neutrophils: importance of localized pathways. Int J Biochem Cell Biol. 2003;35(12):1616–1638.
  • Mazaki Y, Hashimoto S, Tsujimura T, et al. Neutrophil direction sensing and superoxide production linked by the GTPase-activating protein GIT2. Nat Immunol. 2006;7(7):724–731.
  • Premont RT, Perry SJ, Schmalzigaug R, et al. The GIT/PIX complex: an oligomeric assembly of GIT family ARF GTPase-activating proteins and PIX family Rac1/Cdc42 guanine nucleotide exchange factors. Cell Signal. 2004;16(9):1001–1011.
  • Bustelo XR. The VAV family of signal transduction molecules. Crit. Rev. Oncog. 1996;7(1–2):65–88.
  • Razidlo GL, Schroeder B, Chen J, et al. Vav1 as a central regulator of invadopodia assembly. Curr Biol. 2014;24(1):86–93.
  • Fujikawa K, Miletic AV, Alt FW, et al. Vav1/2/3-null Mice Define an Essential Role for Vav Family Proteins in Lymphocyte Development and Activation but a Differential Requirement in MAPK Signaling in T and B Cells. J Exp Med. 2003;198(10):1595–1608.
  • Shepherd TR, Hard RL, Murray AM, et al. Distinct ligand specificity of the Tiam1 and Tiam2 PDZ domains. Biochemistry. 2011;50(8):1296–1308.
  • Wang Y, Xu X, Pan M, et al. ELMO1 directly interacts with Gβγ subunit to transduce GPCR signaling to Rac1 activation in chemotaxis. J Cancer. 2016;7(8):973–983.
  • Kunimura K, Uruno T, Fukui Y. DOCK family proteins: key players in immune surveillance mechanisms. Int Immunol. 2020;32(1):5–15.
  • Fukui Y, Hashimoto O, Sanui T, et al. Haematopoietic cell-specific CDM family protein DOCK2 is essential for lymphocyte migration. Nature. 2001;412(6849):826–831.
  • Diviani D, Soderling J, Scott JD. AKAP-Lbc anchors protein kinase a and nucleates Gα12-selective Rho-mediated stress fiber formation. J Biol Chem. 2001;276(47):44247–44257.
  • Cavin S, Maric D, Diviani D. A-kinase anchoring protein-Lbc promotes pro-fibrotic signaling in cardiac fibroblasts. Biochim Biophys Acta Mol Cell Res. 2014;1843(2):335–345.
  • Aittaleb M, Boguth CA, Tesmer JJG. Structure and function of heterotrimeric G protein-regulated Rho guanine nucleotide exchange factors. Mol Pharmacol. 2010;77(2):111–125.
  • O’Connor KL, Chen M, Towers LN. Integrin α6β4 cooperates with LPA signaling to stimulate Rac through AKAP-LBc-mediated RhoA activation. Am J Physiol Cell Physiol. 2012;302:2–11.
  • Paulucci-Holthauzen AA, Vergara LA, Bellot LJ, et al. Spatial distribution of protein kinase A activity during cell migration is mediated by A-kinase anchoring protein AKAP Lbc. J Biol Chem. 2009;284(9):5956–5967.
  • Blomquist A, Schwörer G, Schablowski H, et al. Identification and characterization of a novel Rho-specific guanine nucleotide exchange factor. Biochem J. 2000;352(2):319–325.
  • Niu J, Profirovic J, Pan H, et al. g protein βγ subunits stimulate p114rhogef, a guanine nucleotide exchange factor for RhoA and Rac1: regulation of cell shape and reactive oxygen species production. Circ Res. 2003;93(9):848–856.
  • Terry SJ, Elbediwy A, Zihni C, et al. Stimulation of cortical myosin phosphorylation by p114RhoGEF drives cell migration and tumor cell invasion. PLoS One. 2012;7(11):e50188.
  • Philippe JM, Lellouch AC, Lecuit T, Garcia De Las Bayonas A. Distinct RhoGEFs activate apical and junctional contractility under control of G Proteins during Epithelial Morphogenesis. Curr Biol. 2019;29(20):3370–3385.e7. .
