Skip to main content

G12/13 signaling in asthma

Abstract

Shortening of airway smooth muscle and bronchoconstriction are pathognomonic for asthma. Airway shortening occurs through calcium-dependent activation of myosin light chain kinase, and RhoA-dependent calcium sensitization, which inhibits myosin light chain phosphatase. The mechanism through which pro-contractile stimuli activate calcium sensitization is poorly understood. Our review of the literature suggests that pro-contractile G protein coupled receptors likely signal through G12/13 to activate RhoA and mediate calcium sensitization. This hypothesis is consistent with the effects of pro-contractile agonists on RhoA and Rho kinase activation, actin polymerization and myosin light chain phosphorylation. Recognizing the likely role of G12/13 signaling in the pathophysiology of asthma rationalizes the effects of pro-contractile stimuli on airway hyperresponsiveness, immune activation and airway remodeling, and suggests new approaches for asthma treatment.

Background

G Protein Coupled Receptors (GPCRs) comprise the largest family of cell surface receptors in the human genome (> 800 members) [1,2,3] and coordinate physiological responses to everything from photons to proteins. In contrast to this diversity of receptors and ligands, the heterotrimeric G proteins that mediate intracellular signaling downstream of GPCR activation seem deceptively simple: sixteen Gα subunits that fall into four mechanistic subfamilies (Gs, Gq/11, Gi/o and G12/13) and five Gβ subunits that form obligate heterodimers with one of twelve Gγ subunits [4]. GPCR signaling is typically described as resulting from specific coupling between a GPCR and a particular G protein, but most GPCRs couple to multiple G proteins [5]. The structural determinants of G protein selection by GPCRs are not well understood [6, 7]. Differential G protein recruitment (G protein bias) is often governed by ligand binding [8, 9] with signaling outcomes determined by the ensemble of G proteins that are activated. Elucidating the structural and mechanistic basis of G protein bias and using it as a tool for improving drug properties is the focus of considerable research [10,11,12].

Ligand binding to GPCRs promotes conformational changes that catalyze guanine nucleotide exchange within the α subunits of associated heterotrimeric G proteins [13]. GTP binding dissociates G protein heterotrimers, revealing protein interaction sites that mediate downstream signaling [14]. Although both Gα and Gβγ complexes contribute to signaling downstream of GPCR activation, signaling pathways are typically categorized by their Gα subunit. Activation of Gs stimulates adenylyl cyclase, which increases the cytosolic concentration of cyclic AMP (cAMP) thereby activating protein kinase A (PKA), while activation of Gi/o inhibits adenylyl cyclase [15]. Activation of Gq/11 stimulates phospholipase Cβ (PLCβ), which generates inositol trisphosphate (IP3) and diacylglycerol (DAG), thereby promoting intracellular calcium flux and activation of protein kinase C (PKC) [16].

In 1991, the G12/13 subfamily was identified as the fourth class of Gα subunits. This subfamily consists of G12 and G13 subunits, which share 67% amino acid sequence homology [17]. Unlike other Gα protein subunit subfamilies, activated G12 and G13 do not regulate enzymes that produce small molecule second messengers such as cAMP, IP3 or Ca2+. Instead, GTP-bound G12/13 subfamily members bind to and regulate the Ras homology (Rho) family of guanine nucleotide exchange factors (RhoGEFs) [18, 19]. RhoGEF complexes activate small Rho GTPases, such as RhoA, which play critical roles in regulating cytoskeletal dynamics [20]. Rho GTPases also stimulate numerous downstream signaling pathways through activation of Rho kinase (ROCK), Lim kinase (LIMK) and c-Jun NH2-terminal kinase (JNK; see Fig. 1) [21]. Deactivation of G12/13 occurs through hydrolysis of GTP to GDP via their intrinsic GTPase activity, which can be accelerated by GTPase-activating proteins (GAPs) including regulators of G protein signaling (RGS) domains of RhoGEF family members [22]. G12/13 couple to more than 30 GPCRs, including angiotensin II receptors, cysteinyl leukotriene receptors (CysLTR), histamine (H) receptors, lysophosphatidic acid (LPA) receptors, protease-activated receptors (PAR), sphingosine-1-phosphate (S1P) receptors, and thromboxane A2 receptors [5].

