(1)
Project-team INRIA-UPMC-CNRS REO Laboratoire Jacques-Louis Lions, CNRS UMR 7598, Université Pierre et Marie Curie, Place Jussieu 4, Paris Cedex 05, France
Abstract
Guanosine triphosphatases intervene in: (1) signal transduction from the intracellular edge of the plasma membrane and intracellular domain of transmembrane receptors; (2) protein synthesis at the ribosome (Vol. 1 – Chap. 5. Protein Synthesis); (3) control of cell division (Vol. 2 – Chap. 2. Cell Growth and Proliferation); (4) proper protein folding; (5) translocation of proteins through the membrane of the endoplasmic reticulum; and (6) vesicular transport within the cell (Vol. 1 – Chap. 9. Intracellular Transport).
Guanosine triphosphatases intervene in: (1) signal transduction from the intracellular edge of the plasma membrane and intracellular domain of transmembrane receptors; (2) protein synthesis at the ribosome (Vol. 1 – Chap. 5. Protein Synthesis); (3) control of cell division (Vol. 2 – Chap. 2. Cell Growth and Proliferation); (4) proper protein folding; (5) translocation of proteins through the membrane of the endoplasmic reticulum; and (6) vesicular transport within the cell (Vol. 1 – Chap. 9. Intracellular Transport).
Guanosine triphosphatases include: (1) translation factors of the TRAFAC family that control initiation, elongation, and termination of protein synthesis (or translation at ribosomes; they act as ribosome-assembly factors); (2) translocation ribonucleoproteic factors;1 (3) large GTPases, such as dynamin 2 and members of the septin family3 of polymerizing GTPases; (4) heterotrimeric, relatively large, guanine nucleotide-binding proteins, the so-called G proteins; and (5) monomeric, small guanosine triphosphatases (small GTPases).4
In addition, proteins require assistance of molecular chaperones to get a functional conformation or for refolding of unfolded or misfolded proteins that underwent a stress. Among these chaperones, chaperonins are large complexes that enhance the efficiency of protein folding.5 The reaction cycle consists of a nucleotide-regulated conformational change between an open substrate-acceptor state and a closed folding-active state. Cytosolic, hetero-oligomeric chaperonin with ATPase and GTPase activity (at similar hydrolysis rate) chaperonin containing TCP1 (CCT)6 assists in protein folding. Its 8 different subunits have an ATPase domain and distinct substrate-binding domain. Its substrates include actin and tubulin. Nucleotide GTP induces an open-to-close conformational change in the CCT structure, hence regulating protein folding [776].
Receptor activation primes chemical events that can involve heterotrimeric and monomeric guanosine triphosphatases. These 2 types of proteins — G proteins and small GTPases — cycle between 2 states, as they bind either guanosine di- (GDP) or triphosphate (GTP). These proteic switches are flicked off (inactive, GDP-bound state [GTPaseGDP]) and on (active, GTP-bound state [GTPaseGTP]; Fig. 9.1).7 This hydrolytic activation–deactivation cycle is regulated to avoid pathological states. Once activated, the switch activates the pathway effector immediately downstream from it.


Fig. 9.1
Small GTPase states and effectors. They cycle between active, guanosine triphosphate (GTP)-bound and inactive, guanosine diphosphate (GDP)-loaded states. This cycling is controlled by 3 types of regulatory proteins: (1) guanine nucleotide-dissociation inhibitors (GDI) that stabilize inactive form; (2) guanine nucleotide-exchange factors (GEF) that catalyze the exchange of GDP for GTP; and (3) GTPase-activating proteins (GAP) that enhance the low intrinsic GTPase activity of GTPases.
9.1 Heterotrimeric G Proteins (Gαβγ GTPases)
Heterotrimeric guanine nucleotide-binding proteins (G proteins) are signal transducers attached to the inner leaflet of the plasma membrane. These transducers respond to various extracellular signals, such as hormones, neurotransmitters, chemokines, photons, odorants, tastants, nucleotides, and ions. They are controlled by a set of regulators. Conversely, they regulate the production or influx of second messengers, such as cAMP and calcium. The activation of G proteins is induced by ligand-bound G-protein-coupled receptors (Fig. 9.2; Vol. 3 – Chap. 7. G-Protein-Coupled Receptors).8


Fig. 9.2
G-protein-coupled receptors (GPCR) are associated with 3 G-protein subunits: Gα that binds GDP (inactive state) or GTP after ligand binding (stimulated form), Gβ, and Gγ. Subunit Gα shares homology with small monomeric GTPase of the RAS superfamily. It binds guanine nucleotides in a signal-dependent manner. Heterodimer Gβγ has high affinity for GαGDP form. It is released from active GαGTP after receptor activation. Activated Gα activates an effector. Types of Gα subunits include Gαs (stimulatory), Gαi (inhibitory), Gαq, Gαt, and Gα12. Subunit Gαs stimulates adenylate cyclase (ACase) that produces cAMP messenger. On the other hand, Gαi inhibits adenylate cyclase. Gαq-coupled receptor activates phospholipase-C (PLC) that generates second messengers inositol trisphosphate (IP3) and diacylglycerol (DAG). Subunit Gαt stimulates guanylate cyclase (GCase) that forms cGMP. Subunit Gα12 activates small RhoA GTPase. Protein kinase-A (PKA) regulates myocardial contraction, acting on contractile proteins, activating ion channels, such as ryanodine receptors (RR) of the endoplasmic reticulum (ER), voltage-dependent calcium channels (VDCC), and sodium channels of the plasma membrane (Gβγ inhibits these voltage-gated channels), and carriers. Protein kinase-A also increases metabolic energy, stimulating phosphorylase kinases and hormone-sensitive lipases, and activates gene transcription. Messenger cAMP exerts direct effects on other effectors, such as Rap1GEF. Mediator IP3 binds to its endoplasmic reticulum receptor IP3R and causes the release of stored Ca
which rises transiently in the cytosol. Phospholipase-C also activates plasmalemmal transient receptor potential (TRP) Ca
channels (TRPC), hence Ca
influx (adapted from [777]).



Heterotrimeric G protein is composed of a GαGDP subunit and a Gβ–Gγ dimer (Fig. 9.3). Many isoforms of each of the 3 subunits exist. Genes have been identified for 16 Gα, grouped in 4 main subclasses (Tables 9.1 and 9.2), 5 Gβ, and 15 Gγ subunits, with possible splice variants and post-translational modifications [778]. The G-protein subunit genes encode for [778]: (1) subclass-1 of Gαs subunits with Gs and Gαolf; (2) subclass-2 of Gαi subunits with Gi1 to Gi3, Go1, Go2, Gt1, Gt2, Ggust (gustducin), and Gz; (3) subclass-3 of Gαq subunits with G11, Gq, and G14 to G16; (4) subclass-4 of Gα12 subunits with G12 and G13; (5) subclass of Gβ subunits with Gβ1 to Gβ5; and (6) subclass of Gγ subunits with farnesylated isoforms Gγ1, Gγ11, and Gγ9 (Gγ8cone or Gγ14), and geranyl-geranylated isoforms Gγ2 to Gγ5, Gγ7, Gγ8 (a.k.a. Gγ8olf), Gγ10, and Gγ12.
Table 9.1
The 4 main subclasses of Gα and corresponding targets. Activated GPCRs stimulate G proteins, as they catalyze guanine nucleotide-exchange on Gα subunit. G Proteins are classified according to Gα subunit into Gs, Gi/o, Gq/11, and G12/13. Members of the Gs and Gi/o subclasses converge to the second messenger cAMP. Second messenger activation occurs quickly due to the presence of complexes between Gs and adenylate cyclase as well as Gq and phospholipase-Cβ, but substantial change in second messenger concentration needs much longer time in comparison with the activation kinetics of initial signaling stages.
Gα | Effector |
---|---|
Gs | Adenylate cyclase (stimulation) |
Gi/o | Adenylate cyclase (inhibition) |
Gq/11 | Phospholipase-Cβ |
G12/13 | Small guanosine triphosphatase |
Subunit | Pathway | Effect |
---|---|---|
Gs | ACase–cAMP–PKA | Positive inotropy and lusitropy |
Gi/o | ACase | Inhibition |
NO | Vasodilation | |
PLC–IP3–Ca ![]() | Vasoconstriction | |
PLC–DAG–PKC | ||
Gq/G11 | PLC–IP3–Ca ![]() | Vasoconstriction |
PLC–DAG–PKC | Positive inotropy | |
G12/13 | RhoGEF–Rho–RoCK | Vasoconstriction |
Gβγ | KACh | Negative chronotropy |
A single receptor can stimulate a single G protein (linear signaling). Some receptors promiscuously interact with G proteins, whereas others specifically interact. A given G protein can be regulated by several receptors, either positively or negatively (convergent receptor signaling). A given receptor can activate several G proteins (divergent receptor signaling), such as the α-adrenergic receptor targeting both Gs and Gi subunits.
Activated G protein is dissociated into GαGTP and Gβ–Gγ (or Gβγ; Fig. 9.3). Isolated GαGTP acts on a second messenger. However, although certain G proteins dissociate after receptor activation in vivo, other G proteins undergo simple rearrangements, possibly modulating G-protein effector activity [780].
Once the target has been stimulated, Gα fills its GTPase activity. When GTP is hydrolyzed, Gα returns to the inactivated GDP-bound conformation, dissociates from its target, and reassembles with Gβγ. The duration of G-protein activation is thus controlled by the intrinsic GTPase activity of Gα subunit.
Subunits GαGTP and Gβγ can propagate signals, as they interact with adenylate cyclases, phospholipase-C isoforms, potassium and calcium ion channels, guanine nucleotide-exchange factors for RhoA GTPase, and other effectors.
However, certain ligands modify the conformation of the receptor such that it interacts with either heterotrimeric G proteins or another effector, such as GPCR kinases (GRK) that phosphorylate activated GPCRs and arrestins that bind phosphorylated GPCRs (β-arrestin-biased ligands) and support clathrin-mediated GPCR endocytosis.
Certain G proteins are regulated by homodimerization. These G proteins, activated by nucleotide-dependent dimerization, belong to the set of proteins that also includes signal recognition particles, dynamins, septins, and receptor-interacting protein kinase RIPK7 [775].
In addition to GPCR types and effectors, accessory proteins influence specificity, magnitude, and duration of GPCR signaling. GPCR signaling accessory proteins include scaffold proteins and molecules that directly affect the basal activation state of effectors, such as regulators of G-protein signaling and activators of G-protein signaling (Sect. 9.2).
In dual signaling pathways activated by a single G protein, both activated Gα and Gβγ stimulate effectors. In other pathways, the major regulator is Gβγ, the activity of which can be suppressed by excess Gα liberated by other activated G proteins.
9.1.1 Set of Gα Subunits
Four categories of Gα subunits include: (1) subclass of Gs that stimulates adenylate cyclase and activates calcium channels (Sect. 9.1.1.1); (2) subclass of Gi that inhibits adenylate cyclase and potassium channels (Sect. 9.1.1.2); (3) subclass of Gq that activates phospholipase-C, and protein kinase-C and -D (Sect. 9.1.1.3); and (4) subclass of G12 that regulate cell activity, especially actin cytoskeleton remodeling, via guanine nucleotide-exchange factors (Sect. 9.1.1.4).
9.1.1.1 Gα s Subclass
Ubiquitous Gαs is an activator of adenylate cyclase that produces messenger cAMP from ATP. In its quiescent state, Gαs is associated with Gβ/γ dimer. The intrinsic GTPase activity of Gs causes its deactivation and reassociation with Gβ/γ dimer. In its inactive form, Gs binds to Gβ1/γ1, Gβ1/γ2, Gβ1/γ5, Gβ1/γ7, Gβ2/γ5, and Gβ2/γ7 [781].
Subunit Gαs is activated by G-protein-coupled receptors, such as β-adrenergic, glucagon, luteinizing hormone, calcitonin, and dopamine D1 and D5 receptors [781]. Subunit GsGTP activates adenylate cyclase isoforms. Affinity of Gs for both Gβ/γ and adenylate cyclase is increased by N-terminal palmitoylation.
9.1.1.2 Gα i Subclass
Several types of Gαi exist (Gαi1–Gαi3) that mainly inhibit the cAMP-dependent pathway, but activate the phospholipase-C pathway. In fact, the Gi/o subclass is composed not only of Gαi, but also Gαo, Gαgust, and Gαt (transducin) proteins (Table 9.3).
Table 9.3
Examples of G-protein-coupled receptors (GPCRs) preferentially coupled to members of the Gi/o subclass (Source: [785]).
GPCR type | Gα subunit type |
---|---|
A1 | Gαi1–Gαi2, Gαo |
AT2 | Gαi1–Gαi2, Gαo |
APJ | Gαi1–Gαi2 |
CB1 | Gαi1–Gαi3, Gαo |
D2 | Gαi1 |
GABAB | Gαi1, Gαo |
mGluR2/3 | Gi/o |
LPA1 | Gαi1, Gαo1 |
M2 | Gαi1–Gαi3, Gαo |
M4 | Gαi2, Gαo |
Op3 | Gαi1, Gαi3, Gαo |
P2Y12 | Gi |
S1P1 | Gαi1, Gαi3 |
5HT1A | Gαi1–Gαi3 |
α2a | Gαi1–Gαi2 |
Cells respond to growth factors by either migrating or proliferating. Cell movement and proliferation are indeed mutually exclusive. Growth factor receptors, such as EGFR, VEGFR, and PDGFR receptors, can actually trigger cell motion or division according to the type and concentration of the signaling ligand on the one hand and the density and distribution of receptors on the other. During cell migration, phospholipase-Cγ1 and phosphatidylinositol 3-kinase and its effector protein kinase-B are stimulated and coupled to actin remodeling at the leading edge. In proliferating cells, another set of signals (ERK1 and ERK2, Src kinase, and STAT5b) is involved for the activation of nuclear transcription factors and DNA synthesis. Girdin (girders of actin filaments)9 is an actin-binding PKB substrate10 that operates in actin organization and PKB-dependent cell motility, at least, in fibroblasts. It is phosphorylated upon stimulation by insulin-like growth factor IGF1 that fosters tumor cell movement [782].11 Girdin is a guanine nucleotide-exchange factor for G proteins involved in the regulation of the PI3K–PKB axis, actin cytoskeleton remodeling, and cell migration.12 Proteic subunit Gαi and its ubiquitous guanine nucleotide-exchange factor girdin orchestrate the migration–proliferation dichotomy downstream from EGFR signaling [784]. Girdin directly interacts with EGFR and associates Gαi to the receptor. A Gαi–girdin–EGFR complex indeed assembles, EGFR autophosphorylates (a process necessary for cell migration, but not for mitosis), and EGFR residence in the plasma membrane is prolonged (signaling from the cell surface rather than from endosomes). Plasma membrane-based motogenic signals (PLCγ1 and the PI3K–PKB axis) are triggered and cell migration starts. Activation of Gαi by girdin is required in cell migration primed by EGF and insulin during epithelial wound healing, macrophage chemotaxis, and tumor cell migration. In addition, cancer angiogenesis, invasion, and metastasis also rely on girdin mediator following VEGF and IGF stimulation. On the other hand, a GEF-deficient girdin splice variant promotes mitogenic signals and cell proliferation occurs.13
Gα i1
Ubiquitous Gαi1 inhibits adenylate cyclases AC1, AC5, and AC6 [785]. On the other hand, associated Gβγ primes the activation of acetylcholine-activated inwardlyrectifying K + GIRK channels (KIR3). In addition, Gαi1 can also activate KIR3 activity.
In platelets, adrenaline stimulates the association of Src with Gαi1 Protein Gαi1 can then trigger the PI3Kγ–PKB pathway. In endothelial cells, stimulation of β2-adrenoceptor activates the Gαi1–Src–PKB axis, whereas in cardiomyocytes, this receptor activates predominately the Gβγ–PI3K–PKB pathway [785].
Activated Gi (Gi1–Gi3)-coupled receptors can signal via Jun N-terminal kinase. Protein Gαi1 mediates not only GPCR signaling, but also that of receptor Tyr kinases. Both Gαi1 and GGαi3 downstream from epidermal growth factor receptor use GAB1 to activate the PKB–TORC1 pathway [785].
Protein Gαi1 can be N-myristoylated and palmitoylated to enhance membrane insertion and affinity for Gβγ subunit on the one hand, and to reduce the affinity for the GAP and RGS4 proteins [785].
Protein Gαi1 intervenes in synaptic transmission and platelet aggregation. It is linked to serotonin 5HT1A , dopamine D2, cannabinoid CB1, metabotropic glutamate mGluR2 and mGluR3, muscarinic acetylcholine M2, and μ-opioid, as well as adenosine A1, angiotensin-2 AT2, apelin, sphingosine-1-phosphate S1P1, lysophosphatidic acid LPA1, and metabotropic P2Y12 receptors [785]. Gi1-coupled α2a-adrenoreceptor activates voltage-dependent CaV channels in rat portal vein myocytes.
Gα o
The “other” G-protein subunit Gαo that constitute the Go protein with Gβγ dimer is the most abundant G protein in the central and peripheral nervous systems. Two splice variants — Gαo1 and Gαo2 — exist. It is activated not only by many G-protein-coupled receptors bound to neurotransmitters and hormones (Table 9.4), but also by 43-kDa growth cone-associated protein GAP43,14 and amyloid-β (A4) precursor protein (APP) [786].
Chemokine receptors | CXCR4 |
---|---|
Morphogen receptors | Fz |
Neurotransmitter GPCRs | α2a, D2, 5HT1P, |
M2, M4 (Go1), GABAB | |
Nucleoside receptors | A1 |
Nucleotide receptors | P2Y2 |
Opioid receptors | Op1, Op3, Op4 |
Peptide receptors | Galanin, neuropeptide-Y, somatostatin (Sst2; Go2), |
corticotropin-releasing factor receptor (CRF2) |
Protein Go also plays a role in visual signal transduction, olfactory reception, and neurotransmitters monoamine and glutamate uptake after exocytosis by their cognate vesicle membrane transporters [786]. Frizzled receptor that transmits developmental signals relies on Gαo subunit.
In the heart, Go protein is preferentially synthesized in endocrine atriomyocytes and peripheral neurons. It enhances calcium cycling and contractility [786]. In addition, it supports the cardiac control by the parasympathetic activity mediated by muscarinic acetylcholine receptors.
Activated Gαo can interact directly with many types of ion channels [786]. It impedes transmitter release, as it precludes the activity ofCaV2.2 channel in presynaptic terminals. Moreover, opioid receptors inhibit CaV2.1 channel in dorsal root ganglion neurons. In ventriculomyocytes, muscarinic GPCRs prevents the activation of CaV1.2a channel. On the other hand, Go protein activateKIR3 (GIRK) channel. It also modulates Na + channel gating and prevents the activaty of transient receptor potential cation channel TRPM1L in retinal bipolar cells.
Protein Gαo directly binds to Rap1GAP protein, then targeting it for proteasomal degradation. It also interacts with Rab5 and Rit1 GTPases. It activates the GTPase activity of tubulin. Activated Gαo protein interacts directly with zinc finger and BTB domain-containing transcription factor ZBTB16, thereby enhancing its transcriptional repression.
Gα z
The Gz subunit is activated by GPCRs (adenosine [A1], adrenergic [α2a], complement C5a, corticotropin-releasing hormone, dopaminergic receptors [D2–D5], endothelin, fractalkine, lysophosphatidic acid, macrophage inflammatory protein-3α, melatonin [MT1 and MT2], muscarinic [M2], neurokinin, nociceptin, opioids, serotonin receptors [5HT1A ], and thrombin) that activate other members of the Gi subclass. It is unique among the Gi subclass, with very low rates of GDP–GTP exchange (0.02/mn) and GTP hydrolysis (0.05/mn).
Because Gz is recognized by several RGS proteins (RGS4, RGS10, RGS17, RGS19, RGS20, and retinal RGS1 [RetRGS1]), sustained state of activation due to slow GTP hydrolysis can be shortened [788]. Myristoylation and palmitoylation are needed for stable anchorage to the plasma membrane. Palmitoylation prevents GTP hydrolysis by RGS proteins. Protein Gz is phosphorylated by protein kinase-C and P21-activated protein kinase. Phosphorylation disrupts Gαz–Gβγ interaction. Phosphorylation (Ser16) blocks binding to RGS17, RGS19, and RGS20 proteins.
Protein Gz directly binds and inhibits adenylate cyclases AC1, AC5, and AC6, but indirectly stimulates adenylate cyclase AC2 [788]. Its other effects include: (1) inhibition of CaV2.2 channels; (2) stimulation of inwardly rectifying K + channels; (3) repression of Rap1 signaling by interacting with Rap1 GTPase-activating protein; (4) release of tumor-necrosis factor-α from immunoglobulin-E-stimulated mastocytes; (5) natural killer cell-mediated lysis of allogeneic and tumor cells; and (6) reduction in insulin secretion.
9.1.1.3 Gα q Subclass
Subunit Gαq links certain types of activated G-protein-coupled receptors to intracellular signaling cascades. Various types of GPCRs couple to Gαq, such as acetylcholine, angiotensin, catecholamine, endothelin, glutamate, histamine, lysophospholipid, and serotonin receptors [789] (Tables 9.5 to 9.8).
Table 9.5
Examples of plamalemmal, heptahelical Gαq ∕ 11-protein-coupled receptors (Part 1; Source: [789]).
Type | Name |
---|---|
Receptors for amino acids and dicarboxylic acids | |
Metabotropic glutamate receptor | mGluR1, mGluR5 (class C) |
α-Ketoglutarate receptor | GPR99/OXGR1 (subfamily A11) |
or oxoglutarate receptor | |
Succinate receptor | GPR91 (subfamily A11) |
LL-amino acid receptor | GPRC6A (class C) |
(Larginine and Llysine) | |
Receptors for biogenic amines | |
Muscarinic acetylcholine receptor | M1, M3, M5 (subfamily A18) |
Adrenergic receptor | α1A, α1B, α1D (subfamily A18) |
Histamine receptor | H1 (subfamily A18) |
Serotonin receptor | 5HT2A, 5HT2B, 5HT2C (subfamily A17) |
Table 9.6
Examples of plamalemmal, heptahelical Gαq ∕ 11 protein-coupled receptors (Part 2; Source: [789]).
Type | Name |
---|---|
Receptors for ions | |
Calcium-sensing receptor | CaSR (class C) |
Receptors for nucleotides and nucleosides | |
Purinergic receptor | P2Y1 (ADP/ATP), P2Y2 (UTP/ATP), |
(subfamily A11) | P2Y4 (UTP/ATP), P2Y6 (UDP), |
P2Y11 (ATP) |
Table 9.7
Examples of plamalemmal, heptahelical Gαq ∕ 11 protein-coupled receptors (Part 3; Source: [789]).
Type | Name |
---|---|
Receptors for lipids | |
Fatty acid receptor | GPR40, GPR43, GPR120 (subfamily A11) |
Leukotriene C4/D4 receptor | CysLT1, CysLT2 (subfamily A5) |
Lysophosphatidic acid receptor | LPA1/2/3/5 (subfamily A13) |
Platelet-activating factor receptor | PAF (subfamily A12) |
Prostaglandin F2a receptor | PF2R (subfamily A14) |
Prostaglandin E2 receptor | EP1, EP3 (subfamily A14) |
Sphingosine-1-phosphate receptor | S1P2, S1P3 (subfamily A13) |
Thromboxane A2 receptor | TP (subfamily A14) |
Table 9.8
Examples of plamalemmal, heptahelical Gαq ∕ 11 protein-coupled receptors (Part 4; Source: [789]).
Type | Name |
---|---|
Receptors for peptides and proteins | |
Angiotensin-2 receptor | AT1A, AT1B (subfamily A3) |
Bradykinin receptor | B1, B2 (subfamily A3) |
Calcitonin receptor | CT (class B) |
Calcitonin gene-related peptide receptor | CGRP1 (CALCRL; class B) |
Cholecystokinin-8 receptor | CCK1, CCK2 (subfamily A6) |
Endothelin-1, -2, -3 receptors | ETA, ETB (subfamily A7) |
Galanin receptor | GAL2 (subfamily A5) |
Gastrin-releasing peptide/bombesin receptor | BB2 (subfamily A7) |
Ghrelin receptor | GHSR (subfamily A7) |
Gonadotropin-releasing hormone receptor | GnRHR (subfamily A6) |
Kisspeptin and metastin receptor | GPR54 (subfamily A5) |
Melanin-concentrating hormone receptor | MCHR2 (subfamily A5) |
Orexin A/B receptor | OX1R, OX2R (subfamily A6) |
Oxytocin receptor | OxTR (class A) |
Parathyroid hormone receptor | PTH (class B) |
Parathyroid-related peptide receptor | PTHrP1, PTHrP2 (class B) |
Prokineticin-1/2 receptor | PKR1, PKR2 (subfamily A9) |
Substance-P | NK1R (subfamily A9) |
Neurokinin-A/B receptor | NK2R (subfamily A9) |
Thyrotropin receptor | TSH (subfamily A10) |
Thyrotropin-releasing hormone receptor | TRH1, TRH2 (subfamily A10) |
Urotensin-2 receptor | Uts2R (GPR14) (subfamily A5) |
Vasopressin receptor | V1a, V1b (subfamily A6) |
Receptors for peptidases | |
Thrombin receptors | PAR1, PAR3, PAR4 |
(subfamily A15) | |
Trypsin receptor | PAR2 (subfamily A15) |
The subclass of ubiquitous Gαq subunits include 4 families: Gq, G11, G14, and G16 (G15 and G16 are murine and human orthologs, respectively; Table 9.9).
Table 9.9
Properties of the Gq family members (Source: [790]; RIC8: resistance to inhibitors of cholinesterase-8 homolog that serve as Gα protein guanine nucleotide-exchange factor). Regulator of G-protein signaling associated with Gαq belongs to the B/R4 subfamily (RGS1–RGS5, RGS8, RGS13, RGS16, and RGS18). Among them, RGS2, RGS3, RGS4, and RGS18 exhibit GAP activity for Gαq subunits. Members of the Gq/11 and G12/13 subclasses activate RhoA, but via distinct transmission pathways from RhoGEFs to RhoA. Whereas members of the G12/13 subclass use components of the RhoGEF1 subfamily (RhoGEF1, RhoGEF11, and RhoGEF12), members of the Gq/11 subclass target RhoGEF11, RhoGEF12, RhoGEF13 and RhoGEF25 proteins. In addition, Resistance to inhibitors of cholinesterase-8 homolog RIC8a has GEF activity for Gi1, Go, Gq, and G12, but not Gs, whereas RIC8b homolog interacts with Gs and Gq, but not Gi and G12. Scaffold caveolin-1 in calveolae allows efficient Gq–receptor coupling. Lipid raft resident proteins, flotillin-1 and -2, also interact with Gq proteins.
Type | Distribution | Effectors | Partners |
---|---|---|---|
Gq | Ubiquitous | PLCβ1/3/4 | RIC8a/b, caveolin-1, |
RGS, AKAP13, | flotillin-1/2 | ||
RhoGEF11/12/25 | |||
G11 | Ubiquitous | PLCβ1/3/4 | RIC8a/b, caveolin-1, |
RGS, RhoGEF25 | flotillin-1/2 | ||
G14 | Lung, kidney, | PLCβ1/3/4 | |
liver | |||
G16 | Hematopoietic | PLCβ1/2/3 | |
cells |
Gα q
Once G protein-coupled receptors are activated by ligand binding, Gαq releases GDP, binds GTP, and dissociates from Gβγ. Subunits Gαq GTP and Gβγ target phospholipase-Cβ and phosphatidylinositol 3-kinase, respectively. Phospholipase-Cβ catalyzes the hydrolysis of phosphatidylinositol bisphosphate to release inositol trisphosphate and diacylglycerol. Second messengers IP3 and DAG in turn cause calcium influx from intracellular stores via IP3Rs and stimulate protein kinase-C, respectively. Signaling pathways independent of PLCβ are also primed, as Gq-based cue activates Rho guanine nucleotide-exchange factors and interacts with G-protein-coupled receptor kinases [789].
Therefore, owing to Ca
entry into the cytosol, Gαq contributes to the regulation of cell contractility and secretion, especially the control of smooth muscle cell tone in blood vessel and bronchus walls via calcium-dependent activation of myosin light chain kinase that phosphorylates (activates) myosin light chain to heighten actomyosin ( stress fiber)-based contractility with Gα11 as well as membrane fusion of secretory vesicles, particularly in endocrine cells and platelets. In neurons, Gαq modulates the synaptic transmission. In addition, Gαq is involved in pre- and postnatal cardiomyocyte growth, craniofacial development, and phototransduction [789].

Intrinsic GTPase activity of the Gαq subunit hydrolyzes Gαq GTP to Gαq GDP, thereby terminating the signal and allowing the heterotrimeric G-protein to reassemble and take its quiescent state. However, the GTPase activity of isolated G proteins is much lower than that of G proteins in the pool of cellular molecules.
Phospholipase-Cβ (negative feedback) and regulators of G-protein signaling act as GTPase-activating proteins for subunits of the Gαq subclass [791]. Inhibitors of Gαq signaling include RGS1 to RGS5, RGS8, RGS13, RGS16 to RGS19 [789]. In addition to GTPase activation, these RGSs act as effector antagonists, as they are able to reduce PLCβ activity induced by Gαq subunit.
The inhibition by regulators of G-protein signaling on G-protein-mediated signaling can actually be exerted: (1) by serving as GTPase-activating proteins (GAP) for Gα subunits of heterotrimeric G proteins, thereby accelerating G-protein inactivation (GAP mechanism of action); or (2) by blocking Gα-mediated signaling, as they compete by binding to Gα effectors (effector antagonism). Among the members of the R4 RGS subfamily that have the shortest N- and C-terminal flanking regions among the RGS family members and interact with Gq and Gi/o subunits, RGS2 and RGS3 operate mainly by effector antagonism, whereas RGS5 and RGS16 inactivate G proteins [792].
Ubiquitously expressed Gαq undergoes reversible palmitoylation at adjacent cysteines in its N-terminus (Cys9 and Cys10) that enables its localization to the cytoplasmic face of the plasma membrane. In addition, receptor activation promotes palmitate turnover on Gα subunits [789].
Stimulated G-protein-coupled receptors are able to activate Ras–MAPK cascades, albeit with smaller efficacy than that of receptor Tyr kinases. Monomeric GTPases of the RHO superfamily can transmit signaling from certain activated GPCRs. Subunit Gαq can induce activation of small GTPase RhoA, though with a lower potency than that of members of the Gα12 family. Most of the GPCRs that prime Rho-dependent responses, such as GPCRs of bombesin, endothelin, thrombin, lysophosphatidic acid, and thromboxane-A2, as well as calcium-sensing receptor that stimulates stress fibers, focal adhesions, and cell rounding, activate Gq or G12/13 subclass protein. Activation of RhoA mediated by Gαq relies on member of the Rho-specific GEF family, such as RhoGEF11 to RhoGEF13 and RhoGEF25 [793].17
Another group of Gαq interactors corresponds to G-protein-coupled receptor kinases, such as GRK2 and GRK3, that phosphorylate activated Gαq, hence blocking Gαq-mediated signaling and recruit arrestins and other proteins that can desensitize GPCRs [789].
Once bound to angiotensin-2, AT1 receptor triggers multiple signaling axes, particularly those mediated by G proteins (Gαi, Gαq [mainly], and Gα12 ∕ 13) and by β-arrestin, in addition to receptor Tyr kinase transactivation.18 Different types of GPCR agonists can engage distinct signaling pathways (ligand type-dependent signaling), such as: (1) G-protein- and β-arrestin-dependent and (2) G-protein-independent and β-arrestin-dependent activation of ERK1 and ERK2 kinases.19 Both Tyr4 and Phe8 of octapeptide angiotensin-2 are essential for both AT1–ligand binding and G-protein-dependent signaling, i.e., ensure the structure–function relationship for angiotensin-2 and its analogs. Substitution of these residues at position 4 and/or 8 in angiotensin-2 indeed produces agonists with different functional outcomes. These β-arrestin-biased ligands actually trigger β-arrestin-selective signaling downstream from AT1, instead of supporting IP3 production and PKC activation [795]. Different βArr–AT1 complexes resulting from distinct angiotensin-2 analogs, i.e., different changes in AT1 conformation initiated by the analogs and in β-arrestin conformation upon recruitment to AT1 receptor as well as possible selective actions of GRKs (GRK2 and GRK6),20 which can cause β-arrestin-dependent, selective engagement of adaptors and/or signaling effectors, create different activated states and, hence, cellular outcomes ( vascular smooth muscle cell growth or migration) [795]. Efficiency of ERK1 and ERK2 activation is correlated to the stability of endosomal βArr–AT1 complex, which depends on the nature of the ligand–receptor complex, rather than to the extent of β-arrestin recruitment to AT1 receptor [795].21 The propensity of different analogs to promote distinct β-arrestin recruitment and conformational changes depends or not on phosphorylation of AT1 by GRK kinases. Once β2-adrenoceptor is stimulated, ligand promotes phosphorylation of distinct sites by different GRKs and distinctively influences the conformation of β-arrestin. Therefore, different β-arrestin conformations generate distinct signaling types and cell responses.
Gα 11
Widespread Gα11 subunit interacts with multiple G-protein-coupled receptors, such as muscarinic, adrenergic, endothelin, bombesin, cholecystokinin receptors, among others. Gα11 GTP binds to its effectors and regulators, such as phospholipase-Cβ, G-protein-coupled receptor kinases, and regulators of G-protein signaling.
In cardiomyocytes, RGS2 precludes Gq/11-mediated phospholipase-C activation, but not Gq/11-mediated ERK1, ERK2, P38MAPK, and JNK activation downstream from endothelin receptor [796]. In addition, RGS4 inhibits Gq-mediated activation of mitogen-activated protein kinase and phospholipase-C downstream from bombesin receptor [797].
Subunit Gα11 activates phospholipase-Cβ to generate the following effects [791]: (1) stimulation of transcription factor tubby-like protein, as PIP2 tethers tubby-like protein to the plasma membrane; (2) hydrolysis of PIP2 into inositol trisphosphate and diacylglycerol; (3) IP3-triggered Ca
release from its intracellular store; (4) DAG activation of Unc13 (or Munc13) in hyperglycemia that causes apoptosis; and (5) DAG activation of protein kinase-C.