  • Marivin A, Morozova V, Walawalkar I, et al. GPCR-independent activation of G proteins promotes apical cell constriction in vivo. J Cell Biol. 2019;218(5):1743–1763.
  • Souchet M, Portales-Casamar E, Mazurais D, et al. Human p63RhoGEF, a novel RhoA-specific guanine nucleotide exchange factor, is localized in cardiac sarcomere. J Cell Sci. 2002;115(Pt 3):629–640. PubMed 11861769.
  • Lutz S, Freichel-Blomquist A, Yang Y, et al. The guanine nucleotide exchange factor p63RhoGEF, a specific link between Gq/11-coupled receptor signaling and RhoA. J Biol Chem. 2005;280(12):11134–11139.
  • Lutz S, Shankaranarayanan A, Coco C, et al. Structure of Gαq-p63RhoGEF-RhoA complex reveals a pathway for the activation of RhoA by GPCRs. Science. 2007;318(5858):1923–1927. (80-).
  • Rojas RJ, Yohe ME, Gershburg S, et al. Gαq directly activates p63RhoGEF and trio via a conserved extension of the Dbl homology-associated pleckstrin homology domain. J Biol Chem. 2007;282(40):29201–29210.
  • Guo X, Stafford LJ, Bryan B, et al. A Rac/ Cdc42-specific exchange factor, GEFT, induces cell proliferation, transformation, and migration. J Biol Chem. 2003;278(15):13207–13215.
  • Kruse K, Lee QS, Sun Y, et al. N-cadherin signaling via Trio assembles adherens junctions to restrict endothelial permeability. J Cell Biol. 2019;218(1):299–316.
  • Cervantes-Villagrana RD, Adame-García SR, García-Jiménez I, et al. Gβγ signaling to the chemotactic effector P-REX1 and mammalian cell migration is directly regulated by Gαq and Gα13 proteins. J Biol Chem. 2019;294(2):531–546.
  • Sosa MS, Lopez-Haber C, Yang C, et al. Identification of the Rac-GEF P-Rex1 as an Essential Mediator of ErbB signaling in breast cancer. Mol Cell. 2010;40:(6):877–892.
  • Carretero-Ortega J, Walsh CT, Hernández-García R, et al. Phosphatidylinositol 3,4,5-triphosphate-dependent rac exchanger 1 (P-Rex-1), a guanine nucleotide exchange factor for rac, mediates angiogenic responses to stromal cell-derived factor-1/chemokine stromal cell derived factor-1 (SDF-1/CXCL-12) linked to rac. Mol Pharmacol. 2010;77(3):435–442.
  • Cash JN, Urata S, Li S, et al. Cryo-electron microscopy structure and analysis of the P-Rex1-Gβγ signaling scaffold. Sci Adv. 2019;5(10):eaax8855.
  • Damoulakis G, Gambardella L, Rossman KL, et al. P-Rex1 directly activates RhoG to regulate GPCR-driven Rac signalling and actin polarity in neutrophils. J Cell Sci. 2014;127(11):2589–2600.
  • Koh CG, Manser E, Zhao ZS, et al. β1PIX, the PAK-interacting exchange factor, requires localization via a coiled-coil region to promote microvillus-like structures and membrane ruffles. J Cell Sci. 2001;114(Pt 23): 4239–4251. PubMed:11739656.
  • Ward JD, Ha JH, Jayaraman M, et al. LPA-mediated migration of ovarian cancer cells involves translocalization of Gαi2 to invadopodia and association with Src and β-pix. Cancer Lett. 2015;356(2):382–391.
  • Feng Q, Baird D, Yoo S, et al. Phosphorylation of the cool-1/β-pix protein serves as a regulatory signal for the migration and invasive activity of Src-transformed cells. J Biol Chem. 2010;285(24):18806–18816.
  • Feng Q, Baird D, Peng X, et al. Cool-1 functions as an essential regulatory node for EGF receptor and Src-mediated cell growth. Nat Cell Biol. 2006;8(9):945–956.
  • Mayhew MW, Jeffery ED, Sherman NE, et al. Identification of phosphorylation sites in βPIX and PAK1. J Cell Sci. 2007;120(22):3911–3918.