Fig. 1
figure 1

G protein signaling in smooth muscle contraction. Two G protein signaling pathways contribute to airway smooth muscle contraction. Activation of Gq/11 upon receptor-dependent guanine nucleotide exchange stimulates the calcium-dependent contractile pathway, whereby GTP-bound Gq/11 allosterically activates PLCβ-dependent hydrolysis of phosphoinositide bisphosphate (PIP2) into IP3 and DAG, which promote intracellular calcium flux, thereby activating MLCK-dependent phosphorylation of myosin light chain and actomyosin cross-bridge cycling (blue nodes on right side of figure). Calcium sensitization (fuchsia and violet nodes in the center and left of the figure) is mediated by GTP-bound RhoA, which can be generated downstream of activation of either Gq/11 or G12/13, although G12/13 are more potent activators [29]. PKC phosphorylation of CPI-17 promotes inhibition of myosin light chain phosphatase, which increases net MLC phosphorylation. GTP-bound RhoA stimulates actin polymerization through activation of formins (fuchsia nodes) and ROCK activity (violet nodes), which inhibit both filament severing and MLCP. ROCK also phosphorylates CPI-17, which further inhibits MLCP. (Created with BioRender.com)

Asthma is an obstructive airway disease characterized by airway hyperresponsiveness (AHR), inflammation and airway remodeling (AR). GPCRs play critical roles in regulating bronchomotor tone, and, as such, are the targets of numerous therapeutics that are used to treat asthma and other obstructive pulmonary diseases. GPCRs coupled to Gs, such as the β2 adrenergic receptor (β2AR), regulate relaxation of airway smooth muscle, while GPCRs coupled to Gq/11, such as histamine, leukotriene and muscarinic acetylcholine receptors, promote airway constriction. Notably, G12/13 couples to many Gq/11 coupled receptors in vascular and airway smooth muscle [5], and the RhoA signaling pathway downstream of G12/13 activation potentiates smooth muscle contraction [21]. Inflammation plays a fundamental role in the etiology of asthma, thus biologics and corticosteroids are mainstays of therapy. G12/13 dependent signaling plays an important role in lymphocyte activation and migration. Finally, airway thickening and angiogenesis are stimulated in patients with asthma. Increased expression of G12/13 has been reported in animal models of asthma [23], numerous proliferative pathways (mitogen-activated protein kinase (MAPK) [24], Hippo [25], non-receptor tyrosine kinases [26]) are downstream of G12/13 activation, and G13 has been implicated in both airway smooth muscle (ASM) proliferation and angiogenesis in a variety of physiological and pathological settings, including development and cancer [27, 28].

As the most potent upstream modulator of RhoA [29], G12/13 and its downstream signaling pathway constitutes an intriguing target with the potential to impact myriad aspects of the pathophysiology of asthma by inhibiting AHR, inflammation and AR. Since there are limited publications on G12/13 signaling in ASM and asthma, each section of this review summarizes relevant findings from non-muscle tissues first and concludes with smooth muscle or ASM data where available. The preponderance of the evidence suggests that targeting pathways that activate the G12/13 subfamily and its downstream signaling axis may provide a complementary therapeutic approach for managing asthma and other obstructive pulmonary diseases.