Subunits of the Gq/11 subclass can also stimulate the mitogen-activated protein kinase cascade. Vascular endothelial growth factor binds to its cognate receptors on vascular endothelial cells (VEGFR1–VEGFR2). Receptor VEGFR2 triggers signaling, whereas VEGFR1 represses VEGFR2-initiated effect. Both receptors stimulate G proteins. Receptor VEGFR2 activates Gq/11 that target small GTPase RhoA and the MAPK module and provokes Ca
influx via phospholipase-C [798]. On the other hand, VEGFR1 activates small GTPase CDC42. Constitutively active G11 mutant (but not constitutively active Gq mutant) can cause phosphorylation of both VEGFR2 and MAPK. In addition, Gβ/γ subunit intervenes in MAPK phosphorylation and intracellular Ca
mobilization, but does not influence VEGFR2 phosphorylation.


Gα 16
Protein Gα16 is a subunit of the Gq subclass that activates phospholipase-Cβ (PLCβ1–PLCβ4) [799]. This effector enzyme hydrolyzes PIP2, thereby releasing inositol trisphosphate that provoke intracellular Ca
flux and diacylglycerol messenger that activate protein kinase-C.

Protein Gα16 is expressed predominately in hematopoietic cell types. It is coupled to most GPCRs. It is a substrate of 2 types of GTPase-activating proteins (Gα16GAP): PLCβ and many regulators of G-protein signaling [799].
9.1.1.4 Gα 12 Subclass
Members of the subclass 4 of Gα subunits, i.e., G12 and G13, participate in various cellular functions. They activate Rho GTPases. Both Gα12 and Gα13 trigger similar cellular effects that lead to gene transcription, reorganization of the actin cytoskeleton, formation of actin stress fibers, and assembly of focal adhesions, events that are associated with RhoA activation [800]. Nonetheless, Gα12 and Gα13 do not have only redundant activities. Subunit Gα13, but not Gα12, contributes to the development of the vasculature. Factors RhoGEF1 and RhoGEF12 are more effective for G13 than G12. Whereas Gα13 is able to stimulate RhoGEF1 and unphosphorylated RhoGEF12 in vitro, Gα12 only activates phosphorylated RhoGEF12 [800]. Whereas Gα12 activates RhoA for stress-fiber formation upon activation by G-protein-coupled receptors bound to endothelin, thrombin, and vasopressin, Gα13 primes RhoA signaling as a mediator of GPCRs ligated by bradykinin, serotonin, and lisophosphatidic acid. Consequently, Gα12 and Gα13 can be targeted by distinct receptors. They not only possess different affectors, but also different effectors. Furthermore, they can have opposite effect on a given mediator. Subunit G13 activates the transcription factor NRF2, whereas G12 antagonizes it [800]. Besides, G13 is involved in the regulation of NOS2 expression.
Subunits G12 and G13 not only target small GTPase Rho via RhoGEF, but also phospholipase-Cε and -D, MAPK, and Na + –H + exchanger, as G12 and G13 interact with several proteins (Table 9.10). Ezrin, radixin, and moesin connect various receptors, ion channels, and integrins to the cytoskeleton. Four Rho GTPase guanine nucleotide-exchange factors (Sect. 9.4.1.9) are regulated by G12/13 (Table 9.11): (1) RhoGEF1,22 which is produced in blood cells; (2) RhoGEF11,23 which is widespread, but at low level, except in the central nervous system; (3) ubiquitous RhoGEF12;24 and (4) widespread RhoGEF13.25
Table 9.10
Interactors of G12 and G13 proteins (Source: [801]; AKAP: A-kinase anchoring protein; CTK: cytosolic Tyr kinase; HSP: heat shock protein; PP5: protein phosphatase; RasGAP: GTPase-activating protein of Ras; RhoGEF: guanine nucleotide-exchange factor of Rho).
Protein | Function |
---|---|
Cadherin | G12 and G13-mediated release of β-catenin |
Radixin | G13-induced linkage between cytokeletal and plasmalemmal proteins |
HSP90 | G12-induced actin reorganization |
(stress fiber formation) | |
AKAP | G12-mediated stimulation of AKAP13 (especially in heart) |
G13 interaction with AKAP3 in testis and PKA stimulation | |
CTK | G12-mediated activation of BTK |
G13-mediated activation of FAK2 | |
Gi- and Gs-mediated activation of Src | |
PP5 | G12 and G13-mediated activation |
RasGAP | G12-mediated stimulation |
RhoGEF | G13-mediated stimulation of RhoGEF1 (p115RhoGEF) |
G12-mediated stimulation of RhoGEF12 (LARG) |
Table 9.11
G12/13-regulated Rho guanine nucleotide-exchange factors (Source: [802]; AKAP-LBC: A-kinase anchoring protein-lymphoid blast crisis, a cardiac splice variant of LBC-RhoGEF [AKAP13 or RhoGEF13]; FAK: focal adhesion kinase; PAK: P21-activated kinase; PKA(C): protein kinase-A(C); TEC: Tyr kinase expressed in hepatocellular carcinoma). G12 and G13 prime the RhoGEF–RhoA–RoCK pathway. They cause RhoGEF translocation from the cytosol to the plasma membrane and stimulate their GEF activity. Whereas RhoGEF oligomers are inactive, active monomers bind to activated G12/13 at the plasma membrane. Conversely, these RhoGEFs serve as GTPase-activating proteins for G12/13 (negative feedback). Targets of RhoA include Rho kinase (RoCK) that: (1) provokes cell contraction, as it inhibits myosin light chain phosphatase, and (2) initiates serum response factor-dependent gene transcription.
RhoGEF | Effect | GEF Activity regulators |
---|---|---|
RhoGEF1 | G13-induced GEF activation | PKC ( + ) |
(p115RhoGEF) | PAK1 ( − via Rac) | |
RhoGEF11 | G13-induced GEF activation | TEC, FAK ( + ) |
(PDZ-RhoGEF) | ||
RhoGEF12 | G12-induced GEF activation | TEC, FAK ( + ) |
(LARG) | G13-induced GEF activation | |
RhoGEF13 | G12-induced GEF activation | PAK1 ( − ) |
(LBC-RhoGEF) | G13-induced GEF activation | |
(AKAP13) |
Gα 12
Ubiquitous Gα12 protein transmits signals from G-protein-coupled receptors to RhoGEFs to stimulate small Rho GTPase. Unlike G13, G12 does not activate Rho via RhoGEF1, despite its interaction with RhoGEF1 [803]. Protein Gα12 also connects to regulators of G-protein signaling with GTPase-activating protein activity that regulate its functioning. It mainly targets small GTPase RhoA, thereby stimulating its effectors in a Rho-dependent manner. However, it also directly binds and activates certain signaling effectors, such as Bruton’s Tyr kinase, protein phosphatase-2, small Ras GTPase, and N- and E-cadherin [803]. In addition, it also links to Ras(Rap)GAP RasA226 Furthermore, G12 synergistically regulates cell responses with other Gα proteins. Both G12 and G13 cooperate with Gs to integrate cAMP synthesis via adenylate cyclase AC7 [803].
Protein Gα12 can stimulate the activity of Na + –H + exchangers via the Ras–PKC pathway [803]. It contributes to cell proliferation, particularly the G1–S transition via both Ras and Rac small GTPases. It activates the JNK pathways also via Ras, Rac, or CDC42 GTPase, as well as MAP3K1 and MAP3K5 for cell apoptosis.
An active form of G12 binds to the cytoplasmic domain of cadherins to release β-catenin that has an inhibitory effect on intercellular interactions and can then stimulate cell migration [803]. Subunit Gα12 is required for the S1P-induced migration of vascular smooth muscle cells that depends on phospholipase-C It also connects to tight junction protein zonula occludens-1, together with heat shock protein HSP90 and Src kinase [803]. It then disrupts tight junctions. Moreover, G12 or G13 heightens the activity of Rho–Rac-dependent activator protein AP1 that provoke synthesis of transforming growth factor-β1.
In migrating cells, G12 generates backward signals to activate myosin-2 of contractile actin–myosin complexes formed at cell trailing region via Rho small GTPase for a forward motion [803]. Conversely, G12 can impede cell migration, as it can inhibit α2β1-integrin.
Subunit Gα12 binds GDP very tightly. The estimated rate of guanine nucleotide-exchange for G12 is very slow, i.e., 10- to 20-fold slower than that for Gs and Go, but similar to that of Gz and G13 [803]. The GTP hydrolysis rate of Gα12 is very slow, i.e., comparable to that of Gz and G13, but is 5 to 40 times slower than that of other Gα subunits. Kinetics of GTP–GDP exchange are accelerated by GEFs. The duration of Gα12 signaling is controlled by the GAP activity of RGS domain of RhoGEFs, such as RhoGEF1 and RhoGEF12 [803].
Protein Gα12 undergoes post-translational modifications. Palmitoylation determines its subcellular localization, especially to membrane rafts via HSP90, and interaction with other proteins [803]. Phosphorylation by PKCα, PKCβ, PKCδ, PKCε, and PKCζ reduces its affinity for Gβγ subunit.
Subunit Gα12 couples to cognate GPCRs of angiotensin-2 (AT1), cholecystokinin (CCK1), endothelin (ET A and ET B ), galanin, acetylcholine (M1 and M3), peptidase-activated or thrombin (PAR1, PAR2, and PAR4), lysophosphatidic acid and sphingosine 1-phosphate (S1P2 to S1P5), thromboxane-A2, thyroid-stimulating hormone, vasopressin, lysophosphatidylcholine, and formylpeptide, as well as calcium-sensing receptor [803].
Gα 13
Activity of Gα13 is modulated by Rho-specific GEFs (RhoGEF1, RhoGEF11, and RhoGEF12). Subunit G13 also bears post-translational modification, such as acylation and phosphorylation. Protein kinase-A phosphorylates G13 (Thr203) [800]. In response to thrombin or thromboxane-A2, protein kinase-C also phosphorylates G13 subunit.
Subunit Gα13 couples to endothelin ET A and ET B , lysophosphatidic acid, peptidase-activated, sphingosine 1-phosphate, thromboxane-A2, and thyrotropin receptors, as well as angiotensin receptor-1A, cholecystokinin receptor-A, dopamine receptor-D5, galanin receptor-2, neurokinin (substance P) receptor NK1, and serotonin receptor-4 [800].
9.1.2 Set of Gβ Subunits
Heterotrimeric guanine nucleotide-binding proteins that transmit signals from G-protein-coupled receptors to effectors comprise a Gβ subunit. Subunits Gβ1 to Gβ4 are expressed ubiquitously, whereas Gβ5 subunit is produced only in the brain.
The combinatorial linkage of individual subtypes of Gα, Gβ, and Gγ subunits contributes to G-protein specificity for GPCRs. In a cell that contains many different types of Gα, Gβ, and Gγ subunits, only some types are able to heterotrimerize because of differences in affinity of these subunit types for one another.
In general, Gβγ inhibits GTP binding to Gα. Dimer Gβγ can interact with protein kinase-D, G-protein-coupled receptor kinases, Bruton Tyr kinase, several components of the Raf–Ras–MAPK cascade, RhoGEF18 for monomeric GTPases RhoA and Rac1, adaptors Src homology-2 domain-containing protein SHC and kinase suppressor of Ras KSR1, retinal phosducin, calmodulin, membrane integrated qSNARE protein syntaxin-1B and synaptosome-associated protein SNAP25B, tubulin, and dynamin-1, as well as voltage-dependent calcium channels [804]. The G-protein-coupled receptor kinase β-adrenoceptor kinase impedes binding of Gβγto cRaf [805].
9.1.3 Set of Gγ Subunits
Heterotrimeric G proteins contains a Gγ subunit that is irreversibly tethered to a Gβ subunit with variable affinity. Twelve known Gγ subunit types exist. Many, but not all, Gγ subtypes can associate with Gβ protein.
Like G-protein-α subunit that undergoes myristoylation and palmitoylation, Gβγ dimers are covalently modified by lipids for membrane anchorage and specific proteic interactions. Members of Gγ subclass experience prenylation (farnesylation or geranylgeranylation) [806]. The activity of Gβγ is increased by reversible carboxymethylation of Gγ, whereas the nature of isoprenylation (farnesyl or geranylgeranyl group) determines the membrane affinity of Gβγ permanently (irreversible post-translational modification) [804].
Subunits Gγ show greater variation in tissue distribution than Gβ subunits. Subunit Gγ1 is specific to rod photoreceptors; Gγ2 and Gγ7 are present in several tissues, yet they are enriched in brain; Gγ3 and Gγ4 are brain-specific; and Gγ5 is ubiquitous [807]. Subunit Gγ8 could function in chemosensory transduction, both in olfactory and vomeronasal neurons [804]. Subunit Gγ1 interacts poorly with Gβ2 and Gβ3, but fails to bind to Gβ4 and Gβ5; Gγ2 links poorly to Gβ3; Gγ3 binds well with all Gβsubunits; Gγ4 tethers especially well with Gβ5 (preferential interaction with Gβ1, Gβ2, and Gβ5); Gγ5 and Gγ7 favor interactions with Gβ1 and Gβ2 subunits [807]. Subunit Gγ8 is coexpressed with Gβ1 and forms a stable dimer [804].
Gβγ (especially Gβ1γ2 and Gβ2γ2) effectors include adenylate cyclases, phospholipase-Cβ1, -Cβ2, and -Cβ3, cGMP phosphodiesterases, and ion channels.
9.1.4 G Protein and Myogenic Response
Local blood flow is regulated to match the metabolic demand of peripheral tissues. Small-resistance arteries possess the intrinsic property to constrict in response to a rise in intraluminal pressure (Bayliss effect ). Vascular tone determines local vascular resistance and hence organ perfusion. This pressure-induced myogenic vasoconstriction does not depend on endothelium functioning. Myogenic responsiveness actually is an inherent property of vascular smooth muscle cell that can be further fine-tuned by endothelial and neurohumoral factors. Although increased intravascular pressure causes depolarization of arterial myocyte membrane that activates voltage-dependent CaV1 channel, the latter does not influence pressure-induced depolarization, which thus implies another stretch-activated ion channel.
The myogenic response triggered by mechanosensors in vascular smooth muscle cells involves ligand-independent activation of Gq/11-protein-coupled receptors rather than membrane stretch-induced gating of mechanosensitive transient receptor potential ion channels [808]. Mechanically activated receptors adopt an active conformation that allows G-protein coupling and recruitment of β-arrestin. Activated Gq/11 protein signals hastens phospholipase-C that stimulates transient receptor potential channel via DAG, thus leading to membrane depolarization and finally to pressure-induced myogenic vasoconstriction.
9.2 Regulators of Heterotrimeric G Proteins
Heterotrimeric G proteins are activated by a GPCR-induced GDP–GTP exchange at the Gα subunit. This activation is followed by regulation of specific effectors, such as adenylate cyclase, phospholipase-Cβ, kinases, and ion channels. Such a process is controlled by various regulators.
9.2.1 Nucleoside Diphosphate Kinases
Nucleoside diphosphate kinases (NDPK) form proteic complexes, supply nucleoside triphosphates, and catalyze the transfer of phosphate between nucleoside triphosphates and nucleoside diphosphates. In humans, the NDPK family includes 9 known members (mainly NDPKa–NDPKc). They are receptor-independent activators of G-protein. Phosphate conjugation mediated by NDPK acts as an alternative mechanism to the GPCR-induced GDP–GTP exchange. Phosphate is transferred via a plasma membrane-associated complex of NDPKb (but not NDPKa) and Gβγ dimers.
Receptor-independent activation of G proteins via the NDPKb–Gβγ complex requires the intermediate phosphorylation of Gβ subunits [809]. Enzyme NDPKb hence acts firstly as a histidine kinase for Gβ subunits. The phosphate can then be transferred onto GDP. The greater the number of Gαs–Gβγ–NDPKb complexes, the stronger the NDPKb–Gβγ-mediated phosphotransfer to adenylate cyclase-regulating Gαs and Gαi subunits. Conversely, NDPKb depletion leads to strongly reduced amounts of Gβ1γ2 dimers as well as caveolin-1 and -3 [810]. Therefore, caveolins that connect to NDPKb and Gβγ subunits contribute to the regulation of plasmalemmal G-protein content.
In cardiomyocytes, activation of Gs and Gi via GPCRs regulates intracellular cAMP concentration that controls myocardial contractility via stimulation of protein kinase-A and changes in Ca
transients via L-type Ca
channels. β-Adrenoceptor activation can regulate plasmalemmal NDPK content, hence Gαs activity that causes cAMP synthesis [809]. Conversely, the higher the NDPK and Gi levels and density of Gαiβγ–NDPKb complexes, the lower the cAMP concentration and contractility.


9.2.2 Regulators of G-Protein Signaling
The interactions of GPCR, G protein, and effectors also involve fine-tuned regulators of G-protein signaling proteins (RGS), which accelerate GTP hydrolysis by Gα subunits. Regulators of G-protein signaling act as GTPase-activating proteins (Fig. 9.3). They thus inhibit signal transduction mediated by Gi/o and Gq/11 (Tables 9.12 and 9.13).

Fig. 9.3
G-protein, its components, its activation–deactivation, and regulators of G-protein signaling proteins (RGS)
Members of the family of small RGSs are encoded by different genes and share an RGS homology domain. However, their specific activities in GPCR signaling is associated with another structural domain. The smaller RGS proteins likely function as GαGAPs, whereas the larger RGS proteins and RGS-like proteins are Gα effectors. Moreover, certain RGS proteins are involved in the assembly of signaling complexes.
Table 9.12
G-protein-coupled receptors in the cardiovascular system, associated G proteins, regulators (inhibitors) of G-protein signaling, and targets that most often experience a reduced activity, as RGSs terminate G-protein activation by enhancing GTP hydrolysis (Part 1; Source: [779]; AT1: angiotensin-2 receptor; ET X : any endothelin receptor type (X: A or B); IP: inositol phosphate; MAPK: mitogen-activated protein kinase; S1P i : type-i sphingosine 1-phosphate receptor). Receptors ET A and ET B augment and reduce heart contractility, respectively. Receptors ET A and NOS-coupled ET B cause a vasoconstriction and -dilation, respectively. The RGS proteins discriminate not only Gα types, but also GPCRs coupled to the same Gα type to selectively regulate signaling.
GPCR | Gα | RGS | Targets |
---|---|---|---|
AT1 | Gq/11 (Gi) | RGS1/2/3l/4 | MAPK |
RGS5 | MAPK | ||
RGS5 | IP | ||
RGS2 | cAMP | ||
RGS2 | IP | ||
RGS4/5 | Ca ![]() | ||
ET A | Gq/11 (Gi) | RGS3l/4 | MAPK |
RGS4/5 | Ca ![]() | ||
ET X | RGS2/4 | MAPK | |
RGS2/3/4/5 | IP | ||
S1P1 | Gi | RGS1/3l/4 | MAPK |
S1P2 | Gi, Gq/11, G12/13 | RGS1/3l/4 | MAPK |
S1P3 | Gi, Gq/11, G12/13 | RGS1/2/3l | MAPK |
Table 9.13
G-protein-coupled receptors in the cardiovascular system, associated G proteins, regulators (inhibitors) of G-protein signaling, and targets that most often experience a reduced activity, as RGSs terminate G-protein activation by enhancing GTP hydrolysis (Part 2; Source: [779]; AR: adrenergic receptor; GIRK: G-protein inwardly rectifying K + channel; IP: inositol phosphate; M i : type-i muscarinic receptor; MAPK: mitogen-activated protein kinase).
GPCR | Gα | RGS | Targets |
---|---|---|---|
α1aAR | Gq/11 | RGS2 | IP |
α1bAR | Gq/11 | IP | |
α1xAR | RGS2/4 | MAPK | |
RGS2 | MAPK; JNK ( + ) | ||
RGS2/3/4/5 | IP | ||
RGS2 | MAPK | ||
β 2AR | Gs (Gi) | RGS2 | cAMP |
M1 | Gq/11 | RGS2/3/8 | Ca ![]() |
RGS2/8 | IP | ||
RGS2/16 | IP | ||
RGS2/5 | IP, Ca ![]() | ||
M2 | Gi | RGS1/3/4 | GIRK |
RGS3/3s/4/5/16 | MAPK | ||
RGS2/3/3s/4/5/16 | PKB | ||
RGS3/4/5 | cAMP | ||
M3 | Gq/11 | RGS3 | MAPK |
RGS2/3/8 | Ca ![]() | ||
RGS2/8 | IP | ||
RGS2/3/5/16 | IP | ||
RGS2/3 | IP | ||
RGS2/3/4 | Ca ![]() | ||
RGS2/3/3s/5 | MAPK | ||
RGS3/3s | PKB | ||
M5 | Gq/11 | RGS2/3/8 | Ca ![]() |
RGS2/8 | IP |
9.2.2.1 Superfamily of RGS-Box-Containing Proteins
In humans, the RGS superfamily contains at least 37 identified proteins. It can be decomposed into many families [811]: (1) A or RZ family with members (RGS17, RGS19, and RGS20) that are characterized by an N-terminus with many cysteine residues, which can be reversibly palmitoylated; (2) B or R4 family (RGS1–RGS5, RGS8, RGS13, RGS16, RGS18, and RGS21); (3) C or R7 family, members (RGS6, RGS7, RGS9, and RGS11) of which can couple Gα subunits to GPCRs in the absence of Gβγ dimers. They indeed bind Gβ5 via Gγ-like (GGl) domain to form heterodimers;27 (4) D or R12 family (RGS10, RGS12, RGS14); (5) E or RA family that is composed of RGS box-containing axin1 and axin2, negative regulators of the Wnt pathway, that interact with adenomatous polyposis coli protein; (6) F or GEF family that comprises RhoA-specific guanine nucleotide exchange factors (RhoGEF1, RhoGEF11, and RhoGEF12); (7) G or GRK family that encompasses RGS box-containing G-protein-coupled receptor kinases (GRK1–GRK7; Sect. 5.2.11); and (8) H or Snx family that contains RGS box-containing sorting nexins (Snx13, Snx14, Snx25). Other RGS groups include [811]: (1) dual-specificity A-kinase anchor proteins AKAP10a and AKAP10b that bind PKA regulatory subunits and (2) RGS22 isoforms (RGS22a–RGS22c).
Most RGS proteins are GPCR inhibitors, either by their GAP activity for Gα or by effector antagonism, as they can bind activated GαGTP in competition with effectors. On the other hand, members of the F class of RGS proteins are positive regulators; RhoGEF1, RhoGEF11, and RhoGEF12 (Sect. 9.4.1.9) couple Gαq, Gα12, and/or Gα13 subunits to monomeric RhoA GTPase (Sect. 9.3.13.1). Owing to their RGS box, they convert inactive RhoAGDP into active RhoAGTP. These 3 RGS box-containing, RhoA-specific guanine nucleotide-exchange factors (RGS-RhoGEFs) serve as Gα effectors that couple not only GPCRs, but also semaphorin receptors to RhoA GTPases [811].28 The RGS box of RhoGEF1 serves as a GTPase-activating protein motif Gα12 and Gα13 subunits. Protein RhoGEF12 is a Gα-responsive RhoGEF for Gq, G12 and G13 [811]. However, RhoGEF12 stimulation by Gα12 GTP depends on RhoGEF12 phosphorylation by TEC family kinases or focal adhesion kinase. In addition, these 3 RGS-RhoGEFs couple distinct receptors to RhoA activation (RGS-RhoGEF signaling specificity).29
The RGS proteins with a Gαi∕o–Loco interaction (GoLoco or G-protein regulatory [GPR]) motif, such as RGS12 and RGS14, have a guanine nucleotide-dissociation inhibitor activity, as they slow spontaneous exchange of GDP for GTP and inhibit association with Gβγ subunits (sequestration) [811]. GoLoco motif-containing proteins generally bind to GαGDP subunits of the Gi/o subclass. Other GoLoco motif-containing proteins include Rap1GAP (Sect. 9.4.2.3) that accelerates GTP hydrolysis by Rap1 and Rap2-interacting protein (Sect. 9.3.9), as well as G-protein signaling modulators GPSM1,30 GPSM2,31 GPSM3,32 which is able to simultaneously bind more than one Gαi1 subunit, and GPSM4.33 The GPSM proteins do not modify kinetics or magnitude of effector activation, but can reduce agonist binding affinity, hence signaling intensity [811].
In summary, regulators of G-protein signaling serve as: (1) guanosine triphosphate-accelerating protein for Gα proteins (GαGAP), hence as signaling inhibitors; (2) Gα effectors, such as RGS-RhoGEFs that couple receptors to monomeric GTPase RhoA; (3) signaling scaffolds between signaling mediators, such as RGS12 that operates as a nexus between Gα, small guanosine triphosphatase Ras, and protein kinases; (4) coupling factors between Gα and GPCRs, such as dimers composed of C-class RGSs and Gβ5; and (5) guanine nucleotide-dissociation inhibitors (e.g., RGS12 and RGS14).
9.2.2.2 Regulation of Activity
Activity of RGS proteins is regulated within a cell. Phosphatidylinositol (1,4,5)-trisphosphate inhibits RGS proteins [812]. Ca
–calmodulin restores the GAP activity. Both PIP3 and Ca
–calmodulin bind to the same RGS site. Inhibition and disinhibition of GAP activity of RGS4 by these molecules explain oscillations in intracellular calcium concentration.34 Furthermore, RGS proteins are targeted by different protein kinases.35


The RGS proteins are implicated in interactions involving various signaling molecules associated with G-protein signaling pathways and ion carriers at the plasma membrane, such as Ca
, phospholipids (especially phosphoinositides), and Tyr kinases.36 Besides, lipopolysaccharides and angiotensin-2 increase the expression of RGS proteins in vascular cells.

9.2.2.3 RGS Isoforms
RGS1
In humans, RGS1 is identified in heart, aorta, lung, and olfactory bulb, as well as B lymphoblasts, T lymphocytes, dentritic cells, and peripheral blood monocytes. Isoform RGS1 is a GTPase-activating protein for Gi/o and Gq subunits [813]. It interacts with spinophilin and neurabin to lower GPCR-mediated signaling and counteract RGS inhibition, respectively.
RGS3
Ubiquitous RGS3 is a GTPase-activating protein for Gi and Gq subclass members. Alternative splicing generates long (RGS3L) and short (RGS3S) forms, in addition to PDZRGS3 and C2PARGS3 isoforms [813].
Binding of 14-3-3ζ and 14-3-3τ proteins to RGS3 hampers RGS3 activity, whereas Ca
influx through voltage-gated channels activates RGS3 [813]. It interacts with: (1) SMAD2 to SMAD4 transcription factors and interferes with their heteromerization; (2) Na + -dependent inorganic phosphate cotransporters SLC17a1 and SLC34a1; (3) type-1 membrane neuroligins of the postsynaptic membrane, ligands of presynaptical β-neurexins; and (4) EPHb2 and mGlu1A receptors.

9.2.2.4 Cardiac RGSs
Cardiac RGSs hinder phospholipase-C activity via Gq/11, especially phospholipase-C stimulation of endothelin-1.40 Hence, RGS4 has an antihypertrophic effect. Furthermore, cardiac RGSs regulates the activation and deactivation kinetics of Gβγ-gated K + channels, thus the inward rectifier K + channel regulated by acetylcholine via Gi/o (RGSs accelerate GTP hydrolysis rate of Gαi∕o and regulate ACh-dependent relaxation).41
Membrane-attached RGS3, a Gβγ-binding protein [815] that is strongly expressed in the heart [816], attenuates signaling not only via Gαi and Gαq ∕ 11, but also via Gβγ-mediated signaling (via phospholipase-C, mitogen-activated protein kinase, and phosphatidylinositol 3-kinase). Agent RGS6 is relatively abundant in atriomyocytes [817]. Sorting nexin SNx13,42 a Gαs-specific GAP, inhibits adenylate cyclase stimulation induced by the α-adrenoceptor–Gαs complex. It may then modulate the activity of cardiac Ca
channels. It binds to membrane phosphatidylinositol 3-phosphate, thus participating in early endosome structure. The RhoGEF proteins have Dbl- (DH) and pleckstrin (PH) homology domains. The DH domain is responsible for exchange activity and the PH domain is likely involved in subcellular localization. The RGS-like RhoGEFs act on Gα12 and Gα13 [815]. Coupling of GPCRs to G12/13 in cardiomyocytes is associated with contractility [818], and protein kinase-C–mediated activation of sarcolemmal Na + –H + exchangers [819]. GPCR kinases phosphorylate activated GPCR receptors. Agent GRK2, highly expressed in the human heart (as well as GRK5 and GRK6), interacts with Gq/11 (GRK2 sequesters activated Gαq).