  • Chahdi A, Miller B, Sorokin A. Endothelin 1 induces β1Pix translocation and Cdc42 activation via protein kinase A-dependent pathway. J Biol Chem. 2005;280(1):578–584.
  • Chahdi A, Sorokin A. The role of β1Pix/caveolin-1 interaction in endothelin signaling through Gα subunits. Biochem Biophys Res Commun. 2010;391(3):1330–1335.
  • Frank SR, Hansen SH. The PIX-GIT complex: a G protein signaling cassette in control of cell shape. Semin. 2008;19:234–244. Cell Dev. Biol.
  • Zhou W, Li X, Premont RT. Expanding functions of GIT Arf GTPase-activating proteins, PIX Rho guanine nucleotide exchange factors and GIT-PIX complexes. J Cell Sci. 2016;129(10):1963–1974.
  • Feng Q, Albeck JG, Cerione RA, et al. Regulation of the Cool/Pix proteins. Key binding partners of the Cdc42/Rac targets, the p21-activated kinases. J Biol Chem. 2002;277(7):5644–5650.
  • Yoshii S, Tanaka M, Otsuki Y, et al. αPIX nucleotide exchange factor is activated by interaction with phosphatidylinositol 3-kinase. Oncogene. 1999;18(41):5680–5690.
  • Lyda JK, Tan ZL, Rajah A, et al. Rac activation is key to cell motility and directionality: an experimental and modelling investigation. Comput Struct Biotechnol J. 2019;17:1436–1452.
  • Lawson CD, Donald S, Anderson KE, et al., Welch HCE. P-Rex1 and Vav1 cooperate in the regulation of Formyl-Methionyl-Leucyl-Phenylalanine–Dependent Neutrophil Responses. J Immunol. 2011;186(3):1467–1476. .
  • Heo J, Thapar R, Campbell SL. Recognition and activation of Rho GTPases by Vav1 and Vav2 Guanine nucleotide exchange factors †. Biochemistry. 2005;17(17):6573–6585.
  • Bustelo XR. Regulatory and signaling properties of the Vav Family. Mol Cell Biol. 2000;20(5):1461–1477.
  • Vila‐Coro AJ, Rodríguez‐Frade JM, De Ana AM, et al. The chemokine SDF‐lα triggers CXCR4 receptor dimerization and activates the JAK/STAT pathway. Faseb J. 1999;13(13):1699–1710.
  • Fumagalli L, Zhang H, Baruzzi A, et al. The Src family Kinases Hck and Fgr regulate Neutrophil responses to N Formyl-Methionyl-Leucyl-Phenylalanine. J Immunol. 2007;178(6):3874–3885.
  • Aghazadeh B, Lowry WE, Huang XY, et al. Structural basis for relief of autoinhibition of the Dbl homology domain of proto-oncogene Vav by tyrosine phosphorylation. Cell. 2000;102(5):625–633.
  • Fleming IN, Elliott CM, Buchanan FG, et al. Ca2+/Calmodulin-dependent Protein Kinase II regulates Tiam1 by reversible protein Phosphorylation. Biochemistry. 1999;274:12753–12758.
  • Miyamoto Y, Yamauchi J, Tanoue A, et al. TrkB binds and tyrosine-phosphorylates Tiam1, leading to activation of Rac1 and induction of changes in cellular morphology. Proc Natl Acad Sci U S A. 2006;103:10444–10449.
  • Minard ME, Kim LS, Price JE, et al. The role of the guanine nucleotide exchange factor Tiam1 in cellular migration, invasion, adhesion and tumor progression. Breast Cancer Res Treat. 2004;84(1):21–32.
  • Wang S, Watanabe T, Matsuzawa K, et al. Tiam1 interaction with the PAR complex promotes talin-mediated Rac1 activation during polarized cell migration. J Cell Biol. 2012;199(2):331–345.
  • Mertens AEE, Pegtel DM, Collard JG. Tiam1 takes PARt in cell polarity. Trends Cell Biol. 2006;16(6):308–316.