G protein stimulation of myosin light chain phosphorylation regulates smooth muscle contraction and airway hyperresponsiveness

Airway smooth muscle shortening induces narrowing of airways, evoking wheezing, difficulty breathing and chest tightness, which, when coupled with mucus buildup and airway inflammation, is characteristic of asthma [30]. Bronchoconstriction is thought to be mediated by agonist binding to Gq/11 coupled receptors. The guanine exchange activity of agonist-bound GPCRs enables GTP-bound Gq/11 to allosterically activate PLCβ-dependent production of IP3 and DAG, thereby stimulating IP3 receptor-dependent Ca2+ release from the sarcoplasmic reticulum and PKC-dependent activation of Ca2+ channels, which cooperate to increase the concentration of calcium in the cytosol (blue nodes in Fig. 1). Cytosolic Ca2+ binding to calmodulin evokes allosteric activation of myosin light chain kinase (MLCK), which phosphorylates the 20 kDa light chain of myosin to promote actomyosin cross-bridge cycling and muscle contraction [31, 32]. Calcium-bound calmodulin in smooth muscle also activates calmodulin-dependent kinase II, which cooperates with PKC to relieve tonic inhibition of the interaction between myosin and actin by calponin [33]. Interestingly, however, the Gq/11 depsipeptide inhibitor FR900359 (FR) failed to inhibit carbachol-dependent contraction of mouse precision-cut lung slices at 30 nM concentration [34]. A higher concentration of FR (1 µM) only partially inhibited carbachol-dependent contraction of human precision-cut lung slices [35] suggesting that actomyosin cross-bridge cycling still occurs even when Gq/11-stimulated calcium flux is fully inhibited. If Gq/11-dependent Ca2+ flux is insufficient for actomyosin crossbridge cycling, what regulates the effects of acetylcholine on smooth muscle tone?

G12/13 activates the RhoA/ROCK signaling pathway to regulate actin polymerization and inhibit MLCP

The interaction between myosin and actin in smooth muscle is regulated through phosphorylation of myosin light chain, and by maintaining a large pool of depolymerized actin [36]. Both myosin light chain phosphorylation and actin polymerization are necessary for smooth muscle contraction (Fig. 1) [37, 38].

The actin cytoskeleton is a highly dynamic structure that is regulated by polymerization and depolymerization processes interconverting globular (G) and filamentous (F) actin. This mechanism is particularly true in smooth muscle, where the ratio of F actin to G actin is three to four times less than what is observed in cardiac and skeletal muscle [36]. The Rho family of small GTPases -- Cdc42, Rac1, and RhoA -- regulate a diverse array of cytoskeletal dynamics [39, 40]. GTP-bound RhoA activates formins, which stimulate actin polymerization (fuchsia nodes in Fig. 1) [37]. Since each myosin head interacts with two actin monomers in F actin, polymerization of actin is essential for actinomyosin complex assembly and muscle contraction [41]. Microinjection of activated G12 or G13 into Swiss-3T3 cells is sufficient to induce actin polymerization in a RhoA-dependent manner [42].

GTP-bound RhoA also activates ROCK, which phosphorylates the myosin targeting subunit (MYPT1) of myosin light chain phosphatase (MLCP). MYPT1 phosphorylation inhibits MLCP binding to myosin light chain (MLC), thereby increasing net MLC phosphorylation (this is referred to as calcium sensitization – see violet nodes in Fig. 1) [43]. ROCK therefore acts synergistically with MLCK to prolong and strengthen muscle contraction [21]. Both ROCK and PKC phosphorylate and activate CPI-17, a smooth muscle specific MLCP inhibitor, which enhances the pro-contractile effect [44]. Polymerized actin is further stabilized through inhibitory phosphorylation of cofilin by LIMK [45], which is activated upon phosphorylation by ROCK. Cofilin phosphorylation impairs binding to F actin [46] thereby inhibiting filament severing and actin depolymerization [47, 48]. In tracheal smooth muscle, acetylcholine-induced stiffness is independent of MLC phosphorylation and calcium flux, but dependent on ROCK [49]. ROCK inhibition blocks contraction of airway smooth muscle both in vitro and in vivo [50,51,52].