9.2.3 Gα Guanine Nucleotide-Exchange Factors
Resistance to inhibitors of cholinesterase-8 homolog-A (RIC8a)43 acts as a guanine nucleotide-exchange factor for a subset of Gα proteins activated by ligand-bound G-protein-coupled receptors that include Gαi1 to i3, Gαo, Gαq, and Gα13 [820]. It also associates with Gαs [251]. Protein RIC8a connects to GαGDP proteins, stimulates GDP release, and forms a nucleotide-free, transition-state complex with Gα that dissociates upon GTP binding to Gα. In addition, RIC8a binds adenylate cyclase-5 that is highly expressed in the brain striatum and heart. It suppresses AC5 activity upon Gi stimulation [821].44 Therefore, RIC8a potentiates Gα-mediated signaling. During Gq-mediated signal transduction, RIC8a enhances ERK activation [822].
9.2.4 Activators of G-Protein Signaling
Activators of G-protein signaling (AGS) exert distinct effects depending on the G-protein activation–deactivation cycle. They can be classified into 3 distinct functional sets: (1) AGSs that function as direct Gα activators, as they operate as guanine nucleotide-exchange factors; (2) AGSs that bind to Gα, serving as guanine nucleotide dissociation inhibitors; and (3) AGSs that bind to Gβγ that either dissociate the inactive heterotrimer or sequester released Gβγ upon G-protein activation [823] (Table 9.14). Accessory AGS proteins affect the signal features and propagation. In addition, AGSs can facilitate crosstalk between signaling pathways.
Table 9.14
Receptor-independent activators of G-protein signaling (Source: [823]). Activated G-protein-coupled receptors intervene as guanine nucleotide exchange factors (GEF) that trigger the transformation of GαGDP into GαGTP and the dissociation of Gα from Gβγ for signaling. Upon GTP hydrolysis, the heterotrimer then reforms. The stages of G-protein activation–deactivation cycle are regulated to optimize signal magnitude and duration, as well as to keep signal specificity. Inhibitors of G-protein activation are either regulators of G-protein signaling, which act as GTPase-activating proteins (GAP) by enhancing GTP hydrolysis, or guanine dissociation inhibitors (GDI), which inhibit GDP dissociation. Stimulators of G proteins can operate as GEFs for Gα. The AGS subfamily 1 includes AGS1, subfamily-2 AGS3 to AGS6 (AGS5 and AGS6 targeting only Gi3), and subfamily-3 AGS2, AGS7, and AGS8.
Type | Target | Effect |
---|---|---|
AGS1 | Gi2, Gi3 | GEF |
AGS2 | Gi2, Gi3, Gs, G16 | Gβγ binding |
AGS3 | Gi2, Gi3 | GDI |
9.2.4.1 AGS1
Activator of G-protein signaling AGS1 selectively activates Gi/o independently of G-protein-coupled receptors. Protein AGS1 is a member of the RAS hyperfamily of small GTPases that plays the role of guanine nucleotide-exchange factor for Gi subunit. Protein AGS1 can antagonize GPCR by reducing the pool of G-proteins available for GPCR coupling.
In addition, AGS1 impedes increase in activity of Gβγ-regulated inwardly rectifying K + channel (GIRK) activated by muscarinic M2 receptors [824]. In neurons, AGS1 interacts with adaptors, such as NOS-associated CaPON and NCK2, as well as nitric oxide synthase NOS1. Adaptor CaPON competes with adaptor DLg4 involved in the NMDAGlu–NOS1 pathway to form a NMDAGlu–CaPON–NOS1–AGS1 complex [825]. Moreover, AGS1 is expressed in the suprachiasmatic nuclei according to circadian rhythm [826].
9.2.4.2 AGS2
Activator of G-protein signaling AGS2 is similar to a light chain component of cytoskeletal and ciliary dynein. AGS2 interacts with numerous signaling effectors, especially GPCRs. Dynein could organize signaling complexes to regulate organelle displacement. AGS8 is produced by ventricular cardiomyocytes (but not in cardiac fibroblasts, aortic smooth muscle cells, and endothelial cells) in response to hypoxia (but not by tachycardia, hypertrophy, or failure) [827]. AGS8 interacts directly with Gβγ without disturbing the regulation of PLCβ2 by Gβγ.
9.2.4.3 AGS3
Activator of G-protein signaling AGS3, which is widely expressed (with tissue-specific splicing),45 acts on plasmalemmal concentrations of certain receptors and channels by modulating cellular transport of receptors and channels such asKIR2.1. Moreover, AGS3 activates G protein (Gi inhibition and Gβγ stimulation) in a receptor-independent fashion [828]. AGS3 interacts only with members of the Gαi∕o subclass (except Gz), i.e., with both Gi and Go, but it is a guanine dissociation inhibitor for Gi3 only (not for Go) [829]. Agent AGS3 binds and stabilizes Gαi GDP, and then blocks reassociation of Gαi with Gβγ dimer. Therefore, AGS3 inhibits Gαi, but favors Gβγ signaling.
9.2.5 GPCR-Interacting Proteins
Many GPCRs interact with GPCR-interacting proteins (GIP). These proteins are involved in [779]: (1) GPCR compartmentation in membrane rafts and/or caveolae; (2) assembling of large signaling complexes to direct signaling specificity, intensity, and duration; (3) transfer to and from the plasma membrane; and (4) fine-tuning of GPCR signaling.
Some GIPs are transmembrane proteins, whereas others are intracellular. Certain GIPs can interact with several receptors, such as arrestin and G-protein-coupled receptor kinases, whereas other interact specifically with a single type of GPCR, such as AT1 receptor-associated protein (ATRAP).46
Several GPCR-interacting proteins, such as Homer proteins, Gα-interacting protein (GAIP)-interacting protein C-terminus GIPC1,47 14-3-3 proteins, calmodulin, and spinophilin,48 link GPCRs to receptors, ion channels, cytoskeletal proteins, protein kinases, and regulators of G-protein signaling [779].
Several GPCR–GIP interactions are enhanced by ligand stimulation, such as connections between GPCRs and arrestins, GRKs, Na + –H + exchanger regulatory factor NHERF1 [779].49
Among GPCR-interacting proteins, arrestin causes GPCR desensitization via endocytosis. In addition, arrestins are scaffold proteins for Src kinase and several components of MAPK modules, such as cRaf, extracellular signal-regulated kinases, and Jun N-terminal kinase JNK3 [779].
G-protein-coupled receptor kinases also interact with numerous proteins involved in signaling and trafficking, such as Gq/11, Gβγ, PI3K, PKB, MAP2K1, calmodulin, clathrin, caveolin, and actin.
A-kinase anchoring proteins AKAP5 and AKAP12 that link to adrenoceptors also interact with kinases PKA and PKC as well as PP3 phosphatase. Upon stimulation of adrenoceptors, PKA anchored to AKAP5 phosphorylates the receptor that then switches from Gs to Gi coupling and promotes MAPK signaling [779]. It also phosphorylates GRK2 to heighten adrenoceptor desensitization.
9.2.6 G-Protein-Coupled Receptor Kinases
G-protein-coupled receptor kinases (GRK) can complex with heterotrimeric G protein on the inner surface of the plasma membrane. Interactions of G proteins, in particular Gβ1γ2 subunit, with the lipid bilayer facilitate GPCR-catalyzed GTP exchange on the Gα subunit. Ubiquitous G-protein-coupled receptor kinases constitute a 7-member family (GRK1–GRK7; Vol. 3 – Chap. 7. G-Protein-Coupled Receptors and Sect. 5.2.11). These kinases have different distribution patterns among the body’s tissues as well as distinct binding preferences for some receptors.
Subunit Gβ1γ2 links to phospholipase-Cβ and G-protein-regulated inwardly rectifying potassium channels (GIRK or KIR3), as well as GRK2 kinase [830]. Kinase GRK2 can simultaneously interact with activated Gαq and Gβγ, GPCRs, and the plasma membrane. Therefore, GRK2 enables the assembly and organization of signaling complexes at sites of GPCRs. Subunit Gβ1γ2 changes its orientation after binding to GRK2 protein recruited to the plasma membrane by G proteins to phosphorylate activated GPCRs [830].
9.3 Monomeric (Small) GTPases
Guanosine triphosphatases form a set of hydrolases that process guanosine triphosphate. This set comprises both large and small GTPases that operate in signal transduction, protein synthesis, intracellular transport of vesicles, translocation of proteins across cellular membranes, and organization of the cytoskeleton and cell adhesion plaques, hence cell growth, division, differentiation, spreading, and migration. Many small GTPases are dynamically acylated to modify their membrane affinity.
Monomeric, cytosolic, regulatory G proteins constitute a class of approximately 150 members that can be split to form the RAS hyperfamily ( ∼ 36 members). The RAS hyperfamily can be decomposed into the ARF, RAB, and RAS superfamilies, the latter being subdivided into the DIRAS, NKIRAS, RAL, RAN, RAP, RAS, RHO ( ∼ 22 members), RGK, RHEB, and RIT families (Table 9.15).
Table 9.15
Superfamilies of the class of monomeric GTPases.
Category | Subcategories |
---|---|
Members | |
ARF superfamily | |
ARF family | Arf1–Arf6 |
ARL family | |
ARFRP family | |
SAR family | |
RAB superfamily | |
14 families | Rab1–Rab35 |
RAS superfamily | |
DIRAS family | DIRas1–DIRas3 |
NKIRAS family | |
RAL family | RalA–RalB |
RAN family | |
RAP family | Rap1–Rap2 |
RAS family | P21RAS subfamily (hRas, kRas, nRas) |
RRAS subfamily (rRas1, rRas2, mRas/rRas3) | |
RASD subfamily (RasD1–RasD2) | |
RAS-like subfamily | |
RASL10 subfamily (RasL10a–RasL10b) | |
RASL11 subfamily (RasL11a–RasL11b) | |
RASL12 subfamily | |
RASL13 subfamily | |
RERG subfamily | |
RGK family | |
RHEB family | |
RIN/RIT family | RIN subfamily |
RIT subfamily | |
RHO family | CDC42 subfamily |
MIRO subfamily (Miro1–Miro2 [RhoT1–RhoT2]) | |
RAC subfamily (Rac1–Rac3, and RhoG) | |
RHO subfamily (RhoA–RhoC) | |
RHOBTB subfamily (RhoBTB1–RhoBTB2) | |
RHOD/RIF subfamily | |
RHOF subfamily | |
RHOH/TTF subfamily | |
RND subfamily (Rnd1, Rnd2, Rnd3/RhoE) | |
TC10 subfamily (RhoJ–RhoQ) | |
WRCH subfamily (RhoU–RhoV) |
Among large GTPases, dynamins act in endocytosis, organelle division, and cytokinesis. Small GTPases share common features with G proteins, but have different structure and mechanism of action. Monomeric, small GTPases can be categorized according to their functions (Tables 9.16 and 9.17).
Table 9.16
Main functions of small GTPases (Part 1; ARF: adpribosylation factors; CDC42: cell-division cycle-42; PIKE: phosphatidylinositol 3-kinase enhancer). PIKE is a nuclear GTPase with PLCγ1 as guanine nucleotide-exchange factor.
Type | Role |
---|---|
ARF | Control of cell transfer of molecules |
(vesicular transport — vesicular budding and maturation), | |
phosphoinositide metabolism | |
CDC42 | Control of cytoskeleton dynamics, |
formation of filopodia and adhesive-like complexes, | |
activation of the MAPK module and gene expression, | |
initiation of DNA synthesis, regulation of G1 phase progression | |
Gem | Nucleocytoplasmic transport |
Miro | Mitochondrial fusion and transport |
PIKE | Activation of nuclear PI3K activity |
Table 9.17
Main functions of small GTPases (Part 2; Rab: Ras from brain; Rad: Ras associated with diabetes; Ran: Ras-related nuclear proteins; Rap: Ras-related proteins; Ras: rat sarcoma viral proto-oncogene product homolog; RHEB: Ras homolog enriched in brain; Rho: Ras homology; Rit: Ras-like expressed in many tissues).
Type | Role |
---|---|
Rab | Control of vesicle transport, membrane trafficking |
(clathrin-coated vesicle formation, endosomal motility) | |
Rac | Control of cytoskeleton, |
formation of filopodia and adhesive-like complexes, | |
activation of the MAPK module and gene expression, | |
initiation of DNA synthesis, regulation of G1 phase progression | |
Rad | Nucleocytoplasmic shuttling, cardiomyocyte growth |
cardiac excitation-contraction coupling | |
Ran | Control of nucleocytoplasmic transport, |
especially during phases G1, S, and G2 of the cell cycle, | |
and microtubule organization during M phase | |
Rap | Vesicular transport |
Ras | Control of cell differentiation, adhesion, growth, proliferation, |
migration, and apoptosis | |
Rem | Nucleocytoplasmic shuttling, |
muscular excitation–contraction coupling, | |
interaction with ion channels and 14-3-3 proteins | |
RHEB | Cell growth (TOR pathway) |
Rho | Control of cytoskeleton dynamics and integrin activity, |
formation of stress fibers and focal adhesion complexes, | |
initiation of DNA synthesis, activation of gene expression, | |
regulation of progression of cell cycle G1 phase | |
RIN | Neuron development and trophicity |
RIT | Neuron differentiation, axonal and dendritic growth |
The RAS hyperfamily of small GTPases includes 5 major superfamilies — ARF, RAB, RAN, RAS, and RHO — based on sequence homology and specific functions. Members of the RAS superfamily regulate cytoskeletal rearrangement and contraction and are activated upon stimulation of G-protein-coupled and growth factor receptor Tyr kinases.
The ARF superfamily of adpribosylation factors is implicated in the regulation of intracellular vesicle genesis and motion.
The RAB superfamily is composed of about 70 types in humans that regulate vesicle formation, displacement along actin and tubulin filaments, and fusion.
The RAN superfamily of Ras-related nuclear GTPases is involved in nuclear export and import.
The RAS superfamily of small GTPases comprises the DIRAS, RAL, RAP, RAN, RAS, RASD, RAS-like, RGK, RHEB, NKIRAS, and RIT families.
The RAS family embodies 2 subfamilies: the P21RAS subfamily with hRas, kRas, and nRas, and the RRAS subfamily with rRas1, rRas2 (or TC21), and mRas (or rRas3).
The RAS-like family contains the RASL10 (with RasL10a, or RRP22, and RasL10b); RASL11 (with RasL11a and RasL11b); RASL12 (with RasL12);52 and RASL13 (with RasL13)53 subfamilies, as well as Ras-like, estrogen-regulated, growth inhibitor RERG.
The DIRAS family of RAS-like GTPases groups DIRas1 (a.k.a. Rig) to DIRas3 that operate in intracellular protein transport, including nucleocytoplasmic transfer.
The family of NFκB inhibitor-interacting Ras-like proteins comprises NκIRas1 and NκIRas2.
The RGK family consists of Rad, Gem/Kir, and Rem.
The RHO superfamily of small Ras homolog GTPases encompasses the 3 main families, i.e., CDC42, Rac, and Rho families. In fact, it is made up of many families [831]: family 1 with CDC42; 2 with Miro1 and Miro2; 3 with Rac1,54 Rac2,55 Rac3, and RhoG; 4 with RhoA, RhoB, and RhoC; 5 with RhoD; 6 with RhoF;56 7 with RhoH; 8 with RhoJ57 and RhoQ;58 9 with RhoU59 and RhoV;60 10 with RhoBTB1 and RhoBTB2; and 11 with Rnd1 to Rnd3.61
Among the 22 identified members of the RHO superfamily, most are classically activated (i.e., cycle between active GTP-bound and inactive GDP-loaded forms), but 8 are atypical Rho GTPases. Classical Rho GTPases correspond to CDC42, RAC, RHO, RHOD, and RHOF families. Atypical Rho GTPases comprise RhoBTB, RhoH, RhoJ, RhoQ, RhoU, RhoV, and Rnd GTPases. These GTPases are either predominantly GTP-bound (e.g., Rnd and RhoH) or have an increased nucleotide exchange (e.g., RhoU2).
Proteins of the RHO, RAC, and CDC42 families of the RHO superfamily intervene in the reorganization of the actin cytoskeleton as mediators of extracellular signals for formation of stress bundles and focal adhesions (RhoA), membrane ruffles and lamellipodia (Rac1), and filopodia or microspikes (CDC42) [832]. They have additional roles. Members of the RHO family operate during progression through G1 phase of the cell division cycle. Transcription activation by serum response factor in response to lysophosphatic acid requires RhoA GTPase. Proteins CDC42 and Rac1 are able to induce sequential phosphorylations (activation) of Jun N-terminal kinases. Small Rac GTPases participate in the activation of NADPH oxidases in neutrophils and macrophages.
Because small GTPases of the RHO superfamily of the RAS hyperfamily activate various mediators (actin nucleators, protein kinases, phospholipases, and scaffold proteins), they can regulate the cytoskeleton dynamics and thus act in various cell activities that require changes in the cell cytoskeleton, such as intracellular vesicle transport and cell polarity, shape, adhesion, motion, division, and differentiation. About 70 Rho GTPase effectors have been identified in addition to the high number of regulators (Table 9.18).
Table 9.18
Some effectors of small GTPases CDC42, Rac1, and RhoA and corresponding effects (Source: [833]; Dia: diaphanous formin; IQGAP: IQ motif-containing GTPase-activating protein; MRCK: myotonic dystrophy kinase-related CDC42-binding kinase nWASP: neural Wiskott-Aldrich syndrome protein P67PhOx: P67 phagocyte oxidase protein; PAK: P21-activated kinase; Par6: partitioning defective-6; PKN: protein kinase-N; PLC: phospholipase-C; RoCK: Rho-associated coiled-coil-containing protein kinase SRA1: specifically Rac1-associated protein-1; WAVe: WASP family verprolin-homologous protein). CDC42 is involved in organelle positioning; Rac1 in cytoskeleton polarization, microtubule stabilization, formation of tight junctions and cell protrusions; RhoA in formation of actin stress fibers and focal adhesions, membrane retraction, and microfilament contraction.
Effector | Effect |
---|---|
Small GTPase CDC42 | |
IQGAP | Intercellular adhesion, capture of microtubule |
MRCK | Actomyosin-based contraction, kinase |
nWASP | Actin polymerization |
PAK | Actin polymerization, microtubule stabilization, kinase |
Par6 | Cell polarity |
Small GTPase Rac1 | |
IQGAP | Intercellular adhesion, capture of microtubule |
P67PhOx | NADPH oxidation |
PAK | Actin polymerization, microtubule stabilization, kinase |
Par6 | Cell polarity |
SRA1 | Actin polymerization |
WAVe | Actin polymerization |
Small GTPase RhoA | |
Citron | Cytokinesis, kinase |
PKN | Cell cycle, kinase |
PLC | Cell signaling |
RoCK | Factin stabilization, kinase |
Dia | Actin polymerization, microtubule stabilization |
During cell migration, Rho GTPases drive membrane protrusion at the leading edge and contractility of the cell body. Rho GTPases indeed regulate the assembly of filamentous actin (Factin) in response to signaling. Their effectors induce the assembly of contractile actin–myosin filaments ( stress fibers in particular) and integrin-containing focal adhesions. Consequently, small Rho GTPases act in vascular processes, such as smooth muscle cell (SMC) contraction, cell adhesion, endothelial permeability, leukocyte extravasation, platelet activation, and migration of smooth muscle cells and endothelial cells involved in angiogenesis and wall remodeling [834].
Small Rho GTPases can be activated by: (1) G-protein-coupled receptors,62 (2) Tyr kinase receptors, and (3) cytokine receptors. In particular, growth factors recruit Rho, Rac, and ERM.63 Activation of Rho GTPases increases their membrane-associated level and decreases their cytosolic concentration.
Members of the RHO superfamily, such as Rac and CDC42, activate the JNK pathway. Activated JNKs, in turn, phosphorylate (activate) transcription factors Jun and ELk1 that boost transcription from genes with AP1 and SRE reponse elements, respectively. Activated RhoA does not act on JNK, but targets the transcription factor serum response factor that cooperates with ELk1 and Fos to stimulate transcription from promoters containing SRE elements. In addition, Rho, Rac, and CDC42 target the transcription factor nuclear factor-κB.
Small Rho GTPases are involved in vascular disorders associated with pathological remodeling and altered cell contractility. Kinase RoCK, an effector of the small Rho GTPase, is involved in atherosclerosis as well as in post-stenting restenosis. Ezrin, radixin, and moesin of the ERM family are phosphorylated by RoCK [836]. Small Rho GTPase also controls other cellular activities [837]. Small Rho GTPase regulates several enzymes involved in phospholipid metabolism (phospholipase-D and phosphatidylinositol kinase). It controlsdelayed rectifier K + channels.
The activation–inactivation cycle of Rho GTPases is a regulated process. Activation of GTPases into a GTP-bound conformation is controlled by specific guanine nucleotide-exchange factors (GEF; Sect. 9.4.1). Conversely, GTP is hydrolyzed to GDP by GTPase in combination with GTPase-activating proteins (GAP; Sect. 9.4.2). In the absence of signaling, the major fraction of RhoGDP GTPases is sequestered in the cytosol by guanine nucleotide-dissociation inhibitors (GDI; Sect. 9.4.3). In mammals, about 70 RhoGEFs, 60 RhoGAPs, and 3 RhoGDIs have been detected. Certain members of the RHO superfamily are able to regulate activity of other members. Small CDC42 GTPase can activate Rac1; Rac1 can inhibit RhoA GTPase.
Activity of small GTPases Ras and Rho is not only regulated by GEFs, GAPs, and GDIs for Rho GTPases, but also by post-translational modification such as isoprenylation by farnesyl and geranylgeranyl transferases to promote membrane anchorage and subsequent effector association.
Except Ran GTPases, all of small GTPases of the RAS superfamily are post-translationally modified. Small ARF GTPases are myristoylated, Rab GTPases and members of the RAS and RHO superfamilies attach 1 or 2 farnesyl or geranylgeranyl groups. Conversely, Rab and Rho GTPases are sequestered in the cytosol by GDIs that extract GTPasesGDP from the membrane by binding to their prenylated motifs [832].
9.3.1 Superfamily of ARF GTPases
Adpribosylation factors64 (ARF) constitute a superfamily of ubiquitous proteins of the RAS hyperfamily of small GTPases. These myristoylated GTP–GDP switch proteins are engaged in the regulation of vesicular transport in cells. Their activity is controlled by a cycle of successive GTP binding and hydrolysis, i.e., a cycle of activation and inactivation, using ARF guanine nucleotide-exchange factors and GTPase-activating proteins, respectively. They localize to cellular membranes (plasma membrane as well as membranes of the secretory vesicles, endosomes, and lysosomes).
Small ARF GTPases recruit coat proteins for cargo sorting, enzymes to adapt the lipid composition of membranes (e.g., phosphatidylinositol kinases), and cytoskeletal components for motion (Tables 9.19 and 9.20). They interact with regulators of other guanine nucleotide-binding proteins and form molecular platforms constituted by ARFGDP, GEFs, GAPs, and effectors [838]. They can act simultaneously or successively at membranes of the endoplasmic reticulum and Golgi body and at the plasma membrane. Regulators of the ARF network also integrate ARF activities with other GTPase signaling axes. Both ArfGEFs and ArfGAPs can serve as scaffolding effectors, recruiting signaling mediators and promoting conformational changes for appropriate binding and activation.
Table 9.19
Partners and interactors of ARF GTPases. The ARFs regulators recruit: (1) coat proteins that support sorting of cargo into vesicles; (2) enzymes that change lipid composition of membranes; and cytoskeletal factors (Part 1; Source: [838] ERGIC: endoplasmic reticulum–Golgi intermediate compartment; AP: adaptor proteic complex; BBSome: Bardet-Biedl syndrome coat complex (transport of membrane proteins into cilium); CerT: ceramide-transfer protein; CoP: coatomer protein; GGA: Golgi-localized, γ-ear-containing, ARF-binding protein; GCC: GRIP and coiled-coil domain-containing protein; GMAP: Golgi-associated microtubule-binding protein; PLD, phospholipase D; PI4K: phosphatidylinositol 4-kinase; PI4P5K: phosphatidylinositol 4-phosphate 5-kinase). The Golgi-associated retrograde protein (GARP) complex, or vacuolar protein sorting (VPS)-53 (fifty-three [VFT]) tetramer (VPS51–VPS54) is required for the fusion of early and late endodomes with the trans-Golgi network. Pleckstrin homology (PH) domain-containing family-A member PlekHa8, a phosphoinositol 4-phosphate adaptor, also called four-phosphate adaptor protein FAPP2, binds phosphatidylinositol 4-phosphate and ARF1 GTPase and serves as glucosylceramide-transfer protein.
Effector | ARF | Location |
---|---|---|
Coat complexes | ||
AP1 | ARF1/3 | Endosomes, trans-Golgi network |
AP3 | ARF1/3 | Endosomes, trans-Golgi network |
AP4 | ARF1/3 | Trans-Golgi network |
BBSome | ARL6 | Plasma membrane |
CoP1 | ARF1/3 | Golgi body, ERGIC |
CoP2 | SAR1 | Endoplasmic reticulum exit |
GGA1/2/3 | ARF1/3 | Endosomes, trans-Golgi network |
Lipid-processing enzymes | ||
CerT | ARF1 | Golgi body |
PlekHa3/8 | ARF1 | Golgi body |
PI4K | ARF1 | Golgi body |
PI4P5K | ARF1 | Plasma membrane |
PLD | ARF1–6, ARL1 | Plasma membrane |
Tethers | ||
Exocyst | ARF6 | Plasma membrane |
GARP | ARL1 | Endosomes, trans-Golgi network |
GMAP210 | ARF1 | Cis-Golgi network |
Golgin-A1/4, GCC1/2 | ARL1 | trans-Golgi network |
Table 9.20
Partners and interactors of ARF GTPases (Part 2; Source: [838]; arfaptin: ARF-interacting protein; BART: binding partner binder of ARF-like protein-2 (two; ARL2); FIP: family of Rab11-interacting protein; MAPK8IP: MAPK8-interacting protein; NDPKa: nucleoside diphosphate kinase-A; PDE: phosphodiesterase; RhoGAP: Rho GTPase-activating protein; SCoC: short coiled-coil protein; Unc: uncoordinated).
Effector | ARF | Location |
---|---|---|
Regulators | ||
Cytohesin | ARF6, ARL4 | Plasma membrane |
RhoGAP21 | ARF1/6 | Golgi body, plasma membrane |
Scaffolds | ||
FIP3/4 | ARF5/6 | Recycling endosomes |
MAPK8IP3/4 | ARF6 | Endosomes |
Tubulin folding chaperone | ||
β-Tubulin cofactor-D | ARL2 | Cytosol |
Miscellaneous | ||
Arfaptin1/2 | ARF1, ARL1 | Golgi body, trans-Golgi network |
BART2 | ARL2 | Mitochondria, nucleus |
NDPKa | ARF6 | Plasma membrane, cell junctions |
PDE6δ | ARL2/3 | |
SCoC | ARL1 | Golgi body |
Unc119 | ARL2/3 |
9.3.1.1 Families of the ARF Superfamily
The superfamily of ARF GTPases includes several families constitued by: (1) ARF isoforms (ARF1–ARF6);65 (2) secretion-associated and Ras-related protein SAR1; (3) ARF-related protein ArfRP1;66 (4) ARF-like GTPases (ARL1–ARL22);67 and (5) adpribosylation factor domain-containing protein-1 (ARD1 or ARFD1) that corresponds to tripartite motif-containing protein TriM23,68 a ubiquitin-protein ligase and a GTP-binding protein.69
Small ARF GTPases can be classified into 3 subfamilies: subfamily 1 with ARF1 to ARF3 that regulate the assembly of coat complexes onto budding vesicles and activate enzymes that target lipids; subfamily 2 with ARF4 and ARF5 that may operate in Golgi transport and recruitment of coat components to trans-Golgi membranes; and subfamily 3 with ARF6 that controls endosomal-membrane trafficking. Isoform ARF2 is absent in humans [839].
9.3.1.2 ARF Compartmentation and Function
Adpribosylation factors are characterized by their subcellular compartmentation and binding partners that dictates the function of each member.70 However, the majority of ARF effectors can interact with several ARF GTPases. All ARF GTPases are myristoylated (second N-terminus Gly), thus allowing ARF tethering to membranes. Activation–deactivation cycle of ARFs, hence association–dissociation of transport vesicle coat proteins, are regulated by ArfGEFs (Sect. 9.4.1.1) and ArfGAPs (Sect. 9.4.2.1).
The primary role of ARF GTPases is the regulation of vesicular membrane transfer, especially the formation of coated vesicles. The ARF proteins actually regulate structure, budding from donor membrane, motion, and fusion with acceptor membrane of vesicles devoted to intracellular transport of cargos, as they recruit coat proteins, prime proteic complex assembly, modulate phospholipid metabolism, contribute to actin remodeling, especially at the cell cortex, and participate in some signaling pathways. Therefore, ARF GTPases are involved in endo- and exocytosis, phagocytosis, cytokinesis, and cell adhesion and migration. Small ARF GTPases not only regulate vesicular motions, but also organelle structure.
Small ARF GTPases execute their function by anchoring to membrane surfaces, where they interact with other proteins to initiate budding and maturation of transport vesicles. They are mainly involved in the vesicular transport of lipids and proteins between the endoplasmic reticulum, where cargos are synthesized, and the Golgi body, where cargos undergo post-translational modification (Table 9.21 and 9.22).
Table 9.21
Monomeric ARF and ARL GTPases in the secretory pathway (Source: [838]; ERGIC: endoplasmic reticulum–Golgi intermediate compartment; ADRP: adipose differentiation-related protein, or adipophilin; AP: adaptor protein complex; ATGL: adipose triglyceride lipase; CAPS: calcium-dependent activator protein for secretion; CoP: coat protein of the coatomer complex; GGA: Golgi-localized, γ-ear-containing, ARF-binding protein PI4K: phosphatidylinositol 4-kinase).
Site | Involved molecules |
---|---|
Endoplasmic reticulum | SAR1, |
Sec12 (SAR1GEF), Sec23–Sec24 (SAR1GAP), | |
CoP2 | |
ERGIC | ARF1, |
GBF1 (ARF1GEF), | |
CoP1, CoP2, ADRP, ATGL (formation of lipid droplets) | |
Cis-Golgi network | ARF1, ARF4, |
GBF1 (recruited by Rab1 and PI4P), | |
Cert, PlekHa8 (lipid transport) | |
PI4K | |
Medial-Golgi network | |
Trans-Golgi network | ARF3 |
Trans-Golgi network | ARF3 (constitutive exocytosis), |
ARF4, ARF5 (regulated secretion), | |
ARL1 (recruitment of golgins and arfaptin), | |
BIG1, BIG2 (ArfGEF; recruited by PDE3a), | |
AP1, CoP1, GGA, (endosome and lysosome), | |
CAPS (regulated secretion) |
Table 9.22
Small ARF and ARL GTPases at the plasma membrane and in endocytosis (Source: [838]; IPCEF: interaction protein for cytohesin exchange factor).
Type | Involved partners and role |
---|---|
ARF1 | Clathrin-independent endocytosis |
ARF6 | Cortical actin cytoskeleton structuring, |
endosomal membrane recycling, | |
Rac, DOCK1, IPCEF (protrusion), | |
AP2, clathrin (endocytosis), | |
PI4P5K, PLD (clathrin-coated pit formation, | |
sorting endodome), | |
MAPK8IP4 (microtubule motor adaptor for | |
rapid recycling), | |
exocyst | |
RhoGEF24, Rac, RhoG (actin dynamics), | |
cytohesin (cell adhesion assembling), | |
BRAG2, Rac, SOS1 (adherens junction disassembly) | |
ARL2 | Regulation of microtubule-based motion, |
ELMOD2 | |
ARL3 | Ciliogenesis, intraciliary transport |
RP2 | |
ARL4 | Clathrin-independent endocytosis, |
cytohesin | |
ARL6 | Ciliogenesis, intraciliary transport |
(BBS3) | BBSome |
ARL8 | Late endosome (fusion with lysosome) |
ARL13 | Ciliogenesis, intraciliary transport |
Small ARF GTPases contribute to morphology and location of the Golgi body that depends on the cell cytoskeleton as well as actin cytoskeleton-dependent changes in cell morphology. Golgi-associated ARF1 is involved in the formation of focal adhesions via the delivery of adaptor paxillin to the plasma membrane and Rho activation. Small ARF6 GTPase facilitates recruitment of active Rac to the plasma membrane. Small Rac GTPase alone can promote the migration of epithelial cell sheets during wound healing, but not cell scattering that requires cooperation of ARF6 [841].
9.3.1.3 ARF Structure
The ARF proteins possess a 2-domain structure [842]. The myristoylated N-terminal helix used for membrane binding71 is separated from the C-terminus by a flexible linker. This linker may yield a certain degree of adaptability in binding modes for the huge quantity of ARF-interacting proteins, in addition to specific binding sites for lipids on some of these small GTPases.
Exchange of GDP by GTP occurs at the ARF myristoyl binding site. Active ARFs promote and stabilize curved surfaces of budding vesicles [842]. C-terminus positioned on the membrane surface provides an interaction structure for activators, adaptors, effectors, and inhibitory GAP proteins. At least certain ARF types (e.g., ARF4 to ARF6) remain bound to membranes in their GDP-bound conformation.
9.3.1.4 ARF Partners
To function, ARFs must interact sequentially or simultaneously with: (1) guanine nucleotide-exchange factors and lipids that catalyze their activation; (2) proteic adaptors that modulate recruitment of cargo into nascent buds; (3) lipid-modifying enzymes; and (4) GTPase-activating proteins, the 2 latter categories being required for budding and maturation of transport carriers.
Partners of ARFs can be grouped into 3 categories: (1) ArfGEFs that have a Sec7 domain (Sect. 9.4.1.1);72 (2) ArfGAPs that possess a cysteine-rich ArfGAP domain (Sect. 9.4.2.1);73 and (3) effectors and ARF-binding lipids, either non-specific partners that promote nucleotide-exchange, or specific partners, such as PI(4,5)P2 and phosphatidic acid [839]. Several GEFs and GAPs interact with ARF proteins. Proteins of the ArfGEF and ArfGAP sets, like ARF, can translocate to membranes in a regulated fashion. Nucleotide-sensitive partners comprise mitotic kinesin-like protein MKlP1, arfaptin-1 and -2, and arfophilins.74 In addition to arfophilins, JNK-interacting proteins JIP3 and JIP4 (MAPK8IP3 and MAPK8IP4) that also selectively bind ARF6 mediate postendocytic recycling during cytokinesis. All ARF GTPases share several effectors, such as phospholipase-D and several phosphoinositide kinases (PI4P5Kα–PI4P5Kγ).
9.3.1.5 ARF GTPase Isoforms
Soluble ARFs (ARF1–ARF5) are regulators of vesicular transport, particularly to and from the Golgi body, and endosomes, as well as activators of phospholipase-D. Active GTPARF1 to GTPARF5 translocate onto membranes where they recruit: (1) ARF-dependent coat proteins AP1, AP3, AP4, GGA13, and CoP1; (2) scaffolding and trafficking Munc18-interactors MInt1 (a.k.a. amyloid-β A4 precursor-binding protein APBa1 and neuron-specific adaptor X11α), MInt2 (a.k.a. APBa2, X11β, and X11-like protein [X11l]), and MInt3 (a.k.a. APBa3, X11γ, and X11-like-2 protein X11l2);75 and (3) lipid enzymes, such as phospholipase-D, phosphatidylinositol 4-kinase-3, and phosphatidylinositol 4-phosphate 5-kinase [844].
ARF1
Protein ARF1 regulates the membrane association of clathrin-associated adaptor proteic complex AP1 and AP3 used for trans-Golgi network (TGN)-to-endosome and endosome-to-lysosome transport. In its active state, it recruits coat and adaptor proteins, such as clathrin, AP1, and AP3 to the membrane and induces the assembly and nucleation of coated vesicles.
In addition to its myristoylated N-terminus that promotes membrane tethering, inactive, cytosolic ARF1GDP attaches to membranes of the Golgi body and endoplasmic reticulum owing to transmembrane trafficking proteins, transmembrane EMP24 domain-containing trafficking proteins TMED2, TMED3, and TMED1076 on the one hand and membrin, a vesicular fusion mediator SNARE as well as a component of CoP1-mediated coats on the other [840].
Proteins GBF1 and BIG2 (Sect. 9.4.1.1), GEFs for ARF1, stimulate ARF1 in the cis-Golgi network. Upon activation, ARF1GTP dissociates from transmembrane Golgi cargo receptors TMEDs, thereby yielding binding sites for coatomers that can then prime vesicle coat assembly [840]. Active ARF1GTP indeed recruits pre-assembled heptameric CoP1 coatomer complex and controls, together with coatomers, vesicle budding. CoP1-coated vesicles mediate: (1) retrograde transport from the cis-Golgi network to the endoplasmic reticulum77 and (2) retrograde and possibly anterograde, transport between Golgi cisternae. Active ARF1 is also involved in clathrin-coated vesicle formation at the trans-Golgi network for exocytosis to the plasma membrane or cell organelle membranes. Clathrin-coated vesicles transport cargos from the trans-Golgi network. Active ARF1GTP promotes recruitment of heterotetrameric adaptor proteic complexes AP1, AP3, and AP4, as well as monomeric Golgi-localized γ-ear-containing ARF-binding proteins (GGA) from the cytosol onto membranes. Active ARF1 also favors assembly of spectrin and actin on Golgi membranes.
Conversely, ArfGAP1 bound to ligand-coupled KDEL (Lys–Asp–Glu–Leu)-containing receptors and packaged into budding vesicles is activated by coatomers and membrane curvature. It then hydrolizes GTP on ARF1, thereby dissociating CoP1 from vesicles and eliciting cargo packaging, as CoP1-coated vesicles are depleted of cargo. Whereas GTP hydrolysis leads to CoP1-coated vesicle uncoating, it is not sufficient to induce clathrin-mediated coat disassembly [840].
Activated ARF1 can control Golgi membrane–spectrin connection that contributes to the maintenance of Golgi body organization via phosphatidylinositol (4,5)-bisphosphate levels on Golgi membranes, as it causes activation of type-3 phosphatidylinositol 4-kinase78 and type-1 phosphatidylinositol 4-phosphate 5-kinase [840].79 Protein kinase-D is also recruited to the trans-Golgi network, where it causes the fission of vesicles. All PKD isoforms (PKD1–PKD3) phosphorylate PI4K3β.
Protein ARF1 promotes actin assembly on Golgi membranes that favors vesicular budding, as it facilitates ARP2/3-dependent actin polymerization via CDC42 and its effector nWASP [840].80 In addition, ARF1 regulates exocytosis from the Golgi body via the cortactin–dynamin-2 complex. Cortactin and dynamin-2 are involved in actin remodeling and stabilization via the ARP2–ARP3 actin-nucleating complex and vesicle scission, respectively.
ARF3
Adpribosylation factor-3 interacts with adpribosylation factor-interacting proteins-1 (ARFIP1) and -2 (ARFIP2); Golgi-associated, γ-adaptin ear-containing, ARF-binding proteins GCA1 to GCA3; and kinesin-like protein-5 (a.k.a. KiF23 and mitotic kinesin-like protein MKLP1) [251].
ARF4 or ARF2
Adpribosylation factor-4 that belongs to the ARF subfamily-2 interacts with epidermal growth factor receptor [251].
ARF5
Adpribosylation factor-5 interacts with adpribosylation factor-interacting proteins-1 (ARFIP1) and -2 (ARFIP2) as well as β1 subunit of the adaptor-related protein complex-3 (AP3β1) [251].
ARF6
Whereas ARF1 is involved in Golgi membrane dynamics, ARF6 localizes to the plasma membrane and endosomes, where it regulates endocytic membrane trafficking and molecule sorting in the absence of adaptor complex AP2 and clathrin, as well as actin remodeling at the cell cortex via Rac1 GTPase and phospholipids [840]. In vitro, ARF6 binds directly to the β1 subunit of the AP1 complex and both the β3 and δ subunits of the AP3 complex, but not to the AP2 complex [847]. However, ARF6 stimulates the local activity of PI(4)P5K1γ, thereby promoting AP2 assembly at the membrane. In vitro, ARF6 may recruit the AP2 (but not AP1) complex onto liposomal membranes.
Protein ARF6 regulates clathrin-dependent and -independent endocytosis, endosome recycling, and actin reorganization. It operates in highly dynamical events at the cell surface, such as phagocytosis, between-cell adhesion, and cell migration.
In cell migration, ARF6 promotes the activation of Rac necessary for the formation of lamellipodia at the leading edge and mediates the recycling of integrins [847].
Protein ARF6 activates phosphatidylinositol 4-phosphate 5-kinase-1γ and phospholipase-D. The former synthesizes PI(4,5)P2 that regulates clathrin-mediated endocytosis. The latter produces phosphatidic acid, a cofactor in PI(4)P5K activation [840].
Protein ARF6 also regulates clathrin- and caveola-independent endocytosis of various subtances such as major histocompatibility complex class-1 proteins and peripheral myelin-membrane protein PMP22 [840]. Activated ARF6 also favors arrestin dissociation to facilitate internalization of G-protein-coupled receptors. All these molecules are delivered to sorting endosomes with possible plasmalemmal recycling that needs phospholipase-D.
Among transfer of G-protein-coupled receptors, ARF6 controls endocytosis of β-adrenoceptor, angiotensin AT1, vasopressin V2, and endothelin receptors. It also supports neurotransmitter-triggered internalization of AMPA-type glutamate receptors [847].
Small ARF6 GTPase also regulates postendocytic transport (e.g., transferrin recycling) [847]. Several transmembrane and glycosyl-phosphatidylinositol (GPI)-anchored cargos enter an ARF6 + endosomes (e.g., β1-integrins, MHC class-1 molecules, complement regulator CD59,81 syndecans, SNAP25, mucolipin-2, GluT1 and GluT4, T-cell surface glycoprotein CD1, epican (CD44; Indian blood group), CD55 (decay accelerating factor for complement; Cromer blood group), basigin (Ok blood group; or CD147), SLC3a (activators of dibasic and neutral amino acid transport; CD98), and ICAM1 adhesion molecule [847]. Membrane raft components such as GM1 ganglioside are also recycled to the plasma membrane using ARF6 GTPase.
Several effectors bound by active ARF6 intervene in the postendocytic transfer, such as vacuolar ATPase and a component of the exocyst complex Sec10 that mediates docking and fusion of carrier vesicles with the plasma membrane [847].
Protein ARF6 promotes actin remodeling, as it influences lipid metabolism, links to partner of Rac1 POR1 and arfaptin-2, and modulates Rac1 activity. In epithelial cells, ARF6 promotes the internalization of E-cadherin, hence causing disassembly of adherens junctions [840]. Protein ARF6 may promote endocytosis of E-cadherin via nucleotide-diphosphate kinase (NDK) [847]. During mitosis, activated ARF6 localizes to the cleavage furrow and acts via arfophilins and the exocyst complex.82
Among 15 guanine nucleotide-exchange factors ArfGEFs, 12 can activate ARF6, such as 4 cytohesins83 that may act on both ARF6 and ARF1, 3 BRAGs, and 3 EFA6s [847]. Among ArfGAPs that regulate ARF6 activity, ACAP1, ACAP2, and ARAP3 may be specific, whereas others, such as APAP1 and APAP2 act on both ARF6 and other ARFs [847].
9.3.1.6 Family of ARL Regulatory GTPases (ARF Superfamily)
Adpribosylation factor-like proteins are members of the ARF superfamily. The ARL family include 24 members (Table 9.23).
Table 9.23
Members of the ARL family.
Protein | Human genes |
---|---|
ARL1 | Arl1 |
ARL2 | Arl2 |
ARL3 | Arl3 |
ARL4a, ARL4c–ARL4d | Arl4A, Arl4C, Arl4D |
ARL7 | Arl4C |
ARL5, ARL5a–ARL5b | Arl5A–Arl5B |
ARL6 | Arl6 |
ARL8a–ARL8b | Arl8A–Arl8B |
ARL9 | Arl9 |
ARL10 | Arl10 |
ARL11 | Arl11 |
ARL13a–ARL13b | Arl13A–Arl13B |
ARL2L1 | Arl13b |
ARL14 | Arl14 |
ARL15 | Arl15 |
ARL16 | Arl16 |
ARL17 | Arl17 (Arl17B) |
ARL18 | ARFRP1 |
Generally, ARL proteins bind to cell membranes using an N-terminal helix that is inserted into the lipid bilayer once activated. In addition, this N-terminal helix contains a myristoyl or an acetyl group.
ARL1
Ubiquitous ARF-like protein-1 (ARL1) is enriched on vesiculotubular structures on the side of the Golgi body [848]. Binding of ARL1 to membranes depends on co-translational N-myristoylation. Agent ARL1GDP is cytosolic, whereas ARL1GTP is anchored to membranes, especially those of the trans-Golgi network.
ARL2
Ubiquitous ARF-like protein-2 (ARL2) is involved in microtubule dynamics and stability [849]. Isoform ARL2 participates in folding of tubulin and/or tubulin heterodimer assembly by binding to tubulin-specific cochaperone cofactor-D and protein phosphatase-2. Protein ARL2 associates with binder of ARL2 (BART) and adenine nucleotide transporter ANT1 at the inner mitochondrial membrane.
ARL4
Three ARL4 proteins exist. Proteins ARL4a, ARL4c, and ARL4d differ from other members of the ARF superfamily, as they possess a basic C-terminal bipartite nuclear localization signal [850]. Small GTPase ARL4d is similar to ARL4a and ARL4c (60% and 58% identity, respectively). All the ARL4 GTPases recruit cytohesin ArfGEFs to the plasma membrane.
Adpribosylation factor-like protein-4d (ARL4d)84 resides in the nucleus and cytoplasm. Localization of ARL4d at the plasma membrane depends on both GTP binding and N-terminal myristoylation. Human Arl4D mRNA is predominantly detected in the kidney, esophagus, testis, and uterus.
ARL8
The ARL8 subfamily contains 2 isoforms (ARL8a–ARL8b). These proteins contain an N-terminal helix and GTP-binding domains (G1-G5). Both ARL8a and ARL8b localize to lysosomes. Both ARL8a and ARL8b cannot be myristoylated. However, they can be acetylated.
Ubiquitous ARL8a and ARL8b acts mainly in the delivery of endocytosed macromolecules to lysosomes, lysosome motility, chromosome segregation, and axonal transport of presynaptic cargos [851]. In addition, ARL8b associates with β-tubulin.
ARL13
Ubiquitous ARF-like protein-13b (ARL13b) is required for the formation and/or maintenance of cilia, hence associated with sonic Hedgehog signaling [845]. Isotype ARL13b is able to self-associate. Two alternatively spliced transcripts produce long and short isoforms.
9.3.1.7 ArfRP GTPase (ARF Superfamily)
Adpribosylation factor-related protein ArfRP185 is another Ras-related, membrane-associated, monomeric GTPase with some degree of similarity with ARF molecules. Protein ArfRP1 has an unusual feature for an ARF superfamily member: it has a rather high intrinsic GTPase activity. It is mainly associated with the trans-Golgi network [852]. This regulator works synergistically with ARL1 as well as golgin-97 and -245 on Golgi membranes.
In addition, ArfRP1 associates with the plasma membrane. Protein ArfRP1 connects to ArfGEF cytohesin-1 and -2 (Sect. 9.4.1.1. Protein ArfRP1GTP stimulates phospholipase-D to produce phosphatidic acid, a messenger for vesicle formation and trafficking [853]. Phospholipase-D1a and -D1b reside in the perinuclear region, whereas PLD2 is attached to the plasma membrane and is less responsive to ARFs than PLD1 enzyme. Phospholipase-D2 can be stimulated from activated receptor Tyr kinase and G-protein-coupled receptors that prime ARF-, Rho-, and/or PKC-dependent signaling.
9.3.1.8 ARF Domain-Containing Protein ARD1 (ARF Superfamily)
Adpribosylation factor domain-containing protein-1 (ARD1) that pertains to the ARF superfamily of small GTPases corresponds to ubiquitin-protein ligase TriM23 of the tripartite motif (TRIM) family. Members of the TRIM family are implicated in gene transcription, signal transduction, vesicular transport, antiviral defense,86 phospholipase-D activation, and protein degradation via ubiquitination. Three alternatively spliced transcript variants have been detected in humans (ARD1α–ARD1γ).
The TRIM motif includes 3 zinc-binding domains, a RING, B-box type 1 and type 2, and a coiled-coil region. Its C-terminus contains an adpribosylation factor (ARF or P3) domain and a guanine nucleotide-binding site. Its N-terminus possesses a GTPase-activating protein (GAP or P5) domain. Protein ARD1 cycles between active (GTP-bound) and inactive (GDP-bound) forms. Unlike ARFs, ARD1 lacks the myristoylation site in its ARF domain. The GAP activity of ARD1 is restricted to its ARF domain, without GAP activity for any other ARFs [854].
Protein ARD1 localizes to lysosomes and the Golgi body. It operates in the formation of intracellular transport vesicles and their displacement between cellular compartments, as well as phospholipase-D activation [854].
9.3.1.9 SAR GTPase (ARF Superfamily)
Small GTPase secretion-associated and Ras-related protein SAR1, a member of the RAS superfamily, operates as a molecular switch to control protein–protein and protein–lipid interactions during vesicle budding from the endoplasmic reticulum. Unlike all Ras GTPases that use either myristoyl or prenyl groups to direct membrane association and function, SAR1 lacks such modifications, but contains a SAR1-NH2-terminal activation recruitment (STAR) motif [855]. The STAR motif mediates the recruitment of SAR1 to endoplasmic reticulum membranes and facilitates its interaction with membrane-associated guanine nucleotide exchange factor Sec12. In addition, an N-terminal motif assists interaction with the GTPase-activating protein complex Sec23/24. Small GTPase SAR1 coordinates CoP2 coat assembly and disassembly and cargo selection with the recruitment of CoP2 coat to initiate export from the endoplasmic reticulum.
9.3.2 CDC42 GTPase (RHO Superfamily)
CDC42 regulates the actin cytoskeleton and thus cell polarity and migration. CDC42 actually controls the formation of actin-rich filopodia (Vol. 2 – Chap. 6. Cell Motility),89 as well as Rac, RhoD, RhoF, RhoQ, and RhoU [831]. CDC42 induces actin polymerization by binding to Wiskott-Aldrich syndrome protein (WASP ), related nWASP, or via insulin-receptor substrate IRSp53 Tyr kinase to induce actin bundles and branched actin filaments owing to actin-related protein ARP2–ARP3 complex. In addition, CDC42-mediated activation of P21-activated kinase phosphorylates LIMK kinase that phosphorylates (inhibits) cofilin.
CDC42 is involved in chemotaxis of several cell types, such as macrophages, neutrophils, T lymphocytes, and fibroblasts. CDC42 contribution is cell-type specific, as CDC42 is not required for migration of some cell types. In addition, CDC42 regulates cell polarity via partitioning-defective proteins Par6 and Par3 and atypical protein kinase-C. Protein CDC42 also operates via myotonic-dystrophy-kinase-related CDC42-binding kinase (MRCK) to move the nucleus behind the microtubule-organizing center. CDC42 also influences hematopoietic cell differentiation and cell cycle progression.
9.3.3 Superfamily of Rab GTPases
Small Rab (Ras-related proteins in brain) GTPases constitute the largest superfamily of the RAS hyperfamily, with more than 70 members encoded by the human genome.
The Rab proteins regulate all aspects of intracellular transfer of materials between different membrane-enclosed organelles or between organelles and the plasma membrane. They ensure that cargos are delivered to their correct destinations and control membrane identity. Transport of molecules (Vol. 1 – Chap. 9. Intracellular Transport) starts from budding of vesicular or tubular carriers from donor membranes and culminates in their docking and fusion to specific acceptor membranes. Small Rab GTPases of the plasma membrane and organelle membranes behave as membrane-associated switches that regulate vesicle budding, motility, uncoating, docking, and fusion, via recruitment of effectors, such as sorting adaptors, tethering factors, kinases, and phosphatases, and their interactions with coat components, SNARE mediators of membrane fusion, and nanomotors that propel vesicles along microtubules and actin filaments, such as microtubule minus-end-directed dynein, plus-end-directed kinesins,90 and myosins.
Small Rab GTPases control transport fluxes, as they cycle between active and inactive forms. This cycle is regulated by GTPase-activating proteins (RabGAP; Sect. 9.4.2.2) and guanine nucleotide-exchange factors (RabGEF; Sect. 9.4.1.2), such as guanine-exchange factor complex TraPP (transport protein particle).91 The RabGAP proteins inactivate Rab GTPases that are re-activated by RabGEF factors. Active RabGTP GTPases bind their effectors, although some Rab effectors prefer the GDP-bound form [856].
Multiple Rab GTPases interact as they share effectors or via recruitment of selective Rab activators to warrant the spatiotemporal regulation of vesicle transfer. Different Rab GTPases associated with distinct subdomains of the same membrane or distinct types of cellular membranes can actually interact via effectors that are coupled to specific guanine nucleotide exchange factors and GTPase-activating proteins.
A Rab effector complex can contain guanine nucleotide-exchange factors for the same Rab GTPase, thereby generating a positive feedback loop, or for another Rab that is then activated (effector-mediated Rab activation). Effectors of Rab proteins frequently contain separate binding sites for 2 types of Rab GTPases that enable coordination of nanodomains in a given membrane or tethering between 2 membranes (effector-mediated Rab coupling). The effector complex of the secondarily activated Rab GTPase that contains a GTPase-activating protein for the first Rab GTPase triggers a negative feedback loop.
Membrane type | Rab Types |
---|---|
Apical recycling endosome | Rab15, Rab17, Rab25 |
Apical tubule (kidney) | Rab18, Rab20 |
Autophagosome | Rab7, Rab24, Rab33 |
Caveosome | Rab5 |
Centrosome | Rab12 |
Cilium | Rab8, Rab17, Rab23 |
Early endosome | Rab4, Rab5, Rab15, Rab21, Rab22 |
Early phagosome | Rab5, Rab14, Rab22 |
Endoplasmic reticulum | Rab1, Rab2 |
Endosomes | Rab8, Rab13, Rab35 |
Golgi body | Rab1, Rab2, Rab6, Rab8, Rab10, Rab33, Rab40 |
GluT4 vesicle | Rab8, Rab10, Rab13, Rab14 |
Late endosome | Rab7, Rab9 |
Late phagosome | Rab5, Rab7, Rab14, Rab22 |
Liquid droplet | Rab18 |
Lysosome | Rab7, Rab34 |
Macropinosome | Rab34, Rab35 |
Melanosome | Rab27, Rab32, Rab38 |
Mitochondrium | Rab32 |
Nuclear inclusion | Rab24 |
Plasma membrane | Rab3, Rab4, Rab5, Rab8, Rab10, Rab11, Rab14, |
Rab15, Rab17, Rab22, Rab23, Rab25, Rab26, Rab27, | |
Rab34, Rab35, Rab37 | |
Recycling endosome | Rab4, Rab11, Rab15, Rab35 |
Secretory granule | |
in neuroendocrine cells, | Rab18 |
in mastocyte | Rab37 |
Insulin secretory vesicle | Rab3, Rab26, Rab27, Rab37 |
Synaptic vesicle | Rab3 |
Tight junction | Rab13 |
Trans-Golgi network | Rab8, Rab9, Rab22, Rab31 |
Newly synthesized RabGDP GTPases link to Rab escort protein (REP) [856]. The latter presents Rab GTPases to a geranylgeranyl transferase (GGT). Geranylgeranylated, RabGDP are targeted by Rab dissociation inhibitor (RabGDI). The RabGDI sequestrators preclude GDP release from Rab, thereby stabilizing the inactive form. Yet, the Rab–RabGDI complex that chaperones geranylgeranylated Rab GTPases in the cytosol mediates their delivery to their recipient membranes and recycles them back to the cytosol. The Rab–RabGDI complex, indeed, identifies specific membranes via a membrane-bound GDI displacement factor (GDF) [856].
The superfamily of Rab GTPases is the largest set within the hyperfamily of Ras-like monomeric GTPases. In humans, about 70 Rab types have been identified, each with a specific subcellular location, many with a specific tissue distribution (Table 9.24).
Small GTPases Rab1 and Rab2 localize to the endoplasmic reticulum, especially the pre-Golgi intermediate compartment, and Golgi stack;92 Rab3a in secretory vesicles; Rab4 and Rab5 in early endosomes;93 Rab6 in the Golgi body;94 Rab7 and Rab9 in late endosomes; and Rab11 in recycling endosomes.
Proteins of the RAB superfamily have been classified into 8 functional groups according to sequence similarity, subcellular location, and function [859]: Group 1 Rab with Rab1a, Rab1b, and Rab35; 2 with Rab2a, Rab2b, Rab4a, Rab4b, Rab11a, Rab11b, Rab14, and Rab25; 3 with Rab3a to Rab3d, Rab26, Rab27a, Rab27b, and Rab37; 4 with Rab19, Rab30, and Rab43; 5 with Rab5a to Rab5c, Rab21, and Rab22a to Rab22c; 6 with Rab6a to Rab6c and Rab41; 7 with Rab7, Rab9a, and Rab9b; 8 with Rab8a, Rab8b, Rab10, and Rab13. Other components of the dendogram95 comprises the Rab12–Rab15 branch between groups 1 and 3; Rab18, Rab33a–Rab33b, and Rab39a–Rab39b branches between groups 2 and 4; Rab20 and Rab17–Rab24–Rab34–Rab36 branches between groups 2 and 5; Rab28 between groups 5 and 6, the Rab29–Rab32–Rab38 branch between groups 6 and 7; and Rab23 and the Rab40a-to-Rab40c branches between groups 7 and 8. Dendrogram clustering suggests 14 RAB families, but uncharacterized members exist [858] (Table 9.25).
Table 9.25
Families of Rab GTPases (Source: [858]; RasEF: RAS and EF-hand domain-containing protein, or Ras-related protein Rab45, an atypical GTPase with a coiled-coil motif at the mid region, an N-terminal EF-hand domain, and a C-terminal Rab-homology domain). Some members of the RAB superfamily that signal to the nucleus (Rab5, Rab8, Rab24, and possibly others) may cooperate with other RAB superfamily members — Ran GTPases —, which control nucleocytoplasmic shuttling. Small GTPase Rab32 regulates mitochondrial fission and may participate in adaptation to changing energy requirements during growth. Cell growth and differentiation can be modulated by the coordinated actions of Rab GTPases that regulate cell–matrix and cell–cell adhesion (Rab4a, Rab8b, Rab13, and Rab21) and those involved in growth regulation and cell proliferation and apoptosis (Rab6a, Rab11, Rab12, Rab23, Rab25, Rab35, as well as Ran and likely others).
family | Members |
---|---|
1 | Rab23 |
2 | Rab29, Rab32, Rab38, Rab7L1 |
3 | RabL2, RabL3, RabL5 |
4 | Ran |
5 | Rab7, Rab7b, Rab9a/b/c |
6 | Rab28, RabL4 |
7 | Rab34, Rab36 |
8 | Rab6a/a’/b/c, Rab41 |
9 | Rab5a/b/c, Rab17, Rab20, Rab21, Rab22a/b (or Rab31)/c, Rab24 |
10 | Rab18 |
11 | Rab2a/b, Rab4a/b/c, Rab11a/b, Rab14, Rab25, Rab39, Rab42 |
12 | Rab19, Rab30, Rab33a/b, Rab43 |
13 | Rab1a/b, Rab3a/b/c/d, Rab8a/b, Rab10, Rab12, Rab13, Rab15, |
Rab35, Rab40 | |
14 | Rab26, Rab27a/b, Rab37, Rab44, RasEF (Rab45) |
Small Rab GTPases are characterized by their steady-state location at cytosolic surface of particular cell membranes. Each Rab protein indeed has a unique subcellular membrane distribution, although they travel between the cytosol and their target membrane(s).96 In addition, Rab effectors can target at least some members of certain Rab functional groups, without interacting with constituents of other groups. Ten major sets of Rab include Rab1, Rab3, Rab4, Rab5, Rab6, Rab7, Rab8, Rab11, Rab28, and Rab38 groups [860].
Sequential activation of a series of Rabs by a RabGEF module in which RabGEF operates successively is required for intracellular trafficking. Rabs can recruit GEFs that activate subsequently acting Rabs to initiate next transport stage, such as vesicle formation to release or vesicle targeting to docking and then fusion.97 Complexes formed by Rab GTPases and their effectors may exclude other Rab types. Rapid Rab re-activation by specific guanine nucleotide-exchange factors can be prevented by RabGDI that extracts from the membrane and sequesters Rab GTPases into the cytosol.
Recruitment of Rab-specific effectors defines functional nanocompartments that form transport vesicles, link vesicles to motor proteins, and dock and fuse vesicles with their targets. Rab GTPases avoid mixing with one another on a membrane surface, hence disturbances in transport sorting. A given Rab protein can inactivate a previously acting Rab, as it can recruit its cognate GTPase-activating protein to delineate boundaries between Rab GTPases on membranes and determine directionality to membrane traffic. Similar to RabGEF cascade, RabGAP cascade restricts the spatial and temporal overlap of Rab GTPases [862].
9.3.3.1 Group-1 Rab GTPases
Rab1
The Rab1 isoforms (Rab1a–Rab1b) reside at the endoplasmic reticulum and Golgi body as well as intermediate tubules. They regulate the anterograde transport of cargos between the endoplasmic reticulum and Golgi body. They interact with vesicle docking protein, golgin-A2, -A5, and -B1 (giantin), Rab-binding effector protein RabBP1, Rab escort protein-1 and -2, Rab acceptor-1, Rab-interacting factor, Rab1-interacting iporin,98 actin-bundling fascin Fscn1, SLC16a8, cyclin-dependent kinase inhibitor-1A, riboflavin kinase, MiCaL1,99 etc. [251].
Rab1a
Protein Rab1a assists in transferring secretory vesicles from the endoplasmic reticulum to the endoplasmic reticulum–Golgi intermediate compartment (ERGIC) and cis-Golgi network [863]. Three Rab1a splice variants can be observed, isoform Rab1a1 being the reference sequence.
During cardiogenesis, Rab1a is involved in plasmalemmal localization of angiotensin-2 and adrenergic receptors, except α2b-adrenoceptor [863].
In addition, GTPase Rab1a binds to epithelial Ca
channel. It interacts with Rab escort proteins (REP1 and REP2) to facilitate the geranylgeranylation and membrane delivery of Rab proteins [863].