  • Fleming IN, Gray A, Downes CP. Regulation of the Rac1-specific exchange factor Tiam1 involves both phosphoinositide 3-kinase-dependent and -independent components. Biochem J. 2000;351(1):173–182.
  • Hoshino M, Sone M, Fukata M, et al. Identification of the stef gene that encodes a novel guanine nucleotide exchange factor specific for Rac1. J Biol Chem. 1999;274(25):17837–17844.
  • Goto A, Hoshino M, Matsuda M, et al. Phosphorylation of STEF/Tiam2 by protein kinase A is critical for Rac1 activation and neurite outgrowth in dibutyryl cAMP-treated PC12D cells. Mol Biol Cell. 2011;22(10):1780–1790.
  • Emami-Nemini A, Gohla A, Urlaub H, et al. The guanine nucleotide exchange factor Vav2 is a negative regulator of parathyroid hormone Receptor/Gq Signaling. Mol Pharmacol. 2012;82(2):217–225.
  • Schulte G, Wright SC. Frizzleds as GPCRs – more conventional than we thought! trends. 2018;39:828–842. Pharmacol Sci.
  • Gammons MV, Renko M, Johnson CM, et al. Wnt Signalosome Assembly by DEP Domain Swapping of Dishevelled. Mol Cell. 2016;64(1):92–104.
  • Tanegashima K, Zhao H, Dawid IB. WGEF activates Rho in the Wnt-PCP pathway and controls convergent extension in Xenopus gastrulation. Embo J. 2008;27(4):606–617.
  • Li H, Yang L, Fu H, et al. Association between Gαi2 and ELMO1/Dock180 connects chemokine signalling with Rac activation and metastasis. Nat Commun. 2013;4:1–12.
  • Ward JD, Dhanasekaran DN. LPA Stimulates the Phosphorylation of p130Cas via Gαi2 in Ovarian Cancer Cells. Genes Cancer. 2012;3(9–10):578–591.
  • Matsui H, Harada I, Sawada Y. Src, p130Cas, and Mechanotransduction in cancer cells. Genes Cancer. 2012;3(5–6):394–401.
  • Feng H, Hu B, Liu KW, et al. Activation of Rac1 by Src-dependent phosphorylation of Dock180Y1811 mediates PDGFRα-stimulated glioma tumorigenesis in mice and humans. J Clin Invest. 2011;121(12):4670–4684.
  • Kunisaki Y, Nishikimi A, Tanaka Y, et al. DOCK2 is a Rac activator that regulates motility and polarity during neutrophil chemotaxis. J Cell Biol. 2006;174(5):647–652.
  • Wang J, Xu L, Shaheen S, et al. Growth of B Cell receptor microclusters is regulated by PIP2 and PIP3 Equilibrium and Dock2 recruitment and activation. Cell Rep. 2017;21(9):2541–2557.
  • Thelen M, Stein JV. How chemokines invite leukocytes to dance. Nat Immunol. 2008;9:(9):953–959.
  • Park D, Tosello-Trampont AC, Elliott MR, et al. BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature. 2007;450(7168):430–434.
  • Weng Z, Situ C, Lin L, et al. Structure of BAI1/ELMO2 complex reveals an action mechanism of adhesion GPCRs via ELMO family scaffolds. Nat Commun. 2019;10(1). 10.1038/s41467-018-07938-9
  • Afm TI, Yue H, Scavello M, et al. The cAMP-induced G protein subunits dissociation monitored in live Dictyostelium cells by BRET reveals two activation rates, a negative effect of caffeine and potential role of microtubules. Cell Signal. 2018;48:25–37.
  • Islam AFMT, Stepanski BM, Charest PG Studying chemoattractant signal transduction dynamics in Dictyostelium by BRET. In: Methods in Molecular Biology. 2016;1407: 63–77.
  • Galés C, Rebois RV, Hogue M, et al. Real-time monitoring of receptor and G-protein interactions in living cells. Nat Methods. 2005;3(3):177–184.
  • Xu X, Brzostowski JA, Jin T. Monitoring dynamic GP CR signaling events using fluorescence microscopy, FRET imaging, and single-molecule imaging. Methods Mol Biol. 2009;571:371–383.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.