Small GTPases of the Ras superfamily, such as RhoA, must be activated by guanine nucleotide exchange factors to stimulate downstream signaling RhoGEFs can be activated by either GTP-bound Gq/11 or G12/13, but GTP-bound G12/13 are the more potent activators [29]. RhoA and other Ras family members are also regulated by guanine nucleotide dissociation inhibitors (GDIs). GDIs prevent small GTPases from participating in nucleotide exchange, holding them in an inactive state and in some cases blocking them from trafficking to the membrane [53, 54]. In vascular smooth muscle, constitutively activated G12 or G13 promote vasoconstriction, which can be inhibited by botulinum C3 toxin or ROCK inhibition, while dominant-negative forms of G12 or G13 inhibit vasoconstriction [55]. Similarly, conditional knockdown of G12/13 or leukemia-associated RhoGEF (LARG) normalized age-related hypertension in mice [56], and blocked salt-induced hypertension [57], while loss of either G12/13 or the smooth muscle specific RhoGEF, ARHGEF12, in small arteries induced loss of RhoA activation and vasodilation [58]. In a rat model, chronic administration of angiotensin II resulted in increased expression of G12 and elevated blood pressure, while co-administration of GNA12 antisense lowered mean blood pressure [59].

G12/13 couples to several pro-contractile GPCRs in airway smooth muscle cells (H1, CysLTR2, PAR 1 & 2) [5], and G12 and G13 are overexpressed in AHR rats compared to healthy animals [23]. Controversy exists in the optical biosensor literature concerning whether muscarinic 3 acetylcholine receptors (M33R) interact with G12/13 [5, 8, 34], but functional assays suggest interactions between M3R and G12/13 in both human embryonic kidney (HEK293) and human airway smooth muscle (HASM) cells. M3R-dependent activation of G12 in HEK293 cells stimulates phospholipase D activity [60, 61]. Knockdown of GNA13 in primary bronchial smooth muscle cells blocked methacholine-induced phosphorylation of myosin light chain [62]. In HASM cells, interaction between M3R and G12 was confirmed by co-immunoprecipitation, siRNA knockdown of GNA12 reduced phosphorylation of MYPT1 and MLC, expression of a G12 inhibitor (p115RhoGEF-RGS) suppressed carbachol-mediated HASM cell contraction, and RhoA inhibition induced dilation of human precision cut lung slices [63]. Collectively, these data suggest that G12/13 couples to most therapeutically relevant pro-contractile GPCRs in both HEK293 and HASM cells.

Levels of TGF-β1, a cytokine that regulates extracellular matrix formation, cell growth, inflammation, and the epithelial-mesenchymal transition (EMT) are increased in airways of patients with severe asthma [64]. Published research suggests that G12/13 regulates TGF-β1 expression, although the specifics of the regulatory mechanism are unclear. In HASM cells, pretreatment with TGF-β1 increases RhoA translocation via p115RhoGEF. In G12/13 deficient cells, TGF-β1 expression was restored after transfection with constitutively active G12/13 QL mutants, suggesting that G12/13 play a role in modulating TGF-β1 expression levels [65]. TGF-β1 increases bronchomotor tone and enhances carbachol- and histamine-induced excitation-contraction coupling in human airway smooth muscle and precision cut lung slices, respectively. In isolated HASM cells, methacholine-induced cytoskeletal stiffness was increased after TGF-β1 pre-treatment in a dose-dependent manner. Inhibition of ROCK attenuated TGF-β1 induced single-cell contraction, while having no impact on intracellular calcium release [64]. These data indicate that TGF-β1-mediated physiological effects in HASM occur in a G12/13-dependent manner.

In summary, smooth muscle contraction is regulated by Gq/11 dependent activation of MLCK, which is enhanced by ROCK dependent inhibition of MLCP, and by RhoA-dependent actin polymerization, which is stimulated more robustly by activated G12/13 than by activated Gq/11. The pro-contractile and AHR-inducing effects of TGF-β1 also appear to be regulated by G12/13 signaling. The complex interplay between the Gq/11 and G12/13 signaling pathways leads to impaired muscle shortening when RhoA/ROCK activation is compromised, and in elevated resting tone unless both pathways to RhoA/ROCK activation are inhibited.