Rab35
Ubiquitous Rab35 regulates fast recycling of endocytic membranes, especially during cytokinesis as it forms a protein complex to stabilize the bridge between daughter cells prior to abscission. It is also involved in fast recycling of transferrin receptors to the plasma membrane and transport of class-2 major histocompatibility complexes from the plasma membrane to early endosomes and back to plasma membrane in antigen-presenting cells, as well as immunological synapse between antigen-presenting cells and T lymphocytes [864].
9.3.3.2 Group-2 Rab GTPases
Rab4
Small GTPase Rab4 mediates fast endocytic recycling directly from early endosomes [856]. Early and recycling endosomes contain 3 types of nanodomains with: (1) only Rab5, (2) Rab4 and Rab5, and (3) Rab4 and Rab11 GTPases. Small GTPases Rab4 and Rab5 are involved in endocytic recycling and endosome fusion, hence in the formation of recycling and late endosomes, respectively. Whereas Rab4 GTPase is involved in fast endocytic recycling, Rab11 and Rab35 in slow endocytic recycling via recycling endosomes; Rab21 in integrin endocytosis; Rab15 in transfer from early endosomes to recycling endosomes and from apical recycling endosomes to the basolateral plasma membrane [856]. The Rab effector Rabenosyn-5 contains separate binding sites for Rab4 and Rab5 GTPases.
Rab11
The subfamily of Rab11 GTPases includes Rab11a, Rab11b, and Rab25. Ubiquitous Rab11a and Rab11b have redundant functions, whereas Rab25 expression is restricted to polarized epithelial cells. Among Rab11 effectors, Rab11-interacting proteins target all members of the Rab11 group. Small GTPases Rab11 and Rab35 mediate slow endocytic recycling through recycling endosomes [856].
Small GTPase Rab11a regulates the recycling of transferrin receptor, β-integrin, TRPV5 and TRPV6 channels, glucose transporter GluT4, and chemokine CXCR receptor [865]. It binds to epithelial Ca
channel. Active Rab11b interacts with Rab11-interacting proteins. Like Rab11a, Rab11b associates with recycling endosomes. In polarized epithelial cells, Rab11b regulates the apical recycling compartment. It also contributes to localization of recycling endosome to the cleavage furrow during mitosis.

Rab11 family-interacting proteins (Rab11FIP) that are classified into groups 1 (Rab11FIP1–Rab11FIP2 and Rab11FIP5) and -2 (Rab11FIP3–Rab11FIP4) bind to Rab11GTP. They cycle between the cytosol and recycling endosomes. Group-1 Rab11FIPs are implicated in the regulation of endocytic protein sorting and recycling, whereas group-2 Rab11FIPs intervene in endosomal transport.100 Adaptor Rab11FIP2 links Rab11a + endocytic recycling vesicles to myosin-5B for vesicle motility [856]. Myosin-5B is also a direct effector of Rab11a GTPase.
9.3.3.3 Group-3 Rab GTPases
Rab3
Isoforms of Rab3 molecule (Rab3a–Rab3d) operate in synaptic vesicle and calcium-dependent regulated exocytosis. They interact and complex with Rab escort proteins. Small Rab3a GTPase participates in the regulation of neurotransmitter release. Paralogs Rab3a and Rab3b can bind Ca
-binding rabphilin-3, a regulator of neurotransmitter release. Rabphilin, brain-specific scaffolds regulating synaptic membrane exocytosis proteins Rim1 and Rim2, and Rab effector rabphilin-3a-like protein (Rph3aL or Noc2) involved in exocytosis in endocrine cells are Rab3-specific effectors that control vesicle exocytosis in neurons and some endocrine cells.101

Rab3-interacting molecule (RIM) is a scaffold protein that participates in docking and fusion of secretory vesicles at release sites. It tethers to synaptosomal-associated protein SNAP25 and membrane-trafficking synaptotagmin-1. It also binds to pore-forming subunit-α1B ofCaV2.2 channels. It also weakly connects to subunit-α1c of CaV1 channels. In the presence of Ca
, RIM interaction with synaptotagmin-1 rises, but that with SNAP25 decays [867].