G12/13 signaling regulates immune activation and infiltration

Asthma is a chronic disease mediated by immune infiltration in which both symptoms (e.g., mucus production) and pathology (AHR, AR) are driven by inflammatory processes [66]. Asthma patients display heterogeneous clinical phenotypes, in part due to the spectrum of immune mechanisms that are dysregulated in patients with asthma. In approximately half of all patients, asthma is characterized by type 2 immune response and sensitization to allergens [67]. Immune responses in these patients result from production of IgE and Th2-associated cytokines (IL4, IL5, IL13), which stimulate dendritic and mast cell activation, eosinophil invasion and mucus production [68]. As such, these patients are often responsive to inhaled corticosteroids and biologics that antagonize Th2-associated cytokine signaling. In contrast, patients with minimal production of Th2-associated cytokines are often poorly responsive to inhaled corticosteroids. Inflammation in these patients is driven by macrophages, neutrophils and cytokines like TGF-β, IL-1β and IL-6 [69].

G12 and G13 have been implicated in various aspects of immune cell activation and trafficking (Fig. 2). S1P is released by mast cells in response to IgE stimulation. S1P receptors on mature dendritic cells coordinate expression of Th2 cytokines in a G12/13-dependent manner (Fig. 2a) [70, 71], and both S1P concentration and GNA12 expression predict asthma control [72]. In the central nervous system, S1P activates receptors on astrocytes that signal via G12/13, RhoA, phospholipase C-epsilon and protein kinase D to stimulate NFκB dependent transcription of inflammatory genes (Fig. 2b) [73]. Biased inhibition of G12/13 signaling downstream of PAR2 activation blocks inflammation in vivo [12]. Diminished follicular helper T (Tfh) response was observed in a T-cell specific Gα13 deficient mouse model, and G13 knock-out Tfh cells showed an inability to transduce signal through RhoA mediated ROCK activation, highlighting the regulatory role of the G13-Rho-Rock signaling axis in Thf cell differentiation [74].

Fig. 2
figure 2

G12/13 signaling in inflammation. G12/13-dependent activation of RhoA and ROCK promotes inflammation in both Th2-high (a) and Th2-low asthma (c). G12/13 can also stimulate NFκB-dependent production of inflammatory cytokines (b). (Created with BioRender.com)

The expression level of activating protein 1 (AP-1), a transcription factor that is necessary for the proliferation and differentiation of Th2 cells, was reduced in G12/13 knockdown cells [62]. Myeloid-specific knockdown of G12/13 results in increased expression of anti-inflammatory genes in macrophages, which protect against atherosclerosis in wild-type mice [75]. GNA12 regulates C5a-mediated migration in macrophages, indicating an anti-inflammatory role for GNA12 in inflammatory bowel disease [76, 77]. Loss of RGS-containing RhoGEFs such as ARHGEF1, LARG, PRG or p115 that activate RhoA and ROCK downstream of G12/13 compromise immune function (Fig. 2c) [78, 79]. G13 knockdown in microglial cells inhibits LPS-induced activation and Rac-dependent migration [80]. In platelets and leukocytes, outside-in signaling between integrins and G13 regulates Rho-dependent, pro-inflammatory secretion and thrombosis [81,82,83,84]. G12/13 are also implicated in cytokine signaling and inflammation in prostate cancer [85] and mediate renal ischemia-reperfusion injury [86,87,88].

Published evidence suggests that G12/13 and downstream RhoA/ROCK signaling mediate inflammation in diverse physiological and pathological settings. Inhibiting this pathway may cooperate, or even synergize, with biologics and inhaled corticosteroids in Th2 high asthma, and provide a novel approach to suppress inflammation in patients who are refractory to corticosteroid therapy.

G12/13 regulation of airway remodeling and hyperplasia

AR is a hallmark of the pathophysiology of asthma [89]. In airways, hyperproliferation evokes thickening of both smooth muscle and the extracellular matrix, which narrows bronchi and predicts poor patient outcomes [90]. HASM cells from patients with asthma proliferate faster than ASM cells from control patients [91, 92]. Activation of Gq/11-coupled receptors stimulates MAPK, which promotes proliferation of ASM and contributes to hyperplasia, angiogenesis and tissue remodeling in a wide variety of contexts [93]. Proliferation downstream of the G12/13 –RhoA signaling axis is regulated by kinases, pro-proliferative transcription factors, and alterations in gene expression (Fig. 3).

G12 was the first Gα subunit that was shown to be capable of oncogenic transformation upon overexpression [94, 95]. Introduction of mutations that inhibit the intrinsic GTPase activity of G12 (Q229L) resulted in focus forming activity in NIH-3T3 cells that was similar to the most potent viral oncogenes [95, 96]. Similarly, overexpression of G13 is sufficient to transform NIH-3T3 fibroblasts and to promote xenograft growth in nude mice, and the GTPase deficient mutant (Q226L) is a potent oncogene [96, 97]. These genes, which were cloned from a Ewing sarcoma plasmid library (gep oncogenes), are dysregulated in numerous human cancers (reviewed in [98]). Dysregulation of G12/13 signaling has been implicated in cardiac remodeling, angiogenesis and hypertrophy [99,100,101,102], in growth factor-stimulated cell migration [103], and in hepatic, cardiac and pulmonary fibrosis [104,105,106].

The proliferative effects of dysregulated G12/13 signaling were originally attributed to activation of the JNK pathway [24]. Expression of constitutively active G12 leads to increased JNK activity and c-Jun phosphorylation (Fig. 3a) [107]. G12/13-dependent c-fos activation via the serum-response element (SRE) has also been implicated in gep oncogenesis [108]. Serum response factor (SRF) is a transcriptional activator that binds SRE in the promoter region of genes involved in pro-proliferative signaling pathways. Actin polymerization stimulated by RhoGEF/RhoA activation induces translocation of myocardin-related transcription factor A (MRTFA) to the nucleus, where it functions as a co-activator with SRF (Fig. 3b) [109,110,111]. AP-1 dependent transcription serves as an integrated read-out of the actin polymerization (c-fos/SRE) and ROCK stimulation (JNK/c-Jun) functions of activated RhoA [112]. AP-1 activation stimulates expression of MDM2, which promotes degradation of p53 and FOXO1, thus driving malignancy and EMT [113, 114]. G12/13-dependent activation of the JNK pathway also promotes cell migration [78] and invasion [115]. In smooth muscle, S1P dependent activation of G12/13 stimulates growth and proliferation in a RhoA and SRE-dependent manner [116].

Fig. 3
figure 3

G12/13 signaling in proliferation. G12/13 activate RhoGEFs that generate GTP-bound RhoA, activating ROCK and stimulating proliferative gene transcription via AP-1 (a) and SRE (b) promoters. GTP-bound RhoA inhibits phosphorylation of Lats 1/2, thereby blocking negative regulation of the Hippo pathway (c). G12/13 bind to c-Abl, affecting ERK-dependent proliferation (d). (Created with BioRender.com)