Rab26
Rab26 is tethered to membranes of secretory granules and synaptic vesicles. It thus mainly resides in cells with regulated exocytosis or high secretory activity. Scaffold Rim1, a member of the synaptotagmin-like protein family, is a known effector of Rab26 that also interacts with Rab3a to Rab3d, Rab10, and Rab37 [868]. Rab26 constitutes a subfamily with Rab3 isoforms, Rab27 isoforms, Rab37, Rab44, and Rab45.
Rab27
Small GTPase Rab27 regulates exocytosis of granules in cytotoxic T lymphocytes and melanosomes in melanocytes. Members of the synaptotagmin-like protein group (SLP1–SLP4 or SytL1–SytL4)102 of the C2 domain-containing protein family and SLP homologs lacking the C2 domain SLaC2a, or melanophilin, and SLaC2b interact specifically with Rab27aGTP and Rab27bGTP [869].103
Small Rab27 GTPases coordinate vesicle motility, as adaptor SLaC2a also attaches to myosin-5a and -7a and actin [869]. Myosin-5a then transfers Rab27a + vesicles toward the cell cortex. Synaptotagmin-like proteins SLP1 and SLP2 colocalize with Rab27a in melanosomes.104
In addition, small GTPase Rab27 regulates dense core granule secretion in platelets with Rab27-binding protein Munc13-4 [871]. Isoform Rab27a complexes with granuphilin in pancreatic β cells to regulate exocytosis of insulin-containing dense-core granules [872]. Small GTPase Rab27b is most abundantly produced in the pituitary gland. Yet, Rab27b localizes to zymogen granules and regulates pancreatic acinar exocytosis.
Small GTPase Rab27a not only mediates vesicle–nanomotor attachment, but also controls docking of exocytic dense-core vesicles to the plasma membrane. Granuphilin, or SLP4, an effector of Rab27a, interacts directly with Sec1-related syntaxin-binding protein StxBP1 (a.k.a. Munc18-1) [856]. Release of granuphilin precedes SNARE complex assembly and membrane fusion.
Rab37
Small GTPase Rab37 that is closely related to Rab26 contributes to masocyte degranulation.
9.3.3.4 Group-4 Rab GTPases
Golgins are long coiled-coil proteins that localize to particular Golgi subdomains via their C-termini. GRIP domain-containing golgins (golgin-97 and -245 and GRIP and coiled-coil domain-containing proteins GCC1 or GCC88 and GCC2 or GCC185) bind via their C-termini Arf-like protein ArL1 on the trans-Golgi network as well as Rab2, Rab6, Rab19, and Rab30. These molecules of the trans-Golgi network as well as other compartments of the Golgi body such as cis-Golgi network capture Rab-containing vesicles and exclude ribosomes (sise ∼ 25 nm) [873]. Actin and spectrin form a mesh around the Golgi body that reorganizes for vesicle arrival and departure.
Rab19
Small GTPase Rab19 can be detected at high levels in lung, intestine, and spleen and at a lower level in kidney, whereas the brain, heart, and liver contain only very little or no detectable amounts [874].
Rab30
Small GTPase Rab30 can be detected in vascular endothelial cells. Jun N-terminal kinase can influence intracellular transport via small GTPase Rab30 [875].
Rab43
Small GTPase Rab43 (a.k.a. Rab11b, and Rab41) is expressed in the brain, lung, heart, kidney, spleen, colon, uterus, ovary, and testis, but not in liver. It is involved in trafficking between the Golgi body and endoplasmic reticulum [876].
9.3.3.5 Group-5 Rab GTPases
Rab5
Protein Rab5 is involved in budding of clathrin-coated vesicles from the plasma membrane and their transport to early endosomes as well as fusion of early endosomes. It localizes not only to early endosomes, but also phagosomes and caveosomes, in addition to plasma membrane [856]. It mediates endocytosis and endosome fusion of clathrin-coated vesicles, macropinocytosis with Rab34, and maturation of early phagosomes with Rab14 and Rab22 GTPases [856].
The Rab GTPases cooperate with components of the vesicle docking and fusion machinery such as SNARE proteins. Small Rab5 GTPase links to tethering factors early endosome antigen EEA1 and rabenosyn-5 to mediate fusion of endosomal membranes [856]. During membrane fusion of homotypic early endosomes, the complex of RabGEF1 (or RabEx5) and Rab-binding effector protein RabEP1 (or rabaptin-5) stimulates and stabilizes Rab5 protein. The latter can then recruit PI3K to produce PI(3)P as well as Rab5 effectors such as early endosome antigen EEA1 and rabenosyn-5 that assemble with VPS45 vacuolar protein sorting, a member of the Sec1 family of SNARE regulators. In addition, EEA1 and rabenosyn-5 can directly interact with endosomal SNAREs syntaxin-6 and -13 and syntaxin-7, respectively.
During membrane fission, Rab5-binding protein RIn3 interacts with amphiphysin-2 that mediates receptor-induced endocytosis from plasma membranes to early endosomes [877].
Vesicle coat complexes required for cargo sequestration and membrane budding must be shed prior to acceptor membrane fusion. Endocytic vesicles with clathrin coat can recruit cargo adaptor complex AP2 via phosphatidylinositol (4,5)-bisphosphate and adaptin-associated kinase AAK1 that phosphorylates AP2 μ2 subunit. Clathrin-coated vesicle-associated Rab5 and its GEF, GAP, and VPS9 domain-containing protein GAPVD1 coordinate AP2 uncoating, as they promote dephosphorylation of AP2 subunit μ2 and increase PI(4,5)P2 turnover [856].
Small Rab GTPases specify membrane identity, as they control local levels of phosphoinositides and recruit specific effectors to restricted membrane nanodomains. The Rab5 effectors EEA1, rabenosyn-5, and rabankyrin-5 (or AnkFy1) contain PI(3)P-binding FYVE domains. Phosphoinositide PI(3)P, in turn, recruits the retromer subunits — sorting nexins SNx1 and SNx2 — that control the plasmalemmal density of transmembrane receptors. Subunits SNx1 and SNx2 associate with the cargo-interacting retromer VPS26–VPS29–VPS35 subcomplex, an effector of Rab7 GTPase (Rab crosstalk) [856].
Endocytic vesicles experience a Rab5-to-Rab7 conversion. Once it is activated by Rab5GEFs on donor membranes, Rab5GTP is recruited to the endocytic vesicular membrane. Accumulation of PI(3)P105 in the vesicular membrane enables the recruitment of vacuolar fusion protein Mon1b, thereby attracting and activating Rab7 and inhibiting the Rab5–Rab5GEF positive feedback.106 The maturation of Rab5 + early endocytic structures into later Rab7 + vesicles involves Rab5-mediated recruitment of the tether homotypic fusion and vacuole protein sorting complex (HoPS), in which VPS39 subunit is a Rab7GEF.107 Small GTPase Rab7 then recruits a Rab5GAP and causes Rab5–Mon1b–Rab7 negative feedback loop). Then, Rab5GAPs inactivate Rab5 and Rab5GDIs sequester inactivated Rab5 into the cytosol, avoiding its degradation and allowing its transfer to the plasma membrane, where it may once more be activated on newly formed vesicles. Similarly, as the vesicles reach the stage of late endosomes, Rab7GAPs on these membranes inactivate Rab7, and Rab7GDIs sequester it into the cytosol and transporting it to vesicles for Rab5–Rab7 conversion.
Class-1 PI3K kinases that phosphorylate PI(4,5)P2 to PI(3,4,5)P3at the plasma membrane as well as class-3 PI3K kinases that phosphorylate PI to PI(3)P on endosome membranes are effectors of Rab5 [856]. Phosphoinositide 4- and 5- phosphatases that dephosphorylate PI(3,4)P2 to PI(3)P and PI(3,4,5)P3 to PI(3,4)P2, respectively, are also effectors of Rab5 GTPase.
Small GTPase Rab5 then controls the formation of PI(3)P that recruits KIF16b kinesin [856]. Kinesins can hence be indirectly regulated by Rab GTPases for vesicle motility. Once their destination is reached, membrane-tethering complexes enter into action. They often contain RabGEFs. The Rab5 effector rabaptin-5 complexes with RabGEF1 (or rabex-5) to amplify Rab5 activation in nanodomains of endosomal membrane [856].
Endocytosis-associated Rab5 serves as a hub for cooperation between cell signaling and trafficking. Activation of the epidermal growth factor receptor activates Rab5 via RasGEF Son of sevenless SOS1, small GTPase Ras, and Rab5GEF Ras and Rab interactor RIn1 to stimulate EGFR transfer to early endosomes [856]. Small GTPase Rab5 also stimulates macropinocytosis via Rab5 effector USP6 N-terminal-like protein (USP6NL or Related to the N-terminus of Tre [RNTRE]) that interacts directly with actin or via actin-binding actinin-4. Protein USP6NL, a Rab5GAP, is recruited to EGFR via EFGR kinase substrate EPS8 that activates Rac1 GTPase via adaptor Abl interactor ABl1 and RasGEF SOS1. In addition, Rac1 interacts with Rab5 on endosomes via the recruitment of Rab5GEF alsin. The Rab5 effectors — adaptor phosphotyrosine interaction, PH domain, and leucine zipper-containing proteins APPL1 and APPL2 — reside in a subpopulation of endosomes and translocate to the nucleus, where they connect to the chromatin remodeling complex NuRD, an histone deacetylase complex, in response to EGF stimulation.
Rab21
Rab22
Small GTPase Rab22 mediates trafficking between the trans-Golgi network and early endosomes and conversely [856].
9.3.3.6 Group-6 Rab GTPases
Rab6
Golgi body-localized Rab6, Rab33, and Rab40 mediate intra-Golgi trafficking [856]. Kinesins are directly or indirectly regulated by Rab6 GTPase [856]. Protein Rab6 of the Golgi body directly regulates kinesins for vesicle motility, as cytokinesis regulator KiF20a, or rabkinesin-6, is its direct effector. The Rab6 effector Bicaudal-D1 mediates attachment of Golgi vesicles to dynein–dynactin complex in transport from the Golgi body to the endoplasmic reticulum.
The Rab effector Golgi coiled-coil protein GCC2 that contains several Rab-binding sites localizes to Golgi membranes via interactions with Rab6 and adpribosylation factor-like protein ARL1 [856]. It also binds to Rab1, Rab2, Rab9, Rab15, Rab27b, Rab30, and Rab33b.
9.3.3.7 Group-7 Rab GTPases
Rab7
Late endosome-associated GTPase Rab7 mediates maturation from early endosome of late endosomes and phagosomes, and their fusion with lysosomes [856]. Rubicon, a component of the endosomal class-3 PI3K complex (or vacuolar protein sorting VPS34) prevents endosome maturation, as it impedes Rab7 activation [879]. Molecule UV radiation resistance-associated gene product (UVRAG, or VPS38) activates PI3Kc3 and class-C VPS, a Rab7GEF protein. Rubicon sequesters UVRAG from class-C VPS [880]. Active Rab7 competes for Rubicon binding and releases UVRAG to associate with C-VPS, which, in turn, further activates Rab7 GTPase. This feedforward loop ensures rapid amplification of Rab7 activation and subsequent endosome maturation.
Microtubule minus-end-directed nanomotor dynein is an indirect effector for late endosome-associated Rab7 GTPase. The Rab7 effector Rab-interacting lysosomal protein (RILP) recruits a subunit of the dynactin complex that connects to dynein.
Rab9
Late endosome-associated Rab9 GTPase mediates transfer from late endosomes to the trans-Golgi network [856]. Widespread Rab9 GTPase intervenes in cargo-specific coat assembly.
Small Rab9 GTPase operates in the recycling of mannose 6-phosphate receptors (M6PR) from late endosomes to the trans-Golgi network.108 The Rab9 effector — sorting adaptor M6PRBP1 —109 recognizes the cytosolic tail of the 2 mannose 6-phosphate receptors.110
Protein Rab9 activates ubiquitous Rho-related BTB (Broad complex, Tramtrack, and Bric-à-brac) domain-containing GTPase RhoBTB3 for the docking of transport vesicles at the trans-Golgi network [881].
9.3.3.8 Group-8 Rab GTPases
Skeletal muscle is a major site of dietary glucose storage, using insulin-mediated translocation of GluT4 at the plasma membrane via the PKB–TBC1D4 pathway. Agent TBC1 domain-containing protein TBC1D4,112 a RabGAP does not target the same Rab GTPase in skeletal myocytes and adipocytes; Rab8a is involved in GluT4 exocytosis in myocytes and Rab10 in adipocytes. Insulin promotes activation of both Rab8a and Rab13 of the group-8 Rab GTPases, but not Rab10, in rat myocytes, Rab8a activation preceding that of Rab13 [884]. The latter is characterized by a wider intracellular distribution than that of Rab8a GTPase, restricted to the perinuclear region.
Rab8
Rab8a regulates export of vesicles from the trans-Golgi network, transport along actin filaments and microtubules, and fusion with the plasma membrane [885]. Numerous regulators, such as Rab8a-specific GEFs, GAPs, and kinases, modulate Rab8a activity, whereas cytoskeletal nanomotors or motor-binding proteins and effectors coordinate Rab8a function during membrane trafficking.
Small Rab8a GTPase contributes to cell morphogenesis, signaling, and development. Small GTPase Rab8 also participates in translocation of glucose transporter GluT4 vesicle with Rab10 and Rab14 and ciliogenesis with Rab17 and Rab23 GTPases [856]. Myosin-5b is a direct effector of Rab8a GTPase that coordinate vesicle motility [856].
Rab10
Insulin stimulates the translocation of glucose transporter GluT4 from intracellular vesicles to the plasma membrane. Three Rab GTPases — Rab10, Rab11, and Rab14 — lodge on GluT4 + vesicles.
Monomeric Rab10 GTPase can localize to the inner face of the plasma membrane using a lipid anchor. In polarized cells such as epithelial cells, Rab10 mediates transport from basolateral sorting endosomes to common endosomes [886].
9.3.3.9 Other Rab GTPases
Most cells have members of the hyperfamily of small GTPases that are structurally classified into, at least, 5 superfamilies (ARF, Rab, Ran, Ras, and Rho), although expression levels of these members vary. Yet, a few members have a tissue-specific expression. Small GTPase Rab17 is detected only in epithelial cells.
Rab15
Small Rab15 GTPase is involved in the trafficking from early endosomes to recycling endosomes and from apical recycling endosomes to the basolateral plasma membrane [856].
Rab17
In epithelial cells, Rab17 and Rab25 control transfer through the apical recycling endosomes to the apical plasma membrane [856].
Rab18
Small Rab18 GTPase controls the vesicular transport by defining organelle identity and organizing functional membrane nanodomains. It controls the formation of lipid droplets [856].
Rab23
Small Rab23 GTPase is produced in most tissues at a low level. Yet, it is highly expressed in developing brain and spinal cord. It acts as an inhibitor of sonic Hedgehog [887]. Endosomal Rab23 actually causes receptor endocytosis to endosomes, thereby attenuating sonic Hedgehog signaling. It prevents formation of Gli2 activator. It could regulate the subcellular location of Hh signaling components, downstream from Smoothened effector and upstream from Gli mediator.
Rab24
Small Rab24 GTPase that has a very low intrinsic GTPase activity is associated with the autophagosome [888]. It may be implicated in degradation of misfolded proteins as well as in nucleocytoplasmic transport. Protein Rab24 is not efficiently geranylgeranylated. Consequently, Rab24 does not form an observable complex with Rab guanine nucleotide-dissociation inhibitor, as geranylgeranylated RabGDP favors linkage with GDI agents.
Rab32
Small GTPases Rab32 and Rab38 are involved in the genesis of melanosomes. Small GTPase Rab32 also controls mitochondrial fission [856].
Rab44 and Rab45
Small GTPases Rab44 and Rab45 represent unconventional members of the Rab26 family.
9.3.4 Rac GTPases
The Rac family of small GTPases, a subset of the RHO superfamily of the RAS hyperfamily, include 3 isoforms (Rac1–Rac3; Table 9.26) and RhoG [831]. Protein Rac1 is ubiquitous; Rac2 is mainly restricted to hematopoietic cells; Rac3 abounds in brain cells and fibroblasts.113 Small GTPase RhoG is widely expressed albeit at varying levels according to the cell type (Sect. 9.3.17).
Table 9.26
Rac GTPases and their effectors (Source: [889]; BAIAP: brain-specific angiogenesis inhibitor-1-associated insulin receptor substrate; ElMo: engulfment and cell motility adaptor; Ktn: endoplasmic reticulum membrane and microtubule-associated kinectin; p67phox: NADPH oxidase subunit; SH3RF: SH3 domain-containing ring finger ubiquitin-ligase; SRA: steroid receptor RNA activator).
Type | Effectors |
---|---|
Rac1 | PAK1–PAK3, MAP2K |
SH3RF1, SRA1, BAIAP, p67phox | |
Rac2 | PAK1–PAK3 |
Rac3 | PAK1–PAK3 |
RhoG | ElMo, Ktn1 |
Formation and turnover of cell adhesions with adjacent cells and the extracellular matrix is a necessary function of epithelial cells to regulate epithelial barrier integrity and circulating leukocytes that are recruited in tissues to clean and repair. Activated Rac GTPase weakens tight junctions, but stimulates adherens junction formation.
Protein Rac activates actin polymerization during lamellipodium formation via Wiskott-Aldrich syndrome protein family verprolin-homologous protein complex (WAVe) that activates actin-related protein ARP2–ARP3 complex, which generates actin filament branches, and formin diaphanous-2 that nucleates unbranched actin filaments. Moreover, Rac-mediated activation of P21-activated kinase phosphorylates LIMK kinase that phosphorylates (inhibits) actin-severing cofilin. Small Rac GTPases not only stimulate the formation of lamellipodia and membrane ruffles, but also induce membrane protrusions during phagocytosis.114
All components of the renin–angiotensin system, renin, angiotensinogen (or serpin-A8), angiotensin-converting enzyme (ACE), angiotensin-2, angiotensin-2 receptors) localize in the ventricular myocardium, as they are produced by cardiac fibroblasts. Activated angiotensin-2 fosters cardiac remodeling (maladaptive hypertrophy) and fibrosis in overloaded myocardium. In both cardiac myocytes and fibroblasts, integrin-β1, a major mechanosensor, contributes to the regulation of cell growth and gene expression via its effectors of the RHO superfamily and stress-activated protein kinases ( JNKs and P38MAPKs). Transcription of the ATG (angiotensinogen) gene is inhibited by Rac1 via both JNK-dependent and -independent mechanisms, and stimulated by RhoA via a P38MAPK-dependent mechanism [890].115
9.3.4.1 Rac1
Phosphatidylinositol (4,5)-bisphosphate is produced at sites of N-cadherin-mediated intercellular adhesion due to activated Rac1 GTPase. The latter mediates insertion of transient receptor potential channel TRPC5 into the plasma membrane via its stimulation of PI(4)P5K enzyme.
Isoform Rac1 elicits migration of multiple cell types (e.g., macrophages, T lymphocytes, Schwann cells, epithelial cells, and fibroblasts). Response generated by Rac1 relies particularly on chemotactic fMLP peptide. Both Rac1 and Rac2 regulate cell spreading. Both Rac1 and Rac2 intervene in the actin mesh and spectrin scaffold of erythrocytes. Proteins Rac1, Rac3, and RhoG promote axon growth and guidance.
Sumoylation of Rac1 promotes its activity, hence cell migration. Protein inhibitor of activated STAT PIAS3, a small ubiquitin like modifier (SUMo) ligase, interacts with Rac1, particularly Rac1GTP, and primes its sumoylation [891].
9.3.4.2 Rac2
Isoform Rac2 is required for migration and phagocytosis of neutrophils. Protein Rac2 interacts with phagocyte NADPH oxidase NOx2 [892]. Inactive Rac2GDP is bound to RhoGDI that sequesters Rac2 and prevents Rac2 from interacting with membranes as well as GEFs, GAPs, and effectors.
9.3.4.3 Rac3
Ubiquitous Rac3 has an association rate of GDP similar to Rac1, but 13-fold higher than that of Rac2. The intrinsic GTP hydrolysis rate of Rac3 is similar to those of Rac1 or Rac2, but is about 5-fold faster than that of Ras [893]. GTPase-activating protein Bcr can stimulate GTP hydrolysis of Rac3 to a similar degree to that of Rac1. Like other Rac GTPases, guanine nucleotide-exchange factors such as Tiam1 increases the dissociation rate of GDP on Rac3. Protein Rac3 also interacts with guanine nucleotide-dissociation inhibitors.
Its specific effectors include calcium–integrin-binding protein (CIB), nuclear DNA-binding protein C1D, and nuclear receptor-binding protein (NRBP). Constitutively active Rac3 localizes to internal membranes of the endoplasmatic reticulum and Golgi body. Active GTPRac3 can also be detected at the inner leaflet of the plasma membrane and membrane protrusions.
9.3.5 Rad GTPase
Protein Ras associated with diabetes, (Rad; Sect. 9.3.25),116 a member of the RGK (Rad, Gem, and Kir) family of Ras-related GTPases, shares GTP-binding domains with Ras, but lacks the C-terminal CAAX motif involved in prenylation of RAS superfamily members.
Small Rad GTPase interacts with calmodulin and calmodulin-dependent protein kinase CamK2 to preclude its signaling.
In the myocardium, Rad hinders the activity of β-adrenoceptor and ofCaV1.2a channel, thereby reducing cardiomyocyte contractility. Protein Rad then operates as a regulator of cardiac electromechanical coupling and βAR signaling.
Protein Rad contributes to the attenuation of cardiac hypertrophy. It indeed represses connective tissue growth factor (CTGF).117 Cardiomyocytes release Rad that targets cardiac fibroblasts, which produce extracellular matrix, to prevent cardiac fibrosis. In cardiomyocytes,CCAAT/enhancer-binding protein-δ activates CTGF production [894].
In addition, Rad lowers intimal hyperplasia after balloon injury, as it prevents migration of vascular smooth muscle cells activated by the Rho–RoCK pathway in rat carotid arteries [895]. Protein Rad indeed reduces the formation of both focal contacts and stress fibers in this cell type by blocking RoCK signaling.
9.3.6 Rag GTPases
Small Ras-related GTPases Rag include 4 known members (RagA–RagD). Proteins Rag function as heterodimers that consist of RagA or RagB and RagC or RagD.
Subtype RagC associates with the nutrient–energy–redox sensor TOR complex-1 (TORC1) that acts as protein kinase to influence its location within endomembranes of the cell [896]. Small Rag GTPase binds to raptor that mediates Rag–TORC1 interaction.
9.3.7 Ral GTPases
Closely related Ral GTPases — RalA and RalB —118 constitute a family within the RAS hyperfamily (Table 9.27). Small Ral GTPases are activated by a unique nucleotide-exchange factor RalGEF (a.k.a. RalGDS) and inactivated by a distinct GTPase-activating protein RalGAP. The Ral proteins are prenylated to be associated with cellular membranes. Whereas Ras is found almost exclusively in the cell cortex (intracellular egde of the plasma membrane), Ral primarily localizes in cytoplasmic vesicles, in addtion to plasma membrane. Unlike Ras GTPases, Ral does not produce transformed cells when it bears oncogenic mutations that lock it in its active GTP state.
Both RalA and RalB have a calcium-dependent calmodulin-binding domain. Small GTPase Ral and calcium–calmodulin activate phospholipase-Cδ1 [897].
Type | Effectors |
---|---|
RalA | PLD1, PLCδ1, RalBP1 |
RalB |
Protein RalA mediates signaling initiated by plasmalemmal, ligand-bound receptors. Active GTPRalA binds filamin, an actin filament crosslinker and recruiter of membrane and intracellular proteins to actin [898]. It is a CDC42 effector to recruit filamin and generate actin-rich filopodia. In addition, RalA GTPase associates with ARF-responsive, phosphatidylinositol (4,5)-bisphosphate-dependent phospholipase-D1 that hydrolyzes phosphatidylcholine into choline and messenger phosphatidic acid [899]. Moreover, RalA synergistically enhances activation of phospholipase-D1 by ARF1 [900].119
9.3.8 Ran GTPases
Small Ran GTPase intervenes in the transport of proteins between the nucleus and cytoplasm and conversely. Protein Ran can diffuse between the 2 compartments.
The transfer from the cytoplasm to the nucleus is allowed for cytosolic proteins that contain a nuclear localization signal (NLS; proteinNLS). These proteins bind to the importin-α–importin-β complex. The resulting ternary complex crosses the nuclear pore (Vol. 1 – Chaps. 4. Cell Structure and Function and 9. Intracellular Transport). Binding of RanGTP to importin-β dissociates the ternary complex, thereby liberating the NLS-bearing protein.
The transfer from the nucleus to the cytoplasm is allowed for proteins that possess a nuclear export signal (NES). The latter bind to the karyopherin exportin. The resulting complex is exported when bound to RanGTP GTPase.
The basic selectivity associated with structural motifs is complemented by the distributions of proteins affecting the activation state of Ran GTPase. The single known RanGEF — regulator of chromosome condensation RCC1 — is bound to chromatin, whatever the phase of the cell cycle [902]. This regulator continuously generates RanGTP in the nucleus. On the other hand, RanGAPs localize to the cytosol and cytoplasmic face of the nuclear envelope. It dissociating the Ran–exportin–proteinNES complex arriving from the nucleus to the cytoplasm.
9.3.9 Rap GTPases
Monomeric Rap GTPases that are similar in structure to Ras constitute a family of small cytosolic GTPases. These Ras-like small GTPases participate in the control of between-cell and cell–matrix adhesion, especially epithelial and endothelial cell junctions [903].
Small Ras-related Rap GTPases can be subdivided into 2 subfamilies by sequence homology. Subfamily-1 Rap GTPases (Rap1a–Rap1b) are ubiquitous; subfamily-2 Rap GTPases (Rap2) are mainly found in the central nervous system and platelets (Table 9.28).
Type | Effectors |
---|---|
Rap1a | bRaf, cRaf, RalGEF, RGS14 |
Rap1b | bRaf, cRaf, RalGEF, RGS14 |
Rap2a | Rif GTPase, RPIP8, RalGEF, RGS14 |
Rap2b | Rif GTPase, RPIP8, RalGEF, RGS14 |
Rap2c |
9.3.9.1 RAP1 Subfamily
Members of the Rap1 subfamily are able to bind Ras effectors, with an affinity similar to or sometimes higher than Ras, such as cRaf, PI3K, and RalGEFs. They promote activation of certain component of the mitogen-activated protein kinase module, such as bRaf kinase. In addition to their effect on MAPK signaling, Rap1 GTPases participate in cytoskeletal rearrangement and cadherin- and integrin-mediated cell adhesions, as well as the regulation of cell proliferation and differentiation. Both Rap1a and Rap1b contribute to vesicular delivery of E-cadherins with Ral and exocyst complex components ExoC2 and ExoC8, hence controlling plasmalemmal E-cadherin density, as well as cytoskeletal linkage to E-cadherins with catenin-δ1 and Rap1 effector, afadin,121 and thus cell adhesion stabilization. Afadin also binds to Rap1GAP signal-induced proliferation-associated protein SPA1 (or SIPA1). It can then control integrin-mediated cell adhesion via the recruitment of SPA1 and Rap1GTP.
Small GTPase Rap1 is also able to block cell transformation owing to its capacity to form non-productive complexes with Ras effectors. Anti-oncogenic effect of Rap1 indeed relies on sequestration of RasA1 (or p120RasGAP) and cRaf kinase. Conversely, Rap1 can cause Rac activation and mediate relocalization of RacGEFs Vav2 and TIAM1 during intercellular adhesion maturation and cell spreading [903].
In hematopoietic cells, Rap1 provokes lymphocyte aggregation, T-cell anergy, and platelet activation. In the nervous system, Rap1 contributes to the regulation of axonal differentiation, dendritic development, dendritic and spine morphology, and synapse remodeling, in particular synaptic depression by removing AMPARs from synapses via P38MAPK [904]. In addition, Rap1 couples cAMP signaling to ERK1 and ERK2 kinases.
Several Rap guanine nucleotide-exchange factors (RapGEF; Sect. 9.4.1.6)) activate Rap GTPases. They include RapGEF1 (or C3G), RapGEF2 (PDZ-GEF), RapGEF3 and -4 (EPAC1 and -2), Rap(Ras)GRPs, and dedicator of cytokinesis DOCK4.
Ras-related GTPases Rap1 and Rap2 are able to interact with RalGEFs, such as Ral GDP-dissociation stimulator (RalGDS or RalGEF), RalGDS-like (RGL GEF), and RalGDS-like factor (RLF) that, all, are effectors of Ras [905]. In the central nervous system, Rap2 specifically interacts with Rap2-interacting protein RpIP8 (or Rundc3a) [906].
Activation of Rap1 follows its interaction with Rap1-specific RapGEF1 protein. Under basal conditions, RapGEF1 is associated with CRK2 adaptor. Stretch of the extracellular matrix is transduced into intracellular biochemical signals. Small Rap1 GTPase provokes integrin-mediated adhesion and then influences actin dynamics. Protein Rap1 activates integrins via Rap1GTP-interacting adaptor (RIAM) [907].122 Cell stretch activates Rap1 using RapGEF1 and CRK2 (Rap1–CRK2–RapGEF1 pathway) [908]. Stretch-induced phosphorylation of CRK-associated substrate (CAS or BCAR1) of the cytoskeleton is due to SRC family kinases. Adaptor CRK2 binds directly to phosphorylated CAS, especially in cell–matrix adhesion sites.
Small Rap1 GTPase is a member of the shelterin complex, or telosome complex, that regulates the length of telomeres and protects chromosome ends [909].123 Shelterin protects chromosome ends from the activity of DNA-repair molecules.124 Repressor and activator Rap1 protein125 is not involved in the maintenance of telomere length, the organization of telomeric chromatin, and inhibition of non-homologous end joining (NHEJ), a repair process of double-strand DNA breaks at telomeres, but is required for the inhibition of homology-directed repair (HDR) [910].126
Small Rap1 GTPase not only protects telomeres from sister chromatid exchange, but also intervenes in transcriptional regulation. Protein Rap1 may bind to telomeric and extratelomeric (TTAGGG)2 DNA consensus motifs.127 It operates in transcriptional regulation and silences genes in proximal and subtelomeric regions [911].
Small Rap1 GTPase also controls the NFκB pathway that mediates the transcriptional response to various types of cellular and developmental signals and stresses [912]. A significant proportion of cytoplasmic Rap1 is constitutively bound to inhibitor of NFκB kinase (IκB kinases [IKK]). Protein Rap1 enables efficient recruitment of IKKs and phosphorylation of P65NFκB.128 Moreover, NFκB may, in turn, regulate Rap1 production via the NFκB-binding site in the RAP1 promoter region.
Rap1a
Small GTPase Rap1a counteracts Ras mitogenic action, because it competes with Ras to connect to Raf and binds RasGAPs. Protein Rap1a can be phosphorylated by protein kinase-A and calmodulin-dependent kinase.
Rap1b
Small GTPase Rap1b is particularly detected in platelets. It is implicated in cAMP-associated signaling. Like Rap1a, Rap1b can be phosphorylated by cAMP-dependent protein kinase PKA. Phosphorylation of Rap1b by PKA (Ser179) is required for cAMP-dependent cell proliferation and PKB inhibition [913].
9.3.9.2 RAP2 Subfamily
Proteins Rap2a and Rap2b have a low intrinsic GTPase activity. Like Rap1, Rap2 GTPases contribute to synapse remodeling, particularly removal of AMPA receptors via Jun N-terminal kinases. Besides, neither Rap2a nor Rap2b antagonize Ras GTPases. The Rap2 proteins can also be involved in cell signaling, as they can mediate activation of phospholipase-Cε.
Rap2a
Small Rap2a GTPase can be farnesylated. It is ubiquitously expressed, with a higher level in brain and hematopoietic tissues. Kinase MAP4K4 interacts with Rap2 to enhance activation by MAP4K4 of Jun N-terminal kinase [914].
Rap2b
Small Rap2b GTPase undergoes post-translational modifications. It can indeed be polyisoprenylated and palmitoylated. Small GTPase Rap2b is involved in platelet activation.129 Like Rap1b that is activated upon thrombin and ADP stimulation mainly via Ca
-dependent signaling and the Gi–PI3K pathway (Gi-coupled P2Y12 receptor), respectively, Rap2b is stimulated by Ca
, PKC, PI3K upon excitation of G-protein-coupled receptors by ADP and receptor tyrosine kinases by the collagen receptor platelet glycoprotein-6 (GP6) [915]. However, Rap1b rather than Rap2b seems to be directly regulated by thrombin receptors, whereas glycoprotein-6 stimulation preferentially leads to activation of Rap2b rather than Rap1b.


Rap2c
Small GTPase Rap2c is widely expressed (mainly in skeletal muscle, liver, digestive tract, bladder, prostate, uterus, and, to a lesser extent, in brain, kidney, pancreas, and bone marrow) [916]. Small Rap2c GTPase is the predominant type in circulating mononuclear leukocytes. It is also expressed in human megakaryocytes, but not in platelets [917].
9.3.10 Ras GTPases
Membrane-associated Ras130 and Rho GTPases activate intracellular pathways in response to extracellular signals for multiple functions. Activation of Ras by receptor Tyr kinases involves the binding of Ras to Ras-specific GEF Son of sevenless, which has been recruited to the plasma membrane. The binding is followed by nucleotide exchange with SOS agent. G-protein-coupled receptors can also initiate Ras signaling, using a SOS homolog, Ras guanine nucleotide-releasing factor-1 (RasGRF1), which, like SOS, catalyzes Ras nucleotide exchange.
Signal transduction by growth factor receptors proceeds via recruitment to the cell cortex of cytosolic signaling effectors such as adaptor growth factor receptor-bound protein GRB2, which then attracts nucleotide-exchange factor Son of sevenless. Colocalization at the cell membrane increases the effect of effectors. The kinetics of Ras activation by SOS indeed changes according to Ras location. Signaling is more efficient when Ras is tethered to phospholipid membranes instead of being in cytosol, as the activity of SOS catalytic unit is about 500-fold higher when Ras is on membranes [918]. Protein SOS has 2 Ras-binding sites that can both be simultaneously occupied by membrane-bound Ras. Small GTPase Ras transiently binds to the active site for nucleotide exchange. Occupation of the second Ras-binding site of SOS stimulates nucleotide exchange of SOS allosterically, by causing conformational changes of active site and favoring Ras binding. Access to the allosteric site that anchors SOS catalytic unit to the membrane is controlled by SOS regulatory domain. Binding of RasGTP to the allosteric site leads to a positive feedback loop for Ras activation. The N-terminal segment of SOS can occlude the allosteric site, hence blocking membrane anchoring and preventing unchecked activation of Ras. On the other hand, the large increase in activity of SOS catalytic unit requires Ras binding to SOS allosteric site and conversion of RasGDP to RasGTP. Protein SOS responds depending on Ras membrane density, GTP loading, and membrane concentration of phosphatidylinositol (4,5)-bisphosphate. Agent SOS integrates these data to relieve its auto-inhibition with greater efficiency.
9.3.10.1 RAS Hyperfamily
Small Ras GTPases include Ras, Rap, Ral, and others [919]. Small GTPases Rho, Rac, and CDC42 are the 3 best known families of the RHO superfamily. Kinases of the RoCK family are effectors of Rho GTPases. Small Rac and CDC42 GTPases act via P21-activated kinases. Small Rho GTPases regulate cytoskeletal activity during cell motility, shape change, and contraction.131 Each RHO family is characterized by specific effects on the actin cytoskeleton. Agent Rho is involved in the formation of stress fibers, Rac of membrane ruffles and lamellipodia, and CDC42 of filopodia (radial unipolar bundles). Members of all RHO families also regulate the assembly of integrin-containing adhesion complexes, and thus cell–matrix interactions and cell adhesion.
9.3.10.2 Members of the RAS Family
Members of the Ras family constitute 2 subfamilies. A first subfamily includes Harvey hRas, Kirsten kRas,132 and neuroblastoma nRas, whereas related rRas isoforms are grouped in a second subfamily. In mammals, 3 RAS genes generate subfamily-1 Ras GTPases (hRAS, kRAS, and nRAS). The KRAS transcript can be alternatively spliced, thereby producing kRas4a and kRas4b subtypes.
9.3.10.3 Post-Translational Modifications
Post-translational modifications are aimed at enabling proper protein folding and localization, signal transmission, as well as protein degradation. Post-translational modifications of Ras GTPases direct them to various cellular membranes and, in some cases, modulate GTP–GDP exchange [920]. These modifications are either constitutive, such as irreversible farnesylation of the C-terminal CAAX motif,133 reversible palmitoylation,134 methylation, and proteolysis of a C-terminal propeptide,135 or conditional, such as reversible phosphorylation,136 peptidyl-prolyl isomerisation,137 mono- and diubiquitination,138 nitrosylation,139 adpribosylation, and glucosylation.
Membrane tethering and trafficking of small GTPases of the P21RAS subfamily other than kRas4b are regulated by reversible palmitoylation of Cys residues in their C-terminus [920].140 The covalent attachment of the acyl chain of a fatty acid to a protein is called protein acylation. Unlike farnesylation, palmitoylation is reversible. Reversibility is ensured by one or more thioesterases, such as acyl-protein thioesterase APT1. Both hRas and nRas undergo palmitoylation–depalmitoylation (acylation–deacylation) cycles for Ras transfer from (anterograde) and to (retrograde transport) the Golgi body. Palmitoylation is required for the transfer of hRas and nRas from endomembranes to the plasma membrane. Farnesylated Ras has only a slight affinity for membranes, but farnesylated and palmitoylated Ras has a much higher affinity [920]. Mono- and dipalmitoylation control Ras localization.
Cis–trans isomerization of a peptidyl-prolyl bond adjacent to a palmitate in hRAS acts as a molecular timer that regulates depalmitoylation and retrograde transfer.
Phosphorylation of kRas4b in its polybasic region allows kRAS4b dissociation from the plasma membrane. S-nitrosylation of Ras promotes guanine nucleotide exchange.
Mono- and diubiquitination of hRAS regulate its association with endosomes; kRas4b monoubiquitination enhances its activation. Ubiquitination regulates hRas transfer to and from endosomes. Effector of Ras GTPase Ras and Rab interactor RIn1 is required for RabEx5-dependent Ras ubiquitination.
9.3.10.4 Ras Signaling
Small Ras GTPases are involved in the regulation of cell growth, differentiation, and survival. They are encoded by genes that experience mutations in about 30% of cancers in humans.
The components of the P21RAS subfamily have a similar effector domain, which interacts with downstream pathway targets. Both Rap and Ras141 can bind the same effectors to regulate intracellular signaling events.
In the GTP-bound conformation, Ras binds to and activates its effectors, members of the RAF family, phosphatidylinositol 3-kinase, and members of the RalGEF family [922] (Table 9.29). Effector of Ras for small Ras-like GTPases RalA and RalB are the guanine nucleotide-exchange factors Ral guanine nucleotide-dissociation stimulator (RalGDS or RalGEF) and RalGDS-like protein. Other effectors include phospholipase-Cε,142 T-cell lymphoma invasion and metastasis GEF TIAM1,143 and Ras interaction/interference protein RIn1, afadin, and Ras association domain-containing family proteins (RASSF) [923].144
Set | Agents |
---|---|
GEFs | RasGRF, RasGRP, SOS |
GAPS | RasA1/2/3/4, RasAL, neurofibromin-1 |
Effectors | RalGEF, RASSF, RIn1, TIAM1 |
PI3K, PLCε, cRaf |
Small GTPases Ras, hRas and kRas (Table 9.30) in particular, are activated by most growth factors as well as integrins. Activated receptor Tyr kinases dock adaptors such as GRB2 that attract RasGEFs. The latter recruit and activate Ras GTPases. Other receptors can stimulate Ras GTPases via SRC family kinases.
Type | Location | Effectors |
---|---|---|
P21RAS subfamily | ||
hRas | Golgi, PM | cRaf, PI3K, RalGEF |
kRas | PM | cRaf, PI3K, RalGEF |
nRas | Golgi, PM | cRaf, RasGRP2 |
cRaf activation order: | kRas > nRas > hRas | |
RRAS subfamily | ||
rRas1 | bRaf, cRaf, PI3K, PLCε, RalGEF | |
rRas2 | PI3K | |
rRas3 | Raf, PI3K | |
cRaf activation | Weak |
Members of the P21RAS subfamily of small GTPases — hRas, nRas, and kRas (kRas4a–kRas4b) isoforms — mediate mitogenic signaling triggered by growth factor receptors, hence, when mutated, cancer. These Ras proteins participate in cell proliferation and migration via the Raf–MAP2K–ERK pathway, but not survival, at least in mouse embryonic fibroblasts [924].145 The Ras proteins mainly signal via the Raf–MAP2K–ERK, PI3K–PTen–PDK1–PKB, and RalGEF–Ral pathways [924].
In addition to RalGEF, Ras effectors involve other guanine nucleotide-exchange factors, such as TIAM1 and RalGDS. Agent TIAM1 activates small GTPases of the RHO and RAC families to regulate cell polarity, motility, and adhesion. Molecule RalGDS stimulates Ral GTPases that are primarily involved in membrane trafficking.
Additional effectors include components of the PLCε–PKC–Ca
cascade, afadin (or AF6 GEF) that regulates intercellular adhesions and can interact with cytoskeletal structures, Ras and Rab interactor RIn1, a GEF, and Ras association domain family (RASSF) proteins.

On the other hand, constitutively active GTPases, such as members of the RRAS subfamily and embryonic stem cell-expressed Ras (eRas or hRAS2), activate the Raf–ERK pathway.
Small Ras GTPases can influence ion channel activity (Sect. 11.4). Small GTPases Rap and Ras have opposite effects on atrial M2 muscarinic receptor-coupled K + channels (GIRK). Small GTPase Rap1 also antagonizes Ras action on voltage-gated Na + channels, NMDAGlu receptors and AMPAGlu receptors [925].
Tumor suppression by Ras results from cell cycle arrest and/or senescence that can exceed unlimited proliferation and/or transformation [923]. Yet, deregulated Ras signaling causes developmental disorders (cardiofaciocutaneous syndromes) characterized by accumulation of tumors and skeletal, cardiac, and visual abnormalities.
The RRAS subfamily includes rRas isoforms (rRas1–rRas3). Protein rRas146 antagonizes hRas signaling. In vitro, rRas enhances integrin-mediated cell adhesion, whereas hRas inhibits integrin activities. rRas promotes the differentiation of myoblasts, whereas hRas inhibits it. rRas regulates cell survival and integrin activity, particularly in the remodeling of blood vessels. In vivo rRas is mainly expressed by smooth muscle and endothelial cells [927]. In the absence of rRas, neointimal thickening in response to injury and tumor angiogenesis are increased, whereas rRas expression is greatly reduced in hyperplastic neointimal smooth muscle cells and angiogenic endothelial cells.
Specific interactions between isoforms of membrane-anchored Ras and their guanine nucleotide-exchange factors, GTPase-activating proteins, and effectors depends not only on distinct molecular structure (e.g., C-terminal hypervariable region that is modified by lipids, anchors the G domain to the membrane, and allows distinct exocytosis sorting, subcellular localization, including segregation into specific nanodomain in the plasma membrane),147 but also recognition of orientation of the G domain of small GTPase Ras isoforms with respect to the plane of the plasma membrane [928]. Orientation of the G domain of small hRas GTPase that differs from that of kRas, mRas (or rRas3), and nRas, is recognized by effectors cRaf and PI3Kα as well as scaffold galectin-1 (that create nanoclusters of active hRas).
9.3.10.5 Ras Localization, Post-Translational Modifications, and Signaling
Small Ras GTPases lodge in various intracellular membranes, in addition to the plasma membrane. Golgi-associated hRas can be activated after growth factor stimulation with kinetics that differ from that of hRas activation at the plasma membrane. Furthermore, hRas tethered to the endoplasmic reticulum is able to activate the extracellular signal-regulated kinase and protein kinase-B preferentially, whereas a Golgi-tethered hRas can activate predominantly the Jun N-terminal kinase [929]. Therefore, the subcellular localization of Ras influences the type of engaged effectors. The activation of hRas localized at given subcellular compartments may be mediated by specific guanine nucleotide-exchange factors with a restricted residence. The subcellular localization depends on adequate post-translational modifications, especially lipidation (Sect. 9.3.10.9).148
9.3.10.6 Membrane Ras Nanoclusters
Plasmalemmal nanodomains with specialized protein and lipid composition regulate not only cell transport (membrane rafts and caveolae), but also cell signaling. Small Ras GTPases — hRas, kRas, and nRas — either reside in plasmalemmal nanoclusters (bore of ∼ 9 nm; lifetime 0.5–1.0 s) or correspond to freely diffusing monomers. Small GTPases hRas and nRas are farnesylated and acylated, whereas kRas is only farnesylated for stable anchoring to the plasma membrane.
Plasmalemmal Ras clusters are more or less associated with cholesterol and cortical components of the cytoskeleton. Plasmalemmal kRas nanoclusters are cholesterol-independent and actin-dependent, whereas hRas nanoclusters are cholesterol-dependent and actin-independent. Yet, the link with elements of the cell membrane and cortex depends not only on Ras isoform, but also on its bound nucleotide. Plasmalemmal Ras-containing nanoclusters can actually be segregated into GTP- and GDP-loaded protein assemblies. Nanoclusters that contain hRasGDP depend on cholesterol and actin, but not hRasGTP nanoclusters. Nanoclusters that possess kRasGTP depend weakly on actin and do not depend on cholesterol. Assembling of hRasGTP and kRasGTP nanoclusters rely on scaffolds galectin-1 or -3, respectively. The higher the RasGTP concentration, the greater the number of nanoclusters in the plasma membrane.
In particular, the mitogen-activated protein kinase pathway is underpinned on nanoclusters of small GTPase Ras on the inner leaflet of the plasma membrane.149 Ras-containing nanoclusters are required for MAPK-based signaling; indeed, RasGTP nanoclusters recruit Raf and KSR–MAP2K–ERK complexes from the cytosol [591]. Signaling from ERK terminates by spontaneous disassembly of nanoclusters.
Plasmalemmal nanodomains aimed at signaling yield a local concentration of signaling components. They also allow segregation of signaling components inside and outside nanoclusters. These sites act as switches by recruiting and activating pathway effectors causing maximal output above a given threshold rather than eliciting a graded signaling. However, a graded response directly proportional to input can be provided by the formation and activation of nanoclusters.
Signal transmission deeply depends on Ras spatial organization, i.e., on transient Ras cluster tethered to the inner leaflet of the plasma membrane. The plasmalemmal concentration of specific signaling effectors increase the efficiency and specificity of signaling using the Ras–Raf–MAP2K–ERK module. A set of plasmalemmal nanodomains of Ras GTPases convert graded ligand inputs into adapted outputs of activated extracellular signal-regulated kinase [930]. The higher the ligand concentration, the larger the number of transient Ras clusters (linear relationship).
Activation of Ras in association with scaffold proteins such as galectins triggers the formation of signaling clusters that can then activate the MAPK module. The regulated motion of scaffold proteins can control the extent of Ras clusters and hence modulate MAPK signaling magnitude. The recruitment of MAPK components to nanoclusters during cluster lifetime not only locally increases the kinase concentration and favors activation, but also protects them from degradation by phosphatases.
Post-translational lipid processing (palmitoylation) of Ras allows its attachment to the cell membrane, a prerequisite to its activation. The addition of a farnesyl isoprenoid lipid is catalyzed by farnesyl transferase. This reaction is followed by proteolytic cleavage processed by Ras-converting enzyme-1 and carboxymethylation by isoprenylcysteine carboxymethyltransferase-1 [923]. Farnesylated kRas4b is transported directly to the plasma membrane, whereas hRas, nRas, and kRas4a undergo palmitoylation in the Golgi body.
9.3.10.7 hRas
Small hRas GTPase (Harvey rat sarcoma virus oncogene homolog) is encoded by the HRAS gene. It relays growth regulatory signals that are initiated from plasmalemmal receptors, as, upon activation by inducers, hRasGTP can interact with effectors. Ubiquitous hRas is predominantly localized to the inner leaflet of the plasma membrane.
The weak intrinsic GTPase activity of hRas is enhanced by association with GTPase-activating proteins, such as RasA1 (or p120RasGAP) to RasA4 and neurofibromin-1 [931]. Conversely, hRasGDP is converted into active hRasGTP when hRas links to guanine nucleotide-exchange factors, such as SOS1, SOS2, RasGRF1, RasGRF2, and RasGRP4.
Effectors of hRas include Raf isoforms (aRaf–cRaf), Ral guanine nucleotide-dissociation stimulator RalGDS (or RalGEF), RalGDS-like proteins RGL1 and RGL2, which bind hRasGTP, but not hRasGDP, Ras-responsive Ub ligase Impedes mitogenic signal propagation (IMP) that inactivates KSR scaffold, which couples activated Raf to MAP2K [932], RalGEFs AF6 and Ras and Rab interactor RIn1,150 Ras-association (RalGDS–AF6) domain-containing proteins RASSF1 and RASSF5, PI3Kc1α, PI3Kc1δ, and PI3Kc1γ subunits, and PLCε [931].
Small hRas GTPase decreases the activity of inward rectifierKIR2.1 channel (IRK1), as it promotes its internalization [925]. On the other hand, it elevates CaV3 channel functioning.
9.3.10.8 kRas
Small kRas GTPase (Kirsten rat sarcoma viral oncogene homolog) is encoded by the KRAS gene. (Mutations of the KRAS gene produce potent oncogenes.) Protein kRas is usually tethered to cell membranes via a C-terminal isoprenyl group. Like hRas and nRas, kRas is farnesylated, but does not contain a palmitoylatable cysteine. Phosphorylation of a serine residue causes kRas to target mitochondria. It can also reside in the endoplasmic reticulum and late endosomes [902].
Small GTPase kRas interacts with cRaf of the MAPK module, Ras-association domain-containing protein RASSF2, Ral guanine nucleotide dissociation stimulator (RalGDS or RalGEF), and PI3Kγ. In addition, small GTPase kRas increases activity of epithelial Na + channel (ENaC) via PI3K and PI(4)P5K enzymes.151
9.3.10.9 nRas
Small nRas GTPase (neuroblastoma rat sarcoma viral oncogene homolog) is encoded by the NRAS gene that generates to 2 main transcripts. Both hRas and nRas are continuously exchanged between 2 cellular pools, the Golgi body and plasma membrane, at a relatively fast rate; this spatial cycle that depends on reversible palmitoylation is superimposed on the temporal GTPase cycle. Palmitoylatable hRas and nRas proteins are farnesylated, hence weakly attached to membranes and able to rapidly diffuse throughout the cell. However, because hRas and nRas are reversibly palmitoylated (at cysteine residues close to the farnesylation site), the resulting additional hydrophobicity confers higher stability on cellular membranes [902]. Switching between high- and low-membrane-affinity associated with the palmitoylation–depalmitoylation cycle due to the action of palmitoyltransferases (for palmitoylation and repalmitoylation) and thioesterases (for depalmitoylation) together with localized acylation–deacylation events creates the spatial cycle. Depalmitoylation occurs throughout the cytoplasm by acyl protein thioesterase-1 (APT1). Due to its ubiquitous distribution within the cell, APT1 enzyme converts by depalmitoylation mislocalized palmitoylated Ras to the fast-diffusing solely farnesylated form.
9.3.10.10 rRas
Small rRas GTPases (related rat sarcoma viral oncogene homolog) are encoded by genes of the RRAS group (RRAS1–RRAS3). Proteins rRas2 and rRas3 are also termed TC21 and mRas (muscle rat sarcoma viral oncogene product homolog), respectively.
Small rRas GTPase interacts with Ras-association domain-containing protein RASSF5, RalGEF, adaptor NCK1 that links receptor Tyr kinases to Ras, anti-apoptotic molecule BCL2, and kinase aRaf [251]. Protein aRaf binds to MAP2K2, translocase of inner mitochondrial membrane TIMM44, EGF-containing fibulin-like extracellular matrix protein EFEMP1, PRP6 pre-mRNA processing factor PRPF6, a bridging factor between snRNPs in the spliceosome, and TH1-like of the NELF complex that interacts with the DSIF complex to repress transcriptional elongation by RNA polymerase-2.
9.3.10.11 Ras-Association Domain Family Members
Molecular switches Ras control cell proliferation, differentiation, motility, and apoptosis according to extracellular signals. Their effectors specifically bind to RasGTP GTPases. These effectors include not only Raf kinases of the MAPK module and PI3K, but also Ras-association (RA; RalGDS/AF6) domain153 family proteins (RASSF1–RASSF8) that act as tumor suppressors [936].
RASSF1
Seven different transcripts are generated (RASSF1a-RASSF1g) by differential promoter use and alternative splicings [936]. Ubiquitous RASSF1a and RASSF1c are the major isoforms. Subtype RASSF1b is expressed predominantly in hematopoietic cells. Four isoforms RASSF1d to RASSF1g are splice variants of RASSF1a protein. Transcripts RASSF1d and RASSF1e are expressed specifically in cardiac and pancreatic cells, respectively. Protein RASSF1 interacts with plasma membrane calmodulin-dependent calcium ATPase PMCA4b [936].
Isotype RASSF1a is a microtubule-binding and -stabilizing molecule that control genome stability during the cell division cycle, as it impedes activated Ras to cause genomic instability. Subtype RASSF1a localizes to microtubules in interphase, relocalizes to separated centrosomes during prophase, then to spindle fibers and poles during metaphase and anaphase, and finally to the midbody during cytokinesis [936]. Isoform RASSF1c has identical cellular locations as those of RASSF1a. Protein RASSF1a primes cell cycle arrest by engaging the checkpoint retinoblastoma proteins that regulates entry into S phase.
In addition, RASSF1a as well as RASSF5 may serve as detectors of pro-apoptotic signals initiated via the Ras pathway. Effectors RASSF1a or RASSF1c can bind scaffold Connector enhancer of kinase suppressor of Ras CnK1 that allows Ras to activate Raf kinase. Isoform RASSF1a can also connect to kinases STK3 and STK4 (Sect. 10.7), hence favoring CnK1-induced apoptosis [936]. Moreover, binding of RASSF1a to Modulator of apoptosis MAP1 provokes MAP1 association with BAX protein. Activation of BAX leads to cell death. Besides, RASSF1c is a partner of Death-associated protein DAP6 (DAXX).
RASSF2
Protein RASSF2, originally termed Rasfadin, possesses 3 isoforms (RASSF2a–RASSF2c), but only the RASSF2a transcript is translated [936]. Overexpression of RASSF2 causes apoptosis via caspase-3 activation and cell cycle arrest.
RASSF3 and RASSF4
The RASSF3 gene can produce 3 transcripts (RASSF3A–RASSF3C) by alternative splicing. Protein RASSF3a corresponds to RASSF3 that is longer than RASSF3b and RASSF3c isoforms [936]. Protein RASSF4 is broadly expressed in human tissues (heart, brain, lung, liver, skeletal muscle, pancreas, and placenta) [936].
RASSF5
Protein RASSF5154 possesses 2 isoforms RASSF5a and RASSF5b [936]. It associates with hRas and kRas as well as, with a comparable affinity, Ras-like GTPases, such as Rap1 and Rap2, as well as rRas, rRas2, and rRas3 (mRas). Like RASSF1, it impedes cell proliferation. It also modulates Ras signaling triggered by stimulated TCR via recruitment of active Ras to the plasma membrane and control of localization of caspase recruitment domain family member CARD11, a membrane raft-associated regulator that supports TCR-induced NFκB activation.
9.3.10.12 Small GTPases Ras and Cancer
Many cancers are associated with mutations in RAS genes that remain in the active (GTP-bound), oncogenic state. Mutations of the KRAS gene initiate and maintain tumor growth by stimulating the PI3K–PKB pathway that phosphorylates (activates) nitric oxide synthase NOS3 [937]. The latter synthesizes nitric oxide that promotes Ras nitrosylation and GTP binding of hRas and nRas needed for growth and maintenance of kRas-mutant tumors.
9.3.10.13 Small GTPases Ras and Immunity
Many GTPases of the RAS hyperfamily (Ras, Rap1a, CDC42, Rac1, Rac2, and RhoA) contribute to signal transduction primed by antigen receptors, costimulators, and cytokine and chemokine receptors to regulate the immune response [938]. Small Ras GTPase is activated by T-cell antigen and cytokine receptors (particularly IL2R and IL15R). It mediates some of preTCR signals that select thymocytes.156 In mature T lymphocytes, Ras targets transcription factors that regulate cytokine genes, such as ELk1, SRF, AP1, and NFAT factors.
In lymphocytes, Ras GTPases are regulated by RasA1 (or p120RasGAP) and phosphorylated adaptors such as docking proteins DOK1 and DOK2 on the one hand and the single RasGEF in lymphocytes SOS that tethers to GRB2 on the other [938].157 In addition, lymphocytes express Ras guanine nucleotide-releasing protein (RasGRP) that associates with diacylglycerol.
Among members of the RHO superfamily of the RAS hyperfamily, CDC42 regulates interactions between T lymphocytes and antigen-presenting cells and chemokine-stimulated lymphocyte migration [938]. Other member RhoA controls thymocyte development as well as integrin-mediated cell adhesion. Small Rac1 GTPase is involved in T-cell activation. The RacGEF Vav1 is phosphorylated in response to engagement of antigen receptor or costimulator CD28 with their ligands.158 Both Rac1 and Rac2 modulate distinct T-cell functions. Only Rac1 can direct thymocytes from positive to negative selection, whereas Rac2 causes apoptosis and depletion of CD4 + , CD8 + double-positive lymphocytes [938].
Small Rap1a GTPase is rapidly activated after antigen receptor ligation in both B and T lymphocytes via Fyn kinase that promotes formation of a complex with adaptors CBL and CRKL and RapGEF1 [938]. Small GTPase Rap1a can also be activated via increase in intracellular calcium concentration, diacylglycerol release, and cAMP synthesis. It can antagonize Ras, and, conversely, can be antagonized by CD28 agent. However, Rap1A and Ras may be involved in different signaling pathways, so that active Rap1A does not interfere with Ras signaling and T-cell activation [938].
9.3.11 RHEB GTPase
Small GTPase Ras homolog enriched in brain RHEB is involved in cell growth and nutrient uptake. Tuberous sclerosis complex TSC2 is a GTPase-activating protein that deactivates RHEB GTPase.159 Proteins TSC1 and TSC2 form a complex that inhibits phosphorylation of S6K and 4eBP1 proteins. The PI3K–PKB axis activates RHEB by inhibiting TSC1 or TSC2. In response to nutrients and cellular energy status, RHEB then activates Ser/Thr protein kinase target of rapamycin that phosphorylates kinase S6K and translational repressor initiation factor-4E for eukaryotic translation (eIF4e)-binding protein 4eBP1 [939]. In addition, RHEB promotes the formation of late endosomes [940]. It regulates endocytic trafficking pathway independently from the mTOR pathway.
9.3.12 Superfamily of RHO GTPases (CDC42, Rac, and Rho)
The RHO superfamily of small GTPases encompasses 3 major families: RHO (RhoA–RhoC), RAC (Rac1–Rac3) and CDC42 families. Each of these monomeric GTPases controls the formation of a distinct cytoskeletal constituent.160 In fact, families of the RHO superfamily include: (1) RHO family with its 3 isoforms; (2) RAC family also with 3 isoforms and RhoG; (3) CDC42 family with 2 isoforms; (4) RND family with 3 isoforms (Rnd1, or Rho6, Rnd2, or Rho7, and Rnd3, or RhoE; (5) MIRO family with 2 isoforms (Miro1–Miro2, or RhoT1–RhoT2); as well as families that contain (6) RhoJ and RhoQ; (7) RhoU and RhoV; (8) RhoD, (9) RhoF, and (10) RhoH GTPase.
Major activators of members of the RHO superfamily comprise G-protein-coupled receptors, receptor Tyr kinases, adhesion molecules, and mechanical stresses (tension, compression, and shear). These signaling mediators are, indeed, activated upon stimulation by hormones, growth factors, cytokines,161 adhesion molecules, especially integrins162 and cadherins, to reorganize the actin cytoskeleton and, hence, regulate vesicle release, transfer, and uptake, activate NADPH oxidase, contribute to gene transcription, execute cell division cycle or apoptosis, control morphogenesis, and participate in platelet activation and aggregation during blood coagulation as well as neutrophil activation and phagocytosis in immune response, among other biological events. Small GTPases CDC42, Rac1, and RhoA modulate dynamics of contractile actin–myosin-2 filaments via their associated kinases and other target proteins (Tables 9.31 to 9.33).
Table 9.31
Targets of small GTPases of the Rho superfamily (Part 1; Sources: [941–943]; ACK: activated CDC42-associated kinase; ARP: actin-related protein; ERK: extracellular signal-regulated kinase; ERM: ezrin, radixin, and moesin; JNK: Jun N-terminal kinase; IQGAP: IQ motif-containing GTPase-activating protein; MLC: myosin light chain; MLK: mixed lineage kinase; MRCK: myotonic dystrophy kinase-related CDC42 binding kinase; Par: partitioning defective protein; Par6–Par3–aPKC: Par6–Par3–atypical PKC complex; PIP2: phosphatidylinositol (4,5)-bisphosphate; PIP5K: phosphatidylinositol 4-phosphate 5-kinase; PKB/C: Protein kinase-B/C; WASP: Wiskott-Aldrich syndrome protein; WIP: WASP-interacting protein).
Effector | Effect |
---|---|
CDC42 | |
ACK1/2 | Focal adhesion formation |
Binders of Rho GTPases | Cytokinesis via septins |
Coatamers | Membrane trafficking |
IQGAP | Intercellular adhesion, microtubule orientation |
MRCK | Promotion of cell contraction and focal adhesion |
LIMK2 phosphorylation (activation) | |
P21-activated kinases | LIMK phosphorylation (cofilin inactivation) |
(PAK1–PAK4) | Stathmin phosphorylation (microtubule stability) |
Triggering of cRaf–MAP2K1–ERK1 axis | |
Stimulation of MLK–JNK pathway | |
(activation of transcription factors Jun, Fos, Elk1) | |
LIMK1 activation (PAK1/4) | |
MLCK inhibition (PAK1) | |
MRCK phosphorylation | |
Par6–Par3–aPKC | Cell polarity (tight junction) |
Reorientation of the microtubule organizing center | |
during cell migration | |
PI3K | Stimulation of RacGEFs, |
WASP (actin polymerization); PKB activation | |
PIP5K | PIP2 production |
(ERM processing for subsequent phosphorylation) | |
WASP, WIP | Formation of filopodia via PI(4,5)P2 and ARP2/3 |
Table 9.32
Targets of small GTPases of the RHO superfamily (Part 2; Sources: [941–943]; ARP: actin-related protein; ERK: extracellular signal-regulated kinase; JNK: Jun N-terminal kinase; IQGAP: IQ motif-containing GTPase-activating protein; MLK: mixed lineage kinase; p140SRA1: specifically Rac1-associated protein; Par: partitioning defective protein; Par6–Par3–aPKC: Par6–Par3–atypical PKC complex; PIP2: phosphatidylinositol (4,5)-bisphosphate; PIP5K: phosphatidylinositol 4-phosphate 5-kinase; PKB/C: Protein kinase-B/C; WASP: Wiskott-Aldrich syndrome protein; WAVe: WASP family verprolin homology domain-containing protein).
Effector | Effect |
---|---|
Rac | |
IQGAP | Intercellular adhesion, microtubule orientation |
NADPH Oxidase | Synthesis of reactive oxygen species |
(activation of nuclear factor-κB) | |
p140SRA1 | Membrane ruffling |
P21-activated kinases | LIMK phosphorylation (cofilin inactivation) |
(PAK1–PAK4) | Stathmin phosphorylation (microtubule stability) |
Triggering of cRaf–MAP2K1–ERK1 axis | |
Stimulation of MLK–JNK pathway | |
(stimulation of AP1-dependent gene expression) | |
Par6–Par3–aPKC | Cell polarity (tight junction) |
Partner of Rac1 (POR1) | Membrane ruffling |
Phospholipase-D | Phosphatidic acid production |
PI3K | Activation of WASP and PKB |
PIP5K | PIP2 production |
WAVe | Activation of the ARP2–ARP3 complex |
(actin polymerization) |
Table 9.33
Targets of small GTPases of the RHO superfamily (Part 3; Sources: [941–943]; ARP: actin-related protein; CRIK: citron Rho-interacting kinase; ERM: ezrin, radixin, and moesin; MBS: myosin-binding subunit; MLC: myosin light chain; NHE1: sodium–hydrogen exchanger; PIP2: phosphatidylinositol (4,5)-bisphosphate; PIP5K: phosphatidylinositol 4-phosphate 5-kinase; WASP: Wiskott-Aldrich syndrome protein).
Effector | Effect |
---|---|
Rho | |
Binders of Rho GTPases | Cytokinesis via septins |
CRIK | Cytokinesis, cell division cycle progression |
Diaphanous | Microtubule stability, actin polymerization |
GDIA | |
Kinectin | |
LIMK1/2 | Cofilin phosphorylation (inactivation) |
MLC kinase | Actin–myosin assembly, cell contraction |
MLC phosphatase | Actin–myosin disassembly, cell relaxation |
(MBS PP1δ) | |
NET1 | Guanine nucleotide-exchange factor |
Phospholipase-D | Phosphatidylcholine hydrolysis |
PIP5K | PIP2 production (ERM processing for |
phosphorylation) | |
Protein kinase-N | Endosome-mediated transfer |
(PKN1 [PRK1]–PKN2) | |
RoCK | Phosphorylation of MLC and MBS |
(inactivation of myosin phosphatase) | |
LIMK2 phosphorylation (cofilin inactivation) | |
ERM and NHE1 phosphorylation | |
(actin–membrane link) | |
Phosphorylation of vimentin and desmin | |
(reorganization of intermediate filaments) | |
Rhophilins-1/2 | Rhophilin-2-induced disassembly of stress fibers |
Rhotekins-1/2 | Lymphocyte function (rhotekin-2) |
WASP | Activation of the ARP2–ARP3 complex |
(promotion of actin polymerization) |
Multiple Rho-associated Ser/Thr kinases interact with and are regulated by their partner GTPases: citron Rho-interacting kinase (CRIK), mixed lineage kinases (MLK) of the MAPK module (MAP3K9–MAP3K11 and MAP3K15, in addition to MLK4), myotonin-related CCD42-binding kinase (MRCK), P21-activated kinase (PAK), protein kinase novel (PKN), and RoCK kinase. All of these kinases can dimerize [943].
Myosin phosphorylation depends mainly on the balance of 2 enzymes: Ca
-dependent regulatory myosin light-chain kinase and phosphatase. Phosphorylation of myosin-2 light chain provokes its interaction with actin and subsequent activation of myosin ATPase to cause cell contraction. Kinase RoCK phosphorylates MBS subunit (Thr697, Ser854, and Thr855) to prevent dephosphorylation and also phosphorylates regulatory myosin light chain (Ser19). In addition, it phosphorylates LIMK2 (Thr505) that phosphorylates (inactivates) cofilin, an actin-depolymerizing agent, to promote actin–myosin-2 assembly.

Among Rho interactors and indirect effectors, activated proteins ezrin, radixin, and moesin (ERM) connect directly to adhesion molecules, such as intercellular adhesion molecules ICAM1 to ICAM3 and indirectly with other integral membrane proteins such as Na + –H + exchanger NHE3 (SLC9a3) via NHE regulatory factor (NHERF) [941].163 Activated ERM proteins also bind to RhoGDI to activate Rho GTPase that, in turn, can stimulate ERM proteins beneath the plasma membrane (positive feedback). Kinectin (Ktn1), a Dia-related protein, interacts with RhoA and CDC42 GTPases. Citron Rho-interacting kinase (CRIK or STK21) is a RoCK-related kinase that operates in cytokinesis and other aspects of cell cycle progression. Diaphanous proteins (Dia1–Dia3) mediate both actin polymerization and stabilization of microtubule plus-ends during cell migration.
P rotein kinases PKN1 and PKN2164 are Rho effectors involved in endosomal trafficking. Enzyme PKN also phosphorylates (inactivates) intermediate filament proteins, such as neurofilament, vimentin, and glial fibrillary acidic protein, as well as microtubule-associated protein Tau [944]. Widespread PKN has a catalytic domain homologous to that of members of the PKC family and a unique regulatory region. It can bind to small GTPase RhoA to be activated [941] as well as RhoB [943].
Ubiquitous PKNα (PKN1) is expressed especially in central neurons. It is concentrated in a region of the endoplasmic reticulum and its derived vesicles localized to the apical compartment of the juxtanuclear cytoplasm, as well as Golgi body, late endosomes, multivesicular bodies, and secretory vesicles, in addition to cell nucleus [944]. It phosphorylates myristoylated alanine-rich C kinase (MARCK) and vimentin. It can be stimulated by phosphoinositide-dependent protein kinase PDK1 as well as arachidonic and linoleic acids, cardiolipin, phospholipids, such as phosphatidylinositol (4,5)-bisphosphate and (3,4,5)-trisphosphate, and lysophospholipids, such as lysophosphatidic acid and lysophosphatidylinositol. Isoform PKNα interacts with RhoA, RhoB, RhoC, and Rac1 GTPases [941, 943]. It sustains the GTPRhoA form [944]. In addition, PKNα is able to bind to actin crosslinking protein α-actinin as well as actin-cytoskeleton proteins, such as caldesmon and Gactin.
Isoform PKNβ (PKN3) attaches to GTPase-activating proteins for RhoA GRAFs (GTPase regulator associated with focal adhesion kinase) and CDC42 [944].
Isozyme PKNγ (PKN2) tethers to NCK and GRB4 adaptors [944]. It can associate with MAP3K2 kinase. However, MAP3K2 may act as a scaffold to regulate PKNγ activity, similarly to PKCα that interacts with and activates PLD independently of its kinase activity. It can also phosphorylate PDK1 for maximal PKB activation, but can inhibit PDK1 autophosphorylation, hence impeding the ability of PDK1 to phosphorylate PKCζ and PKCδ, as well as S6K and SGK [944]. In addition, it interacts with protein Tyr phosphatase PTPn13 to modulate activity of the actin cytoskeleton.
Alias PKN can be used to designate a protein kinase novel (PKN1–PKN3 or PKNα–PKNγ), some being also called PKC-related kinase (PRK1/2), and P21-activated kinases. Some authors describe both types of kinases in a review on Rho-associated kinases [943].
The P21-activated kinases (PAK1–PAK3; i.e., group-1 PAKs) interact with activated Rac1 to Rac3 and CDC42, as well as other small GTPases, such as RhoQ, RhoU, and RhoV proteins, but neither other Rho types (RhoA to RhoC, RhoE, and RhoG) nor other RAS superfamily members [943]. They require autophosphorylation to become active. They are also implicated in transcription via mitogen-activated protein kinase cascades. Membrane recruitment of PAK1 via NCK and GRB2 adaptors stimulates its kinase activity, possibly by PDK1 kinase. Kinases PAK1 to PAK3 complex with focal adhesion-associated CDC42–Rac1GEF Rho(Arh)GEF6 protein.
Non-conventional members of group-2 P21-activated kinases include ubiquitous PAK4, brain-enriched PAK5, and androgen receptor-interacting PAK6. Isoform PAK4 binds to CDC42GTP and, to a much lesser extent, RacGTP. Binding of CDC42GTP to PAK4 does not stimulate kinase activity, but instead causes PAK4 translocation to the Golgi body [943]. Similarly, linkage between CDC42 or Rac and PAK5 or PAK6 does not enhance PAK activity.
Mixed lineage kinases (MAP3K9–MAP3K11, MLK4, and MAP3K15) influence cytoskeletal organization (Sect. 6.5.3) [943]. Both MLK1 to MLK4 are auto-inhibited. Kinase MAP3K11 binds activated CDC42 and Rac1 activators to dimerize and autophosphorylate. Ubiquitous myotonin-related CDC42-binding kinase (MRCK), an effector of CDC42 and Rac GTPases, targets both RoCK and LIMK2 kinases. Myotonic dystrophy-associated protein kinase DMPK1 is not regulated by small GTPases [943].
9.3.13 Family of RHO GTPases (RhoA, RhoB, and RhoC)
Table 9.34
Small Rho GTPases and their effectors (Source: [889]; CRIK: citron Rho-interacting, serine/threonine kinase STK21; Dia: diaphanous formin; FHoD1: formin homology-2 domain-containing protein). RhoA is more closely related to RhoC than RhoB. Whereas RhoA and RhoC promote cell growth, RhoB most often inhibits cell growth. Rho GTPases link to a large number of proteins, such as members of the Rho-associated kinase family and mammalian Diaphanous proteins. They participate in gene expression, cell morphology control, smooth muscle contraction, formation of stress fibers and focal adhesions, cytokinesis, cell cycle progression, axon guidance and extension, vesicular transfer, tissue development, and cell transformation.
Type | Effectors |
---|---|
RhoA | RoCK1/2, PAK1, CRIK, Dia1/2 |
RhoB | RoCK1/2, PAK1, CRIK, Dia1/2 |
RhoC | RoCK1/2, PAK1, Dia1/2, FHoD1 |
RhoD | Dia3 |
RhoF | Dia2 |
Members of the RHO superfamily are prenylated and methylated at their C-termini and thus can reside in cell membrane. Similarly to other Ras-related GTPases, members of the RHO superfamily are inefficient GTPases, as they tightly bind both GTP and GDP and slowly catalyze GTP hydrolysis. The rates of GTP hydrolysis and guanine nucleotide release are greatly accelerated by GTPase-activating proteins and guanine nucleotide-exchange factors, respectively. Therefore, Rho GTPase functions primarily as a molecular switch. Specific GAPs favor GTP hydrolysis, as they transfer the switch into its inactive GDP-coupled form. Specific GEFs assist GTP binding and displace the switch into its active GTP-charged form that can bind effectors to generate and/or transmit signals.
9.3.13.1 RhoA
Small RhoA GTPase is the prototypical member of the RHO family that is involved in multiple signal transduction pathways, as it associates with a large number of proteins. It undergoes adpribosylation by C3 ADPribosyltransferase [945]. Cytoskeleton assembly and actin and myosin dynamics are regulated by the balance between RhoA, Rac1, and CDC42 activities.
Small RhoA GTPase binds to RoCK1 and RoCK2 kinases that phosphorylate and activate LIMK1 kinase that, in turn, phosphorylates (inactivates) cofilin, hence inhibiting depolymerization of filamentous actin. The RoCK kinases also enhance myosin activation, as they phosphorylate regulatory myosin light chain and inhibit MLC phosphatase (Sect. 5.2.14). In mammals, Diaphanous-related formins (DRF) that include Dia1 to Dia3, Disheveled-associated activators of morphogenesis DAAM1 and DAAM2, and FRL1 to FRL3 that bind and bundle actin filaments are direct effectors of RHO family proteins. They are auto-inhibited by intramolecular interactions. Binding of GTP to Rho relieves auto-inhibition. Isoform Dia1 specifically binds to and is activated by RhoA to RhoC, whereas Dia2 and Dia3 are activated by RhoA to RhoC, CDC42, and RhoD [945]. Formins initiates actin filament assembly, as they bind and stabilize actin dimers or trimers. Diaphanous initiates the assembly of new actin filaments from actin monomers, extension of non-branching actin filaments, and formation of thin actin stress fibers. Effectors RoCK kinases and Diaphanous cooperate in Rho-induced actin reorganization to create actin fibers of various thicknesses and densities according to the balance of RoCK and Dia activities. On the other hand, RoCK antagonizes Dia in Rho-dependent Rac stimulation via Src activation and formation of the CAS–CRK–DOCK180 complex [945].
RhoA and Cell Migration
In migrating cells, CDC42 and Rac1 operate at the leading edge, in filopodia and lamellipodia, respectively, whereas RhoA stimulates contractile actin–myosin filaments in the cell body and at the cell rear to induce tail retraction as well as in lamellipodia and membrane ruffles, in which RhoA associates with Dia1 to control actin assembly (Vol. 2 – Chap. 6. Cell Motility). The Rho–Dia pathway regulates cell polarity and focal adhesion turnover, as it localizes adenomatous polyposis coli protein and Src kinase at their respective sites [945].
RhoA and Phospholipids
Small RhoA GTPase associates with phospholipase-D that generates phosphatidic acid. The latter activates PI(4)P 5-kinase that locally produces PI(4,5)P2. Small RhoA GTPase also connects to PI(4)P 5-kinase. Synthesized PI(4,5)P2 regulates actin-binding proteins, such as cofilin and gelsolin [945]. Phosphatidylinositol 4-phosphate 5-kinase-1γ associates with N-cadherin-mediated intercellular adhesions owing to activated RhoA GTPase. Agent PI(4,5)P2 regulates intercellular adhesion strength via actin-binding gelsolin [946]. In addition, RhoA links toKV1.2 channel subunit. It also heightens the activity of ENaC channels.
RhoA and the Cell Cycle
Small GTPase contributes to the control of cell cycle progression, as it regulates cyclin-dependent kinase inhibitor and cyclin-D1 concentrations [945]. During G1 phase, the RhoA-RoCK pathway prevents the activity of CDK inhibitors CKI1a and CKI1b via adhesion-dependent signaling and causes sustained extracellular signal-regulated kinase stimulation that controls expression of cyclin-D1 and repression on Rac and CDC42 signals. In addition, the RhoA–Dia pathway promotes ubiquitin-mediated degradation of CKI1b, thereby stimulating the G1–S transition. Last, but not least, RhoA and its effectors CRIK kinase and Dia2 actin nucleator colocalize to the cleavage furrow. Small GTPase RhoA is activated by RhoA-GEF epithelial-cell transforming gene ECT2 at the cell equator to form the contractile ring of actin–myosin-2 filaments that cleaves the cell into 2 daughter cells. Simultaneously, at the midbody during cytokinesis, Aurora-B colocalizes with RhoA and RacGAP1 and phosphorylates RacGAP1 (Ser387) to focus RhoA activity only to the cleavage furrow.
RhoA and Vesicular Transfer
Both RhoA and RhoB are involved in the regulation of exo- and endocytosis, as they stimulate actin dynamics via Wiskott-Aldrich syndrome protein and WASP family verprolin homologous proteins [945].
RhoA and Epi- and Endothelium Integrity
Cadherins suppress RhoA activity partly by increased RhoGAP35 activity. Catenin-δ1 not only interacts with cadherins to prevent their endocytosis, but also inhibits RhoA to avoid disruption of apical junctions and maintain epithelial architecture [945]. Ezrin–radixin–moesin proteins promote cortical actin assembly and cell polarity. They link transmembrane proteins to apical actin cytoskeleton of epithelial cells. A negative feedback exists between activated ERM and RhoA GTPase [945]. Small RhoA GTPase causes formation and contraction of actin stress fibers, hence endothelial hyperpermeability once stimulated by vascular epidermal growth factor, angiopoietin-1 and -2, and thrombin.
Regulation of Activity
Activity of RhoA is governed by (1) activation by GEFs and deactivation by GAPs; (2) adpribosylation (inactivation); (3) prenylation (geranylgeranylation) for interactions with membranes as well as kinesin-associated protein KIFAP3, RhoGDIs, and GEFs; (4) phosphorylation by protein kinase-A166 and dephosphorylation167 that regulates RhoA activity and location; (5) methylation; (6) ubiquitination for degradation; and (7) crosstalk with Rac or Ras GTPase pathways.168
Activity of RhoA can be regulated by angiopoietin-2, bombesin, chemoattractants, lisophosphatidic acid, sphingosine-1-phosphate, thrombin, Wnt morphogens, as well as hepatocyte, platelet-derived, transforming- (TGFβ), and vascular endothelial growth factors, and plexin-B and EPHa ligands via particular GEFs linked to G-protein-coupled receptors and receptor Tyr or Ser/Thr kinases [945]. Cadherins, integrins, fibronectins, and T-cell receptors also influence RhoA activity possibly via focal adhesion kinases that phosphorylate various GEFs.
9.3.13.2 RhoB
Small RhoB GTPase localizes to endocytic vesicles and regulates endocytosis, especially receptor internalization. It influences nuclear transport of protein kinase-B in endothelial cells.
9.3.13.3 RhoC
Small RhoC GTPase favors cell migration, particularly metastasis of tumor cells.169 It indeed contributes to the regulation of actin stress-fiber assembly and focal adhesion formation. It interacts with numerous affectors and effectors: (1) RhoC-GEFs include Scambio and RhoGEF5 (Tim); (2) RhoC-GAPs p50RhoGAP, p190RhoGAP, Myr5, and RhoGAP26; (3) RhoC-GDIs RhoGDIα, RhoGDIβ, and RhoGDIγ. Its effectors comprise phospholipase-Cε, scaffold rhotekin (inhibitor of GTPase activity of Rho), rhophilin-1 (Rho GTPase-binding protein), Rho-associated kinase RoCK1/2, Rho-interacting Ser/Thr kinase-21 Citron (STK21), Diaphanous Dia1, and PAK1 [947].
9.3.14 Atypical RhoBTB GTPases
Small RhoBTB GTPases (BTB: Broad complex, Tramtrack, and Bric-à-brac) constitute a family of atypical GTPases within the RHO superfamily. In addition to the GTPase domain (most often non-functional), RhoBTB is constituted by a tandem of 2 Bric-à-brac domains.
In humans, the RhoBTB family includes 3 isoforms (RhoBTB1–RhoBTB3). The RhoBTB1 and RhoBTB3 isoforms are ubiquitous, but with different distribution patterns [948]. Subtype RhoBTB1 is produced at high levels in the kidney, skeletal muscle, stomach, placenta, and testis, and RhoBTB3 in the nervous system, heart, pancreas, placenta and testis. On the other hand, RhoBTB2 is expressed at much lower levels than the other 2 isoforms.
Small GTPases RhoBTB can recruit cullin-3, a scaffold of ubiquitin ligases [949]. Once RhoBTB2 is bound to cullin-3, the latter regulates RhoBTB2 level by ubiquitinating RhoBTB2 directly for degradation [950]. The RhoBTB proteins may participate in cell cycle regulation and vesicle transport, as they tag substrates for degradation by 26S proteasome.
9.3.15 Classical RhoD GTPase
Small RhoD GTPase regulates early endosome movement between the cell cortex and central region. A splice variant of human Diaphanous Dia2c specifically binds to GTPase RhoD and is recruited onto early endosomes. Together with Dia2c, RhoD aligns early endosomes along actin filaments and reduces their motility, once Src kinase has been recruited and activated [951]. Small RhoD GTPase antagonizes RhoA, as RhoD causes loss of stress fibers and focal adhesions, whereas RhoA favors formation of actin stress fibers and associated focal adhesions [952].
9.3.16 Classical RhoF GTPase
Small RhoF GTPase (Ras homolog gene family, member F [in filopodia], also named RIF) is expressed in lymphocyte subpopulations. Cells of B-cell origin expressed higher RhoF levels than their T-cell counterparts [953]. It is upregulated in transformed follicular lymphoma, the most common form of non-Hodgkin lymphoma.
In addition to filopodium formation caused by GTPase CDC42,170 GTPase RhoF induces actin-rich filopodia that contain parallel actin filaments bundled by actin crosslinking proteins and help cells to sense their environment. RhoF-mediated filopodium formation occurs via Diaphanous-related formin Dia2 [954].
9.3.17 Atypical RhoG GTPase
Small GTPase RhoG shares a significant degree of sequence identity with Rac1 and CDC42 [955]. Protein RhoG not only accumulates in the perinuclear region, but concentrates in plasmalemmal spots. Constitutively active RhoG causes cytoskeletal changes similar to those generated by a simultaneous activation of Rac1 and CDC42 (formation of microvilli, ruffles, lamellipodia, filopodia, and partial loss of stress fibers). Small GTPase RhoG controls a pathway that requires microtubules and activates Rac1 and CDC42 independently of their stimuli, such as PDGF or bradykinin [955]. Monomeric GTPase RhoG is activated at the transcriptional level in the mid-G1 phase in fibroblasts [956].
9.3.18 Atypical RhoH GTPase
Small RhoH GTPase is predominantly expressed in hematopoietic cells. Monomeric RhoH has low or no GTPase activity and is thus constitutively bound to GTP [831]. Small GTPase RhoH hinders Rac recruitment to the plasma membrane. Protein RhoH precludes proliferation and homing of murine hematopoietic progenitor cells. It is needed to keep leukocytes in a resting, non-adhesive state [957]. It also intervenes in signal transduction during T-cell development.
Activity of RhoH is regulated by tyrosine phosphorylation (ITAM-like motif). In addition, RhoH C-terminal CAAX box is targeted by farnesyl transferase and geranylgeranyl transferase. Isoprenylation of RhoH and translocation yield further elements for fine-tuned signaling [957]. Small GTPase RhoH is used by the NFκB, PI3K, and MAPK pathways.
9.3.19 Atypical RhoJ GTPase
Small RhoJ GTPase171 is related to CDC42 and RhoQ (or TC10). It localizes to the plasma membrane and early endosomes [958].
Receptor-dependent endocytosis of transferrin remains unaffected in the absence of endogenous RhoJ, but transferrin accumulates in Rab5 + uncoated endocytic vesicles and fails to reach early endosome and pericentriolar recycling endosomes. Monomeric GTPase RhoJ is thus needed for clathrin-dependent endocytosis of receptors via early endosomes and direct recycling without accumulating in perinuclear recycling endosomes.
Small RhoJ GTPase participates in the early stage of adipocyte differentiation, probably linked to the peroxisome proliferator-activated receptor PPARγ pathway [959]. It regulates the expression of specific genes.
9.3.20 Atypical RhoQ GTPase
Small Ras-like RhoQ GTPase172 is a unusual member of the RHO superfamily. Monomeric RhoQ GTPase with 2 RhoQ homologous transcripts belongs to Ras-related teratocarcinoma (TC)-associated proteins, such as Ran (TC4), rRas2 (TC21), Rac1 (TC25), and Rab11. Small RhoQ GTPase is predominantly located at the plasma membrane and endosomes due to post-translational prenylation [960].173 It colocalizes with caveolin and flotillin at the plasma membrane.
Protein RhoQ regulates various cellular processes, such as actin cytoskeletal organization and cell shape, intracellular protein transport, signal transduction associated with the mitogen-activated protein kinase module, and cell growth and proliferation. Ubiquitous RhoQ [960]: (1) promotes formation of filopodia; (2) activates Jun N-terminal kinase; (3) stimulates nuclear factor-κB- and serum response factor-dependent transcription; and (4) synergizes with activated Raf kinase and contributes to full hRas action in cell transformation. Small RhoQ GTPase interacts with actin-binding and filament-forming profilin.
Small RhoQ GTPase binds to several signaling effectors, such as mixed lineage kinase MLK2 (MAP3K10), CDC42-binding protein kinase-α (a.k.a. myotonic dystrophy-related CDC42 kinase [MRCK]), P21-activated protein kinases, the Borg family of CDC42 effector proteins (CDC42EP), partition-defective homolog Par6, and nWASP [961].
Small RhoQ GTPase intervenes in intracellular protein transport. Upon insulin stimulation, RhoQ regulates assembly and fusion of vesicles responsible for GluT4 transporter exocytosis to the plasma membrane [962]. Protein RhoQαGTP (but not RhoQβ) promotes insulin-stimulated GluT4 translocation. Active RhoQGTP fastens to Exo70, a subunit of a proteic complex that tethers vesicles to sites of secretion. Protein Exo70 is involved in GluT4 transfer. Whereas CDC42-interacting protein CIP4 mediates the association of WASP with microtubules [963], RhoQ-interacting protein CIP4 is recruited to the plasma membrane in response to insulin to enhance GluT4 translocation [964]. Phosphoinositides contribute to GluT4 exocytosis. In fact, GluT4 translocation requires activation not only of RhoQ, but also phosphatidylinositol 3-kinase, as phosphatidylinositol (3,4,5)-trisphosphate serves in GluT4 translocation to the plasma membrane. On the other hand, phosphatidylinositol 3-phosphate is produced at plasmalemmal rafts upon stimulation by insulin via RhoQ to be incorporated into endosomes [965].
Protein RhoQ inhibits a cystic fibrosis transmembrane conductance regulator-interacting protein, CFTR-associated ligand (CAL), that mediates CFTR degradation in the lysosome, hence raising plasmalemmal CFTR density [966]. Active RhoQGTP redistributes CAL from the Golgi body to the plasma membrane, where it cannot influence CFTR transport to lysosomes for degradation. Monomeric RhoQ recruits effectors, such as CDC42-interacting protein, to the plasma membrane.
Small RhoQ GTPase is activated by RapGEF1 and inactivated by RhoGAP1 and RhoGAP35 [967]. It interacts with α, β, and γ isoforms of P21-activated kinases. It is also able to bind to mixed lineage kinase MLK2 (MAP3K10) that links to huntingtin as well as carrier IQGAP1 involved in neurite outgrowth. It tethers to nWASP that activates the ARP2–ARP3 complex as well as profilin that converts globular into filamentous actin and myotonic dystrophy kinase-related CDC42-binding kinase (MRCK; also known as CDC42-binding protein kinase CDC42BPα), a Ser/Thr kinase. Small GTPase RhoQ can recruit PKCζ and -λ to the plasma membrane in adipocytes. Activated RhoQ stimulates transcription factors or promoters, such as that of the JNK–Jun axis, as well as SRF, cyclin-D1, and NFκB promoters [967].
Binder of Rho GTPases (BORG) are CDC42 and RhoQ GTPase-interacting proteins that behave as negative regulators [968]. Whereas BORG3 binds only to CDC42, BORG1, BORG2, BORG4, and BORG5 tether to both CDC42 and RhoQ in a GTP-dependent manner. Both full-length BORG1 and BORG2 proteins as well as fragments of BORG1, BORG4, and BORG5 are able to interact with RhoQ.
9.3.21 RhoT GTPase
Small RhoT GTPase is closely related to RhoQ agent. Protein RhoT is predominantly expressed in heart and uterus. It is also induced during neuronal differentiation. Whereas RhoQ generates actin filament-containing peripheral processes longer than CDC42-launched filopodia, RhoT produces much longer and thicker processes that are also composed of actin filaments. Both RhoQ and RhoT assist neurite outgrowth [969]. They as well as CDC42 bind to and activate nWASP to cause ARP2/3-mediated actin polymerization.
9.3.22 Atypical RhoT1/2 GTPases (Miro-1/2)
Monomeric GTPases Ras homolog gene products RhoT1174 and RhoT2175 have a C-terminal transmembrane domain that serves as a label for mitochondria. Protein RhoT2 induces mitochondrium aggregation. In addition, Miro interacts with kinesin-binding proteins, thereby linking mitochondria to microtubules [970].
9.3.23 Atypical RhoU GTPase
Monomeric RhoU GTPase176 actuates filopodium formation and stress fiber dissolution. In addition, it localizes to focal adhesions and Src-induced podosomes to enhance cell migration [971].
Small RhoU GTPase is targeted by transcription factor signal transducer and activator of transcription STAT3 that is stimulated by growth factors and morphogen Wnt1 using the non-canonical planar cell polarity pathway with activation of Jun N-terminal kinase [972].
9.3.24 Atypical RhoV GTPase
Small RhoV GTPase177 has been initially identified as a protein interacting with the regulatory domain of PAK2 kinase. Activated RhoV stimulates the JNK pathway and provokes the formation of small lamellipodia and focal adhesions [973]. Activity of RhoV depends on palmitoylation to associate with cellular membranes. It is insensitive to RhoGDI agents.
9.3.25 RGK (Rad–Gem/Kir–Rem) Family of GTPases
The RGK family of small GTPases includes Rad, Rem1,178 Rem2, and Gem179 that are involved in the nucleocytoplasmic transport. Among the small GTPases of the RGK family, Rem2 is the single member that abounds in neurons.
Rem-interacting proteins include 14-3-3 isoforms (ε, η, θ, and ζ). Protein Rem is phosphorylated and then binds to 14-3-3ζ, as the presence of protein phosphatase-1 abolishes this association [974]. Protein 14-3-3 may recruit mediators of Rem-dependent signaling.
Small Ras GTPase associated with diabetes (Rad; Sect. 9.3.5),180 is highly expressed in human skeletal and cardiac muscles and lung. It inhibits glucose uptake. It is overexpressed in skeletal myocytes in insulin-independent type-2 diabetes [975]. In addition, Rad GTPase is an inhibitor of cardiac hypertrophy via calmodulin-dependent kinase-2δ [976].
Small GTPase Rem alters the excitation–contraction coupling by reducing the number of functional plasmalemmalCaV1 channels [977]. In excitable cells, inhibition of voltage-gated Ca
channels by RGK GTPases participates in the regulation of Ca
influx, in addition to channel expression, among other mechanisms. In fact, RGK GTPases are potent inhibitors of Ca
influx via high-threshold voltage-gated Ca
channels CaV1.2 and CaV2.2, as small RGK GTPases acts on channel auxiliary CaVβ1 and -2 subunits [978, 979]. Effect of RGK corresponds to a decrease in amplitude without modification in voltage dependence and ion flux kinetics [979]. In neurons, Rem2 nearly abolishes calcium currents arising from high-voltage-gated Ca
channels without affecting low-voltage-gated Ca
channels. Small Rem2 GTPase localizes to the plasma membrane and interacts with calcium channel β subunits in the pre-assembled CaV2.2 channel, thereby forming a non-conducting carrier [980].






Insulin secretion by pancreatic β cells also requires Ca
ingress. Monomeric Rem2 GTPase that is synthesized upon exposure to high glucose level associates with CaV1.2 and CaV1.3, thereby preventing Ca
import and glucose-stimulated insulin secretion [981].


Small Gem GTPase interacts with the microtubule network through kinesin-like protein KiF9. Moreover, it regulates actin dynamics downstream from RhoA by inhibiting kinase RoCKβ and interacts with Rho GTPase-activating protein Gmip [982].
9.3.26 RND Family of Atypical GTPases
The family of Rnd GTPases, a subset of the RHO superfamily, includes 3 members (Table 9.35): Rnd1 or Rho6, Rnd2 or Rho7, and Rnd3 or RhoE. They regulate actin cytoskeleton dynamics. The Rnd proteins antagonize small GTPase Rho, as they hinder the formation of and even disrupt contractile actomyosin stress fibers. Constitutively active RndGTP is regulated by its synthesis level, location, phosphorylation, and degradation [983].
Table 9.35
Rnd GTPases and their effectors (Source: [889]). Formin-binding protein FnBP1 (a.k.a. Rnd2 apostle rapostlin) that binds to microtubules favors neurite branching. Pragmin (pragma of Rnd2) causes cell contraction via the RhoA–RoCK pathway. Socius (a.k.a. UBX domain-containing protein UBXD5) is a RndGAP involved in disassembly of actin stress fibers. It connects to Gα subunits.
Type | Effectors |
---|---|
Rnd1 | Socius |
Rnd2 | FnBP1, pragmin |
Rnd3 | RocK1, socius, p190RhoGAP |
The Rnd proteins operate in fibroblast-growth-factor-receptor-1 signaling. Small Rnd GTPases are involved in axon guidance. They interact with plexins. Plexins (plexin-A–plexin-D) are semaphorin receptors that govern cell adhesion, migration, and axon guidance. Among plexins, plexin-A1 and -B1 operate as rRasGAP. Activity of rRasGAP requires Rnd1 binding. Plexin-D1 also acts as rRasGAP and inhibits cell migration with the help of Rnd2 [984]. Like other plexins, Plexin-C1 exhibits rRasGAP activity. Nevertheless, it can prevent cell migration without Rnd GTPase.
9.3.26.1 Rnd1 GTPase
Isotypes Rnd1 and Rnd3 interact with RhoGAP35, and recruit this protein at sites where Rho should be inhibited. Both RhoGAP35 isoforms, RhoGAP35a and RhoGAP35b, stimulate Rnd1 GTPase. Isotype Rnd1 interacts with plexin-A1 and -B1, the semaphorin-3A and -4D receptors, UBX domain-containing protein UBXD5 (or Socius; a RndGAP involved in disassembly of stress fibers), GRB7 adaptor, and rRas [985]. In addition, Rnd1 interacts with membrane-anchored, PTB domain-containing fibroblast growth factor receptor substrates FRS2a and FRS2b that are docking proteins, which recruit signaling proteins, such as PKCλ and NTRK1 kinases, PTPn11 phosphatase, CBL and GRB2 adaptors, and SOS guanine nucleotide-exchange factor to the plasma membrane during fibroblast growth factor stimulation. Interaction of Rnd1 with FRS2b prevents Rnd1 ability to downregulate RhoA GTPase.
9.3.26.2 Rnd2 GTPase
Subtype Rnd2 is predominantly synthesized in testes as well as, to a lesser extent, the brain and liver. In neurons, it regulates the actin cytoskeleton [983]. Small GTPase Rnd2 interacts with RhoGAP5, albeit less strongly than Rnd1 and Rnd3 GTPases. In the testis, Rnd2 also binds to RacGAP1. In neurons and testis cells, it works with UBXD5, or UBX domain-containing protein UBXn11, a RndGAP molecule.
Small GTPase Rnd2 is less efficient at eliminating stress fibers in fibroblasts than other Rnd isoforms [985]. In neurons, it induces neurite branching together with rapostlin (apostle of Rnd2) and nWASP protein.
Unlike Rnd1 and Rnd3, Rnd2 can have a GTPase activity. Like Rnd1, but not Rnd3, Rnd2 interacts with FRS2a and FRS2b substrates. These molecules sequester Rnd2 in an inactive complex. In addition, Rnd2 interacts with vacuolar protein sorting VPS4a in early endosomes. Isoform Rnd2 can link to the plasma membrane as well as other membranes.
9.3.26.3 Rnd3 GTPase
Isotype Rnd3, or RhoE, is ubiquitous, but at very low levels. Its activity is regulated by RoCK1 [985]. Small Rnd3 GTPase contributes to the regulation of P53-mediated stress response that triggers actin depolymerization [986]. It inhibits RoCK1 during genotoxic stress, thereby suppressing apoptosis. Monomeric Rnd3 GTPase also participates in the regulation of cell cycle progression, eventually by decreasing adhesion and inducing dissociation of integrin-based focal adhesions that hamper cell cycle progression, and above all by inhibiting induction of cyclin-D1 [983]. Protein Rnd modulates smooth muscle contractility. In the brain, they are implicated in neurite extension (growth cone). Amphetamine and cocaine upregulate expression of Rnd3 in mice brain by distinct pathways [987].
9.3.27 Family of RIN and RIT GTPases
Small GTPases RIN (Ras-like protein expressed in neurons), a neuron-specific and calmodulin-binding Ras-related protein, and RIT (Ras-like protein expressed in many tissues; Table 9.36) are 25-kDa Ras-like GTPases that are 64% identical [988]. They lack a binding motif (Ras-like proteins without a terminal cysteine–aliphatic amino acid–aliphatic amino acid–any amino acid [CAAX] motif) for C-terminal lipid anchor that enables plasma membrane association that is essential for their activity.181 Nonetheless, transiently produced RIN and RIT localize at the plasma membrane.
Table 9.36
Small RIN and RIT GTPases (Source: [889]). Whereas RIT is ubiquitous, RIN is detected only in neurons.
Type | Aliases |
---|---|
RIN | RIBa, RIT2, Roc2 |
RIT | RIBb, RIT1, Roc1 |
9.3.27.1 RIN1 (RIT2)
Small RIN GTPase (or RIN1), a Ras-related protein as well as a Ras effector, is a calmodulin-binding protein expressed predominantly (if not solely) in neurons. It is also called Ras-like without CAAX motif type-2 protein (RIT2).182 The Rin1 gene encodes a Ras effector that regulates epithelial cell behavior.183 The transcriptional repressor Snai1 (Snail homolog) that silences the CDH1 (E-cadherin) gene as well as other proepithelial genes hampers RIN1 synthesis [989]. Moreover, DNA methylation within the Rin1 promoter and first exon can also contribute to gene silencing.
Monomeric RIN GTPase forms a family with RIT agent. Protein RIN localizes to both the plasma membrane and nucleus. The GTP dissociation rate is 5-to 10-fold faster than most Ras-like GTPases. Small GTPase RIN is activated by Src kinase following stimulation of both Gs and Gi subunits.
Protein RIN1 activates Abl kinases and Rab5 GTPase to regulate cytoskeletal remodeling and endocytic pathways. It thus reduces motility of epithelial cells, as activated Abl kinases precludes cytoskeletal rearrangements needed for cell dissociation and migration [989]. Molecule RIN operates via HSP27 and the cAMP–PKA pathway. Neuropeptide pituitary adenylate cyclase-activating polypeptide PACAP38 acts via G-protein-coupled receptors and RIN for its neurotrophic and neurodevelopmental effects [990]. Signaling based on RIN appears to contribute to nerve growth factor-dependent neuronal differentiation in cooperation with both Rho and P38MAPK mediators.
Protein RIN can associate with Ca
–calmodulin [991]. Active RINGTP is able to interact with Par6 scaffold and bRaf kinase, whereas RINGDP can associate with transcription factor POU (pituitary-specific Pit1/octamers Oct/Unc) class-4 homeobox gene product POU4F1, or brain-specific homeodomain-containing protein Brn3a.

9.3.27.2 RIT1
Protein RIT1 (Ras-like protein expressed in many tissues)184 is ubiquitous, but transiently expressed at the plasma membrane. The GTP dissociation rate is 5- to 10-fold faster than most Ras-like GTPases. Small GTPase RIT intervenes in neuronal differentiation. It selectively activates neuronal bRaf isoform. Protein RIT1 can also complex with Par6 and CDC42 [992].
9.3.28 Phosphoinositide 3-Kinase Enhancer: GTPase and GAP
Phosphoinositide 3-kinase enhancer (PIKE) is a GTPase that binds to and stimulates PI3K kinase. Protein PIKE exists in 3 types: 2 brain-specific isoforms, a long form PIKEL (or PIKE1) and a short form PIKES (or PIKE2) and a ubiquitous PIKEa subtype. Whereas PIKES is exclusively nuclear, PIKEL resides in both the nucleus and cytoplasm. The latter contains a C-terminal extension with an adpribosylation GTPase-activating protein domain and 2 ankyrin repeats, in addition to the N-terminal GTPase domain. Therefore, it is also termed ArfGAP with GTP-binding protein-like, ankyrin repeat and pleckstrin homology domain-containing AGAP2 protein.
Isoform PIKEa contains the domains of PIKEL, except the N-terminal proline-rich domain that binds PI3K and PLCγ1 enzymes. Yet, PIKEa specifically binds to active protein kinase-B.
Small PIKE GTPase is also known as centaurin-γ1 (encoded by the CentG1 gene) and GTP-binding and GTPase-activating protein GGAP2. It has a strong GTPase activity. Moreover, it actually acts as a GTPase-activating protein for ARF1 and ARF5 GTPases.
Cytoplasmic PI3K and its products phosphatidylinositol (4,5)-bisphosphate and (3,4,5)-trisphosphate regulate membrane translocation of many signaling molecules, as they bind to and activate these mediators. Small PIKE GTPases are regulated by PI(4,5)P2 and, to a lesser extent, by PI(3,4,5)P3. Phosphatidic acid potentiates PIP2 stimulation.
Nerve growth factor activates PIKES, as it triggers nuclear translocation of phospholipase PLCγ1 that acts as a guanine nucleotide-exchange factor for PIKES [993].
Isoform PIKEL associates with Homer-1, a metabotropic glutamate receptor mGluR1-binding adaptor. The Homer1–PIKEL complex couples PI3K to mGluR1 to prevent neuronal apoptosis. In addition, PIKEL may regulate postsynaptic signaling by metabotropic glutamate receptors. Isoform PIKES also binds to and enhances PKB activity to prevent apoptosis. Besides, it also regulates adaptor protein-1-dependent transport of proteins in the endosomal system.
9.4 Regulators of Monomeric Guanosine Triphosphatases
Monomeric GTPases exist in 3 states: (1) a transient, active, GTP-bound state, in which they connect to signaling effectors; (2) a transient, free, inactive, GDP-loaded state that results from the action of GTPase-activating proteins (GAP); and (3) a sequestered, GDP-bound, guanine nucleotide-dissociation inhibitor (GDI)-complexed state.
The bimodal functioning of small GTPases during transmission of intracellular signals involves a cascade of between-protein interactions modulated by chemical modifications, structural rearrangements, and intracellular relocalizations. The GDP–GTP binding cycle enables monomeric GTPases to filter, amplify, or temporize receiving signals.
Switching between an inactive and active, effector-binding conformation is regulated by: (1) guanine nucleotide-exchange factors (GEF) that act as activators, as they promote the release of GDP and binding of GTP; and (2) GTPase-activating proteins (GAP) that accelerate the intrinsically slow GTPase activity of monomeric GTPases. Therefore, GTP binding is primed by GEFs (that then do not improve a prerequisite condition, but trigger signal transmission), whereas GAPs enhance intrinsic catalysis rate of small GTPases (hence they do not create a new state) or cause the release of GTP of small GTPases devoid of strong enzymatic capacity.
Membrane anchoring of monomeric GTPases is often a prerequisite for their activity. Therefore, small GTPases require not only the presence of GEFs and GAPs to modulate upstream signaling, but also mediators that relieve their cytosolic sequestration by guanine nucleotide-dissociation inhibitors. The control of the cellular membrane association–dissociation cycle corresponds to an additional regulation level.
9.4.1 Guanine Nucleotide-Exchange Factors
Guanine nucleotide-exchange factors increase the activity of small GTPases, as they catalyze GDP release and cause GTP association (i.e., exchange of GDP for GTP), thereby activating small GTPases (Fig. 9.1).185
Multiple GEFs have been described in humans. Many GEFs can activate various Rho GTPases.186 More than 80 RhoGEFs (Table 9.37) activate Rho GTPases that are encoded by 22 genes and regulate the actin cytoskeleton according to the type of control input. Monomeric GTPases CDC42, Rac1, and RhoA intervene in the formation of filopodia, lamellipodia, and contractile actin–myosin filaments (stress fibers), respectively. Guanine nucleotide-exchange factors link specific cytoskeletal responses to corresponding signaling. Reprogrammed GEFs can act on unrelated processes [994].
Table 9.37
Examples of guanine nucleotide-exchange factors( ARNO: ARF nucleotide site opener; BRAG: brefeldin-resistant ArfGEF; DOCK: dedicator of cytokinesis; EFA6: exchange factor for ARF6; FARP: FERM, RhoGEF and pleckstrin domain protein, a.k.a. FERM domain including RhoGEF [FRG] and pleckstrin homology domain-containing family-C member PlekHc3; FBx: F-box only protein; DEF6: differentially expressed in FDCP6 homolog, a.k.a. SWAP70L and IRF4-binding protein (IBP); PREx: PIP3-dependent Rac exchanger; RabIn3: Rab3a-interacting protein; RIn: Ras and Rab interactor; RGL: Ral guanine nucleotide-dissociation stimulator-like protein; SOS: Son of sevenless; SWAP: Switch-associated protein; TIAM: T-cell lymphoma invasion and metastasis; ).
Category | Examples of members |
---|---|
ArfGEFs | ARF1GEF (GBF1), ArfGEF1–ArfGEF3 (BIG1–BIG3), |
cytohesin-1–cytohesin-4 (ARNO1–ARNO4), | |
BRAG1–BRAG3 (ARF6GEF1–ARF6GEF3), | |
EFA6a–EFA6d (ARF6GEFa–ARF6GEFd), FBx8 | |
CDC42GEF | FARP2, intersectin-1L, Tuba |
RabGEFs | RIn1–RIn3, RabGEF1, |
Rab3IP, Rab5GEF (alsin), RabIn8, RabIn3L | |
RacGEF | DEF6, PREx, SwAP70, TIAM1–TIAM2 |
RalGEFs | RalGEF (RalGDS), RGL1–RGL4 |
RanGEFs | |
RapGEFs | RapGEF1–RapGEF6 |
RasGEFs | RasGRF1–RasGRF3, RasGRP1–RasGRP4 |
DOCK1–DOCK4 | |
RhoGEFs | Duo, FGD1, RhoGEF1–RhoGEF19, |
RhoGEF21/23–25/27, RhoGEF30–RhoGEF31, | |
SOS1/2, Scambio, Vav1–Vav3 |
An additional level of regulation relies on restricted subcellular distributions of GTPases and their GEF and GAP regulators. In addition, GEFs promote crosstalks between different sets of small GTPases. Activated Ras activates GEFs for small GTPases Ral [995], Rab5 [996], and Rac [997].
9.4.1.1 ArfGEFs
Guanine nucleotide-exchange factors that target ARF GTPases possess a Sec7 domain responsible for GEF activity.187 The human genome encodes 15 known Sec7 family members (Table 9.38). They contribute to the regulation of membrane remodeling and trafficking associated with vesicular transport [998]
Table 9.38
Effects of ArfGEFs (Source: [840]; ER: endoplasmic reticulum; AP: adaptor proteic heterotetramer; ARNO: ARF nucleotide site opener; BIG: brefeldin-A-inhibited GEFs for ARFs; BRAG: brefeldin-resistant ArfGEF; CoP: coating protein complex (coatomer); EFA6: exchange factor for ARF6; GBF: Golgi-associated brefeldin-A-resistant GEF; GGA: Golgi-localized γ-ear-containing ARF-binding protein).
ArfGEF | Functions |
---|---|
GBF1 | Endoplasmic reticulum–Golgi body transport, CoP1 recruitment |
BIG1 | Exocytosis from Golgi body |
BIG2 | Exocytosis from Golgi body, AP1 and GGA recruitment |
ARNO | Actin remodeling, GPCR desensitization |
Ca ![]() | |
Cell migration | |
ARNO2 | β2-Integrin adhesion |
Synaptic transmission | |
ARNO3 | Growth-factor signaling |
(e.g., insulin, EGF, and NGF) | |
ARNO4 | |
EFA6a | Endocytosis, recycling, actin remodeling |
EFA6b | Endocytosis, recycling, actin remodeling |
EFA6c | |
EFA6d | |
BRAG1 | Vesicle trafficking at synapses |
Association with NMDA receptors at excitatory synapses | |
BRAG2 | Endocytosis and recycling of β1-integrin |
BRAG3 |
Only 2 among 7 ArfGEF sets exist in all eukaryotes: (1) BIG set of brefeldin-A-inhibited guanine nucleotide-exchange factors for adpribosylation factors (BIG1–BIG7) and (2) GBF set of Golgi-associated brefeldin-A-resistant guanine nucleotide-exchange factors (GBF1–GBF3) that are also termed guanine nucleotide-exchange on ARF (GEA).188 The set of eukaryotic ArfGEFs is subdivided into 5 families based on overall structure and domain organization [999] (Tables 9.39 and 9.40): (1) BIG–GBF family of BFA-inhibited GEF (BIG) and Golgi body brefeldin-A-resistance factor (GBF) that has representatives in all eukaryotes; (2) ARNO/cytohesin family of ARF nucleotide-binding site openers (ARNOs) or cytohesins;189 (3) EFA6 family of exchange factors for ARF6; (4) BRAG family of brefeldin-resistant ArfGEF; and (5) FBx family of F-box only protein-8 (FBx8).
Table 9.39
Guanine nucleotide-exchange factors of ARF GTPases (ArfGEFs) in humans (Part 1; Sources: [838, 998, 999] and Wiki Professional; ArfD: ARF domain protein; ARNO: ARF nucleotide-binding site opener; CNKSR: connector enhancer of kinase suppressor of Ras; PKAR: protein kinase-A regulatory subunit; PSCD: PH, Sec7, and coiled-coil domain-containing protein). Vesicles from the endoplasmic reticulum travel via the vesicular-tubular cluster (VTC) to the cis-Golgi network (CGN), where they fuse. Materials subsequently progress through the Golgi body. Vesicles also carry substances from the trans-Golgi network (TGN) to their final destination (Endos: endosome; rEndos: recycling endosomes; N: nucleus; PM: plasma membrane). In addition to their substrates, ArfGEF bind to many partners, such as other ArfGEFs, receptors (A2A adenosine receptor; THR: thyroid hormone receptor), ion channels, adaptors (CASP: cytohesin-associated scaffold protein; GRASP: GRP1 [ARNO3]-associated scaffold protein; GRSP: GRP1 [ARNO3] signaling partner; IPCEF: interaction protein for cytohesin exchange factor; VDP: vesicle docking protein [P115]), enzymes (vATPase: vacuolar adenosine triphosphatase), exocyst subunits (Exo), and signaling regulators (Arrβ: β-arrestin).
Type | Aliases | Site | Substrates | Partners |
---|---|---|---|---|
BIG1 | ArfGEF1, | TGN, N, | ARF1/3 | BIG2, FKBP13, |
P200ArfGEF1 | endosome | Exo70, myosin-9b | ||
BIG2 | ArfGEF2 | TGN, | ARF1/3 | BIG1, PKAR1/2, |
BIG5 | rEndos | Exo70, GABAR | ||
BIG3 | ArfGEF3 | |||
GBF1 | CGN, VTC, | ARF1/3/5 | Rab1b, VDP | |
ERGIC | ||||
Cytohesin-1 | PSCD1, ARNO2 | PM, | ARF1/6, | ARP, ARL4, ArfRP, |
endosome | ArfD1 | CASP, CNKSR1 | ||
Cytohesin-2 | PSCD2, ARNO | PM, | ARF1/3/6 | ARF6, ARL4, A2A , |
endosome | ARL4 | Arrβ, CASP, GRASP, | ||
CNKSR1, HER, | ||||
IPCEF1, vATPase | ||||
Cytohesin-3 | PSCD3, ARNO3, | PM | ARF1/6 | ARF6, ARL4, CASP, |
GRP1 | endosome | CNKSR1, GrASP, | ||
GRSP1, THR | ||||
Cytohesin-4 | PSCD4, ARNO4 | ARF1/5 |
Table 9.40
Guanine nucleotide-exchange factors targeting ARF GTPases (ArfGEFs) in humans (Part 2; Sources: [998, 999] and Wiki Professional; ArfGEP: ARF guanine nucleotide-exchange protein; BAIAP: brain-specific angiogenesis inhibitor 1-associated protein [or insulin receptor substrate P53 (IRSP53)]; BIG: brefeldin-A-inhibited GEFs for ARFs; BRAG: brefeldin-resistant ArfGEF; EFA6: exchange factor for ARF6; FBx: F-box only protein; FBS: F-box, Sec7 protein; IQSec: IQ motif and Sec7 domain-containing protein; PSD: pleckstrin- and Sec7 domain-containing protein). Vesicles from the endoplasmic reticulum travel to the cis-Golgi network (CGN), where they fuse. Cargos subsequently progress through the Golgi body. Vesicles also carry substances from the trans-Golgi network to their final destination (Endos: endosome; N: nucleus; PM: plasma membrane; PsD: postsynaptic density). In addition to their substrates, ArfGEF bind to many partners, such as other ArfGEFs, receptors, ion channels (K2P 1.1: two pore-forming K + channel [KCNK1]), adaptors (Homer-1; DLg: Disc large homolog), enzymes, exocyst subunits, and signaling regulators.
Type | Aliases | Site | Substrates | Partners |
---|---|---|---|---|
EFA6a | EFA6, PSD, | PM | ARF6 | K2P 1.1 |
PSD1, TYL | ||||
EFA6b | PSD4, TIC | PM | ARF6 | |
EFA6c | PSD2 | PsD | ARF6 | |
EFA6d | PSD3 | PsD | ARF6 | |
BRAG1 | IQSec2, | PsD | ARF6 | BAIAP2 |
ArfGEF4 | ||||
BRAG2 | IQSec1, | PM, Endos, N | ARF6 | AMPAR |
ArfGEP100 | ||||
BRAG3 | IQSec3, Sag, | PsD | ARF6 | DLg1/4, Homer1 |
synArfGEF | dystrophin | |||
FBx8 | FBS |
Cytohesins
In humans, the cytohesin family contains 4 known members (cytohesin-1–cytohesin-4).190 Cytohesin-2 and -3 are ubiquitous. Cytohesin-1 lodges principally in leukocytes; cytohesin-4 is more leukocyte specific. Cytohesins reside primarily in the cell periphery. Two splice isoforms of cytohesin-1, -2, and -3 are synthesized [999].
Cytohesin-1 is a guanine nucleotide-exchange factor for membrane-associated ARF GTPases. It is highly synthesized in natural killer and peripheral T cells. It elicits ARF-dependent activation of phospholipase-D. This integrin-binding GEF favors adhesion of lymphocytes mediated by αLβ2-integrin, hence their attachment and migration [1000].
Cytohesin-2 targets ARF1, ARF3, and ARF6 that promotes both migration of epithelial cells and outgrowth and branching of neurites [999]. Integrin-mediated activation of rRas leads to cytohesin-2 recruitment via rRas effector RalA-binding protein RalBP1. In migrating epithelia, cytohesin-2 promotes Rac1 activation by recruitment of the DOCK1–ElMo complex, a Rac1GEF (Sect. 9.4.1.7). It is also involved in docking and fusion of secretory granules, postendocytic trafficking via vacuolar adenosine triphosphatase, as well as endocytosis of some G-protein-coupled receptors via β-arrestins.
Recruitment of cytohesins to membranes is done via phosphoinositides and proteic adaptors, such as cytohesin-interacting protein (CytIP) or cytohesin-associated scaffold protein (CASP), GRP1-associated scaffold protein (GRASP or tamalin), and interacting protein for cytohesin exchange factors (IPCEF) [999]. Among these tethering proteins, GRASP couples metabotrobic glutamate receptors and the neurotrophin receptor NTRK3 to ARF activation.
ARF1GEF
Protein ARF1GEF, or Golgi-specific resistance to brefeldin-A factor GBF1, targets both subfamily-1 and -2 ARFs. It localizes to cis-compartments of the Golgi body, where it regulates assembly of CoP1 coat. On the other hand, ArfGEF1 and ArfGEF2, or BIG1 and BIG2, that activate subfamily-1 ARFs concentrate on trans-compartments of the Golgi body, where they control the recruitment of clathrin adaptors [1001].
Protein ARF1GEF presents GEF activity specifically for ARF5 at physiological Mg
concentration [1002]. It associates primarily with vesicular–tubular clusters. It is involved in transport between the endoplasmic reticulum and Golgi body as well as inside the Golgi body. In vesicular–tubular clusters, it interacts with vesicle docking protein (VDP) and actuates ARF-dependent recruitment of vesicle coat protein CoP1 [999].

Agent ARF1GEF is recruited to exit sites of the endoplasmic reticulum and Golgi membranes by interacting with Rab1b GTPase [999]. It rapidly attaches to and detaches from membranes of the inner Golgi compartment (cis-Golgi network). In fact, ARF1GEF substrates include ARF1 among others [1003]. Protein ARF1GEF not only mediates the recruitment of CoP1 coat to cis-Golgi membranes, but also stimulates ARF1 in the cis-Golgi network.
ArfGEF1–ArfGEF3 (BIGs)
Proteins ArfGEF1 to ArfGEF3 correspond to brefeldin-A-inhibited GEFs for ARFs BIG1 to BIG3, respectively. Proteins ArfGEF1 and ArfGEF2 (BIG1 and BIG2) as well as ARF1GEF (or GBF1) can homodimerize. Both ArfGEF1 and ArfGEF2 associate with outer compartments of the Golgi body.
Like ARF1GEF, ArfGEF1 rapidly connects to and is released from membranes. Whereas ARF1GEF is associated with the cis-Golgi network, ArfGEF1 is linked to the trans-Golgi network.
Protein ArfGEF2 regulates the association of heterotetrameric adaptor protein complex AP1 and Golgi-localized γ-ear-containing ARF-binding proteins (GGA) to the trans-Golgi network. Both adaptor-related protein complex AP1191 as well as Golgi-associated, γ-adaptin ear-containing, ARF-binding adaptor complexes are components of clathrin coat complexes. Protein ArfGEF2 is also linked to perinuclear recycling endosomes.
ARF6GEF1 to ARF6GEF3 (BRAGs)
Three brefeldin-resistant ArfGEFs exist (BRAG1–BRAG3), each with splice variants [999]. All activate ARF6;192 in the present text, they are then called ARF6GEF1 to ARF6GEF3 proteins. Agent ArfGEF4, or ARF6GEF1 (BRAG1), is a member of group 3 of large ArfGEFs; ArfGEF1 to ArfGEF3 (BIGs) and ARF1GEF (GBF1) pertain to group-1 and -2.
Like most ArfGEFs, ARF6GEFs are expressed at low levels. Proteins ArfGEF4, or ARF6GEF1 (BRAG1), and ARF6GEF3 (or BRAG3) are mainly produced in the brain; ARF6GEF2 (or BRAG2) abounds in the brain, but is also synthesized in various tissues. Protein ArfGEF4 targets ARFl and ARF6 in neurons.
Postsynaptic densities of excitatory and inhibitory synapses contain diverse types of GEFs (ARF6GEF1, ARF6GEF2b, RapGEF4, RhoGEF9, and RhoGEF24) and GAPs (RasA1, APAP1, AGAP2L, and SpAR RapGAP) for small GTPases [1005]. These regulators modulate the synaptic transmission, as they control the formation and maintenance of dendritic spines and synapses via the actin cytoskeleton structuring. At postsynaptic densities of excitatory synapses, ARF6GEF1 complexes with NMDA-type glutamate receptors. On the other hand, ARF6GEF3, like RhoGEF9, localizes preferentially to postsynaptic densities of inhibitory synapses, with gephyrin, neuroligin-2, dystrophin, syntrophin, α- and β-dystroglycan, α- and β-dystrobrevin, and membrane associated guanylate kinase with inverted domain organization MAGI2, in addition to glycine and/or GABAA receptors. Long ARF6GEF3 splice variant (ARF6GEF3L) interacts with postsynaptic proteins Disc large homologs DLg1 and DLg4 as well as Homer-1.193
Guanine nucleotide-exchange factor ARF6GEF1 (ArfGEF4) is confined to early Golgi compartments. Unlike ARF1GEF, it displays broader ARF substrate specificity [1001].
In non-neuronal cells, ARF6GEF2 regulates endocytosis of some cargos. Agent ARF6GEF2 cycles between the cytoplasm, where it contributes to endocytosis regulation at the plasma membrane, and the nucleus, where it can regulate nucleolar architecture [1006]. Its 2 splice variants — ARF6GEF2a and ARF6GEF2b — can both enter in and exit the nucleus.
ARF6GEFa to ARF6GEFd (EFA6s)
Exchange factor for adpribosylation factor ARF6 (EFA6) localizes mainly at the apical pole of polarized epithelial cells. It contributes to the stability of the apical actin ring onto which the tight junction is anchored [1004]. In response to E-cadherin engagement, it is also involved in tight junction formation.
Protein EFA6 was the first identified set member that activates ARF6, especially in developing and mature neurons, where it regulates actin cytoskeleton reorganization associated with intracellular transfer of cargos. Additional ARF6-specific GEFs constitute a family of 4 structurally related proteins (EFA6a–EFA6d); in the present text, they are called ARF6GEFa to ARF6GEFd, as it is done for BRAG family members, called ARF6GEF1 to ARF6GEF3 for a straightforward meaning. Members of the EFA6 family are highly selective for ARF6 [999]. They also interact selectively with PI(4,5)P2 and localize mostly to the plasma membrane. These EFA6 isoforms promote the reorganization of cortical actin into microvillus-like structures.
Protein EFA6a is synthesized predominantly in the central nervous system, but also in intestine; EFA6b is more widespread with its highest levels in the pancreas, spleen, thymus, and placenta; EFA6c is also produced primarily in the brain; EFA6d is ubiquitous [999]. In the brain, EFA6a, EFA6c, and EFA6d have distinct regional distributions.
FBx8
Molecule F-box only protein FBx8 mediates incorporation of proteins via ubiquitination, as it can interact with both ARFs and ubiquitin ligases [999].
9.4.1.2 RabGEFs
Protein Ras and Rab interactor RIn1 that impedes activated Ras is stimulated by Ras and acts as a guanine nucleotide-exchange factor for Rab5 [996] (Table 9.41).
Table 9.41
Guanine nucleotide-exchange factors for Rab (RabGEFs) and their effectors (Source: [889]; RabEx5: Rab5 GDP–GTP exchange factor; RASSF: Ras interaction/interference RIN1, afadin, and Ras association domain-containing protein family member; RIn: Ras and Rab interactor).
Type | Aliases | Effectors |
---|---|---|
RIn1 | Rab5 | |
RIn2 | RASSF4 | |
RIn3 | ||
RabGEF1 | RabEx5 | Rab5 |
9.4.1.3 Rab5GEF (Alsin)
Amyotrophic lateral sclerosis protein-2 (ALS2), also called alsin, acts as a Rab5GEF involved in endosome dynamics [1007]. Endocytosis mediated by Rab5 and appropriate rate of endosomal conversion to lysosomes, i.e., proper lysosomal degradation of internalized glutamate receptors in neurons, requires Rab5GEF [1008]. Agent Rab5GEF preferentially interacts with activated Rac1 that recruits cytosolic Rab5GEF to membranes. Activator Rab5GEF participates in Rac1-activated macropinocytosis [1009].
9.4.1.4 RacGEFs
Dendritic spines, small protrusions from a dendrite that increase the number of possible contacts between neurons, serve for memory storage and synaptic transmission. Dendritic spine morphogenesis and remodeling of excitatory synapses is needed for adequate neuronal development. Calcium–calmodulin-dependent kinases and guanine nucleotide-exchange factors RacGEFs interact to control dendritic spine morphogenesis [1010]. This connection between these 2 agents transduces Ca
influx into small GTPase activity that leads to actin reorganization, hence into spatially and temporally regulated remodeling of dendritic spines.

Small Rap1 GTPase modulates Rac1 activity. The latter is activated and translocates to intercellular adhesion plaques. Proteins Vav2 and TIAM1 are recruited by atrial natriuretic peptide and prostaglandin-E2 to enhance the endothelial barrier (Sect. 9.4.1.6).
TIAM1–TIAM2
Agent T-cell lymphoma invasion and metastasis-inducing protein TIAM1, a Rac-specific guanine nucleotide-exchange factor, can augment endothelial permeability, hence leukocyte transendothelial migration. Platelet-activating factor disrupts interendothelial junctions, as it provokes translocation of Rac1 and TIAM1 [1011]. It binds PI(3,4,5)P3 with high affinity. In vascular endothelial cells, RapGEF3-dependent Rap1 activation causes relocalization and stimulates Rac-specific GEFs TIAM1 and Vav2 and promotes accumulation of suppressor of cytokine signaling SOCS3 (cAMP–RapGEF3–Rap1–SOCS3 pathway), thereby inhibiting signal transducer and activator of transcription [1012]. Protein TIAM1 mediates Ras activation of Rac to stimulate membrane ruffling as well as activate the JNK and NFκB pathways, hence promoting cell survival, independently of phosphatidylinositol 3-kinase [997]. Protein TIAM1 regulates cell migration, as it modulates both intercellular and cell–matrix adhesions.
Other TIAM1 partners include protein kinase-Cα, -Cβ1, -Cγ, -Cδ, -Cε, and -Cζ, protein phosphatase-1, MAPK8-interacting protein-1 and -2, and ankyrins-1 and -3 [251].
Activator TIAM2 that is also called SIF and TIAM1-like exchange factor (STEF) targets Rac1 GTPase (Table 9.42).
Table 9.42
Guanine nucleotide-exchange factors for Rac GTPases (RacGEFs) — TIAM1 and TIAM2 — and their effectors (Source: [889]).
Type | Effectors |
---|---|
TIAM1 | Rac1, Rac2, Rac3 |
TIAM2 | Rac1 |
During cell migration, focal adhesions that attach the cytoskeleton to the extracellular matrix, assemble and disassemble. Numerous agents intervenes in this process, such as ERK, FAK, MLCK, PAK, RoCK, and Src kinases as well as Rho GTPases. Member of the spectraplakin family of cytoskeletal crosslinking proteins Microfilament and actin filament crosslinker protein MAcF1194 binds to both actin and microtubule cytoskeletons, thereby guiding microtubules toward actin filaments connected to focal adhesions. In addition, microtubule growth activates Rac GTPase that, in turn, promotes microtubule growth into the lamellipodium (positive feedback loop), as it targets the microtubule plus-tip protein Cytoplasmic linker-associated protein CLAsP2. The latter is involved in the local regulation of microtubule dynamics in response to positional signals. Agent TIAM1 regulates microtubule stability. Subtype TIAM2 is required during microtubule growth that is involved in focal adhesion disassembly for optimal cell migration [1013]. Protein TIAM2 stimulated by microtubules to activate Rac GTPase promotes the growth of microtubules toward focal adhesions.
PREx1 and PREx2
Guanine nucleotide-exchange factors of the PREX (PI(3,4,5)P3-dependent Rac exchanger) family activate Rac GTPases. The PREX family comprises PREx1 and PREx2 (or PREx2a) and its splice variant PREx2b.
Isoform PREx1 is synthesized in leukocytes. Subtype PREx2 is more widely produced, but not in leukocytes. Variant PREx2b is only generated in the heart. Protein PREx1 is also widespread in the central nervous system, whereas PREx2 is specifically expressed in Purkinje neurons of the cerebellum [100].
Both PREx1 and PREx2 connect signaling from Gβγ subunits of heterotrimeric G proteins and phosphatidylinositol (3,4,5)-trisphosphate synthesized by phosphatidylinositol 3-kinase to Rac activation, hence linking GPCR stimulation to that of PI3K kinase. Members of the PREX family of RacGEFs differ from others in their mode of regulation, because they are synergistically activated by Gβγ subunit and PIP3 agent.
Target of rapamycin complex TORC2 regulates the actin cytoskeleton via Rho GTPases. Both PREx1 and PREx2 interact with TOR kinase, thereby acting as effectors in the TOR signaling for Rac activation for cell migration [1014].
Regulator PREx1 regulates GPCR-dependent Rac2 activation, ROS production, neurotrophin-stimulated neuron migration, and neutrophil recruitment to inflammatory sites [100]. Isoform PREx2a is a direct regulator of phosphatase and tensin homolog on chromosome 10 (PTen) that can stimulate cell proliferation, as it inhibits PTen phosphatase, hence stimulating PI3K signaling. In endothelial cells, PREx2b governs Rac1 activation and cell migration in response to sphingosine 1-phosphate.
Rac and RacGEFs in the Nervous System
Small Rac isoforms — Rac1, Rac2, and Rac3 — control the organization of the actomyosin cytoskeleton, gene expression, and production of reactive oxygen species. In the nervous system, Rac GTPases intervene in all stages of neuronal development (neurite, axon, and dendrite formation; axon pathfinding; dendrite branching; dendritic spine formation; and neuron survival). Small Rac GTPases are activated by guanine nucleotide-exchange factors that control neuronal development [100].
Activator of Rac GTPases TIAM1 regulates neurite and axon outgrowth and dendritic spine formation; Vav2 and Vav3 axon outgrowth from retina to thalamus; RhoGEF23 neurite outgrowth, axon extension, and pathfinding; RhoGEF24 regulates neurite and axon outgrowth as well as dendritic spine formation; Dedicator of cytokinesis DOCK1 neurite outgrowth and DOCK7 axon formation; Rac–CDC42 exchange factors RhoGEF7 and RhoGEF25 dendritic spine formation; and Rab5GEF regulates neuronal survival [100].
9.4.1.5 RalGEFs
Protein Ral GDP-dissociation stimulator (RalGDS or RalGEF) is a Ras effector for RalA and RalB GTPases. In addition, RalGDS-like proteins (RGL1–RGL3) also activate both RalA and RalB (Table 9.43). Molecule RalGDS binds preferentially to active forms of Ras, Rap1a, and rRas GTPases. It competes with Ras1 for connection to hRas.
Table 9.43
Guanine nucleotide-exchange factors for Ral (RalGEFs) and their effectors (Source: [889]).
Type | Effectors |
---|---|
RalGEF (RalGDS) | RalA, RalB |
RGL1 | RalA, RalB |
RGL2 | RalA, RalB |
RGL3 | RalA, RalB |
9.4.1.6 RanGEFs
Small GTPase Ras-related nuclear protein (Ran) is involved in nuclear transfer as well as chromatin condensation via its control of microtubule assembly. Regulator of chromosome condensation RCC1 (or ChC1) is a GEF for Ran GTPase [1007]. Ran-binding protein RanBP3 associates with RCC1 to facilitate its activation and to regulate the nucleocytoplasmic transport.
9.4.1.7 RapGEFs
Rap-specific guanine nucleotide-exchange factors that serve as Ras activators constitute several groups that operate in junctional control. According to activators and binding partners, RapGEFs include the subfamilies of [903]:
1.
RapGEF1;
2.
PDZGEFs, or RAGEFs (RapGEF2–RapGEF6);
3.
cAMP-activated GEF (EPAC or cAMPGEF) with 2 members RapGEF3 and RapGEF4;
4.
calcium- and diacylglycerol-regulated GEF (CalDAGGEF; RasGRP1–RasGRP2); and
5.
atypical RapGEF DOCK4; in addition to
6.
phospholipase-Cε.
Small Rap GTPases are involved, in particular, in the control of between-cell and cell–matrix adhesions.195 Four RapGEF families are implicated in junction regulation.
RapGEF1
Ubiquitous guanine nucleotide-exchange factor RapGEF1 targets Ras GTPase.196 It localizes to endosomes. It activates Rap1 upon E-cadherin and EGFR internalization and nectin engagement [903]. Protein RapGEF1 interacts with many adaptors, such as BCAR1, CRK, CRKL, and GRB2 (Table 9.44). It also tethers to E-cadherin during the initial step of junction formation (Table 9.45).
Table 9.44
Guanine nucleotide-exchange factors for Rap GTPase (RapGEFs) and their effectors (Source: [889]).
Type | Main alias | Effectors |
---|---|---|
RapGEF1 | C3G | BCAR1, CBL, CRK, CRKL, GRB2, NEDD9 |
RapGEF2 | PDZGEF1, | Ral, Rap1, Rap2 |
RAGEF1 | ||
RapGEF3 | EPAC1 | Rap1, Rap2 |
RapGEF4 | EPAC2 | Rap1, Rap2 |
RapGEF5 | GFR | Rap1, Rap2, rRas3 |
RapGEF6 | PDZGEF2, | Rap1, Rap2 |
RAGEF2 |
Pathway | Effect |
---|---|
Nectin–Src–RapGEF1–Rap1 | Adherens junction formation |
Stretch–BCAR1–RapGEF1–Rap1 | Adherens junction formation |
RTK—-Src–RapGEF1–Rap1 | Adherens junction formation |
GPCR–cAMP–RapGEF3–Rap1a | Adherens junction tightening |
RhoG–DOCK4–Rap1 | E-Cadhrin recruitment |
RapGEF2/6–Rap1a | Adherens junction formation and maturation |
PDZ Domain-Containing RapGEF2 and RapGEF6
Guanine nucleotide-exchange factors RapGEF2 and RapGEF6197 localize to cell junctions.
Protein RapGEF2198 connects to Rap1a and Ral guanine nucleotide-dissociation stimulator (RalGDS). Agent RapGEF2 associates with β-catenin both directly and via Membrane-associated guanylate kinases (MAGuK) with inverted domain organization scaffolds MAGI1 and MAGI2 [903]. Protein MAGI1199 localizes to tight and adherens junctions in epithelial cells as well as synapses in neurons.200 It is required for cell adhesion-induced activation of Rap1 GTPase.
Molecule RapGEF6 is closely related to RapGEF2 protein. It has 2 splice variants: RapGEF6a and RapGEF6b [1016]. Like RapGEF2, RapGEF6 is specific for Rap1 and Rap2 downstream from rRas3; it is unresponsive to cAMP and other nucleotides. Protein RapGEF6 has a different tissue distribution than that of RapGEF2 agent. It operates in the cell cortex, whereas RapGEF2 exerts its function in the perinuclear compartment. Protein RapGEF6 does not connect to hRas, nRas, RalA, RIN, RIT, and RHEB [1017]. It is an activator of Rap1 during junction formation and maturation both in epithelial and endothelial cells [903].
cAMP-Regulated RapGEF3 and RapGEF4
Guanine nucleotide-exchange factors for Rap GTPases RapGEF3 and RapGEF4201 can act as RasGEFs. These cAMP-responsive guanine nucleotide exchange factors for Rap GTPase are involved in the regulation of endothelial cell adhesion modulation, myocardial contraction, and insulin secretion, among other tasks.
These proteins possess an N-terminal regulatory region with 1 (RapGEF3) or 2 (RapGEF4) cAMP-binding domains and a C-terminal catalytic sequence with a CDC25-homology domain for GEF activity and a Ras exchange motif (REM).
These 2 isoforms have different tissue-specific distribution. Isoform RapGEF4 is predominantly produced in the brain and adrenal glands, whereas RapGEF3 is ubiquitous. Agent RapGEF3 can activate ERK signaling via Rap2b, PLCε, and subsequent hRas activation.
Agent RapGEF3 binds directly to negatively charged phosphatidic acid using its Disheveled, EGL10, and pleckstrin (DEP) domain under the control of cAMP for subsequent activation of Rap GTPase at the plasma membrane [1018].202 In addition, RapGEF3 can also be recruited to the plasma membrane independently of cAMP via activated ezrin, radixin, and moesin.
Dynamic control of between-endothelial cell adhesions by cAMP that can be stimulated by prostaglandins and atrial natriuretic peptide is mediated by both PKA and RapGEF3 [903]. Junctional Rap1 GTPase is indeed activated by RapGEF3 agent. The cAMP–RapGEF3–Rap1 axis impedes vascular permeability, as it stabilizes adherens junctions. Cerebral cavernous malformation protein CCM1203 actually contributes to signaling between adhesion molecules and the cytoskeleton. It complexes with VE-cadherin, α-, β-, and δ-catenins, and cytoskeleton-anchoring Rap1 effector AF6204 and transmits Rap1 signaling to cell adhesion plaques to tighten intercellular contacts. In addition, CCM1 binds to microtubules to regulate endothelial cell shape.
RapGEF5
Rap(Ras)GRPs
Protein RapGEF5 connects to Rap1 and, in vitro, mRasGTP; RapGEF1 bind Rap1 and Rap2; RasGRP3 activates both Rap1 and Ras [1020]. All these 3 types of GEFs can promote ELk1 activation.
Guanine nucleotide-releasing proteins for Ras (RasGRPs) that are calcium- and diacylglycerol-activated GEFs206 (Sect. 9.4.1.7) constitute a category of Rap guanine nucleotide-exchange factors that abound in the central nervous system.
Protein RasGRP2 has a substrate specificity for Rap1a GTPase. The basal ganglia of the brain is well supplied with RasGRP2 activator. It precludes the Ras-dependent activation of the ERK cascade. Like RasGRP2, RasGRP1 is synthesized in hematopoietic cells. Yet, RasGRP1 fails to activate Rap1a, but activates hRas and rRas in response to Ca
and diacylglycerol.

DOCK4
Rap1GDS1
Protein Rap1 GTP–GDP dissociation stimulator-1 (Rap1GDS1)207 catalyzes GDP exchange for GTP on Rap1a and Rap1b, as well as other small GTPases, such as kRas, Rac1, Rac2, RhoA, RalB, and CDC42 [1020, 1021]. Besides, Rap1a and kRas can be antagonist. However, kRas as well as RhoA and Rac2 are more important substrates for Rap1GDS1 than Rap1a [1021].
9.4.1.8 RasGEFs
Members of the RAS hyperfamily impinge on cell proliferation, differentiation, and apoptosis. Upon gene mutation, these oncogene products provoke cell transformation. They are counteracted by Rap1 GTPase. Guanine nucleotide-exchange factor for Ras GTPases is also called Ras guanine nucleotide-dissociation stimulator (RasGDS).
Families of the RASGEF Superfamily
Three main families of guanosine nucleotide-exchange factors are associated with Ras GTPases (hRas, kRasA, kRasB, and nRas) and link plasmalemmal receptors to Ras activation [1022]: (1) Son of sevenless (SOS1–SOS2); (2) Ras guanosine nucleotide-releasing factors (RasGRF1–RasGRF2); and (3) Ras guanosine nucleotide-releasing proteins (RasGRP1–RasGRP4).
In fact, the superfamily of guanine nucleotide-exchange factors for Ras GTPases also encompass (1) RapGEF1 to RapGEF6; (2) members of the Ral guanine nucleotide-dissociation stimulator family (RalGDS, Ras association [RalGDS–AF6] and pleckstrin homology domain-containing protein RAPH1 [or lamellipodin], and RalGDS-like proteins RGL1, RGL2, and RGL4); (3) RalGEFs with PH domain and SH3-binding motif (RalGPS1–RalGPS2); (4) breast cancer anti-estrogen resistance BCAR3 (a CDC42GEF and RacGEF);208 Rap1GDS1 (Rap1, GTP–GDP dissociation stimulator-1);209 and phospholipase-Cε [1028].
The RasGEF superfamily can be subdivided into 2 epifamilies according to sequence similarities: CDC24- and CDC25 motif-containing RasGEFs. The catalytic domain of RasGEFs, RasGRFs, and RasGRPs corresponds to catalytic CDC25 homology region. Proteins of the RASGEF category that possess a CDC24 motif act on CDC42 GTPase.
Signaling Mediated by RasGEFs
Activators of the SOS and RASGRP categories link plasmalemmal or cytoplasmic Tyr kinases to Ras GTPases via GRB2 adaptor and diacylglycerol (synthesized by phospholipase-Cγ), respectively [1022].210 Most cell types use primarily SOS proteins. In lymphocytes, the main GEF involved in the TCR-mediated activation of Ras is diacylglycerol-dependent RasGRP1.211
Receptor Tyr kinases can be transactivated by Gi- and Gq-coupled receptors to stimulate extracellular signal-regulated kinases (ERK1 and ERK2). In addition, Gβγ dimer associated with Gi- and Gq-coupled receptors can activate Ras via Src kinase, GRB2 adaptor, and SOS activator.212 Moreover, Gi and Gq subunits can activate ERK1 and ERK2 by binding to Rap1GAP1, thereby inhibiting Rap1 that antagonizes Ras GTPase. In particular, Gq-coupled serotonin receptors 5HT2 can trigger the PLC–IP3–Ca
–RasGRP1 and PLC–IP3–Ca
–RasGRF1 pathways. Similarly to RTKs, Gs-coupled receptors can launch the MAPK module via Ras GTPase. Gs-coupled serotonin receptors 5HT4B and 5HT7A excite ERK1 and ERK2 kinases, without transactivation of epidermal growth factor receptors, via Ras, but not Rap1 [1024]. In summary, RasGRP1 is stimulated not only from receptor (e.g., growth factor receptors) and cytosolic Tyr kinases, but also GPCRs (e.g., serotonin HT1 and HT2, dopamine, and prostaglandin receptors) and protein Tyr phosphatase receptors.


On the other hand, RasGRFs are coupled to G-protein-coupled receptors.213 These receptors support RasGRF translocation to the plasma membrane as a result of Ca
influx and their enzymatic activity on account of phosphorylation [1022]. However, RasGRFs do not work only downstream from G-protein-coupled receptors. For example, RasGRF2 also participates in T-cell signaling [1025].

The plasma membrane is not the exclusive platform from which Ras regulates signaling. Activation of Ras at distinct subcellular sites represents a mechanism for signal diversification from a given receptor. Whereas kRas resides exclusively at the plasma membrane, hRas and nRas also lodge in organelle membranes, such as that of the endoplasmic reticulum and Golgi body. Both hRas and nRas can then engage cRaf at organelle membranes during cell signaling with different outcomes. Activation of JNK and ERK is more efficient in the endoplasmic reticulum and Golgi body, respectively [1026]. In T lymphocytes, the plasma membrane remains the single site of TCR-driven nRas activation [1027]. On the oher hand, in response to Src-dependent activation of phospholipase-Cγ1, RasGRP1 can translocate to the Golgi body where it then activates Ras.
RasGRPs
Ras guanine nucleotide-releasing proteins are activated synergistically by Ca
and diacylglycerol (Sect. 9.4.1.6). Four known members (RasGRP1–RasGRP4) are produced in various tissues. Activation of most RasGRPs relies on phospholipase-Cγ-dependent generation of diacylglycerol that facilitates RasGRP translocation to membranes and association with target GTPases (Table 9.46). In neurons, RasGRP2 principally activates Rap GTPases, whereas RasGRP1 activates Ras GTPases.214

Type | Effectors |
---|---|
[signaling axis] | |
RasGRP1 | hRas, kRas, nRas, rRas1–rRas3 |
(hRas > kRas, nRas) | |
[Ras–bRaf–MAP2K3/6–P38MAPK pathway] | |
RasGRP2 | kRas, nRas, rRas1–rRas2, Rap1a, Rap2a |
[Rap–cRaf–MAP2K1/2–ERK1/2 pathway] | |
RasGRP3
![]() Stay updated, free articles. Join our Telegram channel![]() Full access? Get Clinical Tree![]() ![]() ![]() |