The Hippo signaling pathway can modulate cell proliferation downstream of G12/13 induced RhoA signaling. LPA, S1P, and PAR signaling through G12/13 and RhoA inhibits phosphorylation of Lats 1/2, which blocks negative regulation of the oncogenic transcription factors YAP and TAZ (Fig. 3c) [117, 118]. In glioblastoma, S1P/G12/13/RhoA-mediated YAP activation promotes cellular invasion and metastasis [110], while LPA activation of G12/13 and RhoA is implicated in YAP-mediated hepatocellular carcinoma [119]. In addition to affecting Lats 1/2 phosphorylation, G12/13 and RhoA-dependent activation of YAP and TAZ is also significantly regulated by actin polymerization [120]. In vascular smooth muscle cells, knockdown of G12/13, RhoA inhibition, and/or disruption of actin cytoskeleton formation all prevented YAP/TAZ activation, thereby inhibiting tissue remodeling [25]. The response of smooth muscle to mechanical stretch is mediated by the interaction between G13 and integrins, which inhibits Gα13-dependent RhoA activation and YAP/TAZ transcription, thereby preventing proliferation, inflammation, and angiogenesis [84]. G13 binding to integrins stimulates the non-receptor tyrosine kinase c-Src, which promotes cell attachment and spreading [121]. In contrast, focal adhesion kinase (FAK) is activated in a G13-dependent fashion downstream of PAR [29], LPA [122], GRP4 [123] and CCK2 [124], causing sustained RhoA activation that promotes cell migration in ovarian, skin and colon cancer.

Abelson tyrosine kinase (c-Abl) is a primary regulator of actin dynamics and contraction in airway smooth muscle [125]. c-Abl inhibition attenuates actin polymerization, but fails to impact pro-contractile myosin phosphorylation [126]. In platelets, proline-rich tyrosine 2 (Pyk2) activation downstream of G12/13 regulates shape change. Co-immunoprecipitation studies have shown that G13 interacts directly with c-Abl tyrosine kinase in endothelial cells, regulating actin cytoskeletal dynamics and inducing cell remodeling and cell migration [26]. In smooth muscle cells, knockdown of c-Abl inhibited ERK1/2 phosphorylation, a key mediator of smooth muscle proliferation (Fig. 3d) [127].

G12/13 -mediated signaling through JNK, YAP/TAZ and non-receptor tyrosine kinases is implicated in proliferation, angiogenesis and tissue remodeling in diverse disease pathologies, including vascular and airway smooth muscle hypertrophy. Antagonizing pathways that activate G12/13 and its downstream signaling axis may suppress several hallmarks of AR, including increased smooth muscle mass.

G12/13 pathway pharmacology

Therapeutic suppression of G12/13 signaling can be achieved by balanced or biased antagonism of upstream receptors, direct G protein inactivation, blocking of activation-dependent protein-protein interactions or through inhibition of downstream signaling pathways (Fig. 4).

Fig. 4
figure 4

Targets for regulating G12/13 signaling. Hyperactive G12/13 signaling in airway smooth muscle can be attenuated by antagonizing upstream receptors (red), by developing direct inhibitors of G12, G13 (green) or complexes between G12/13 and RhoGEFs (blue), or by inhibiting RhoA or ROCK (purple). (Created with BioRender.com)

Humankind has been treating symptoms of asthma with anticholinergic agents for millennia. The Ebers Papyrus (1550 BC) recommends burning of henbane, which produces anticholinergic tropane alkaloids, to “remove phlegm, alleviate coughs and ease breathing” [128]. Extracts from a variety of other nightshade family plants figure prominently in treatment of dyspnea throughout Western medicine [129], including atropine, which was purified from deadly nightshade (Atropa belladonna) in 1833 [130], and was used to treat symptoms of asthma until the 1970s, when tropane derivatives with improved side effect profiles became available [131]. Despite (or, perhaps, because of) this long, empirical history of asthma treatment with anticholinergic agents, the effects of muscarinic antagonists have largely been evaluated physiologically rather than pharmacologically. This is particularly true for the effects of muscarinic antagonists on G12/13 or RhoA signaling (highlighted in red in Fig. 4). For GPCRs like the M3R that display promiscuous G protein binding, antagonists of one G protein signaling pathway can function as agonists of another [132, 133], or can be biased to prefer one G protein signaling pathway over another [134]. For example, G12 biased agonists of the apelin receptor demonstrate sustained cardiac response [135]. The relative importance of Gq/11 versus G12/13 signaling in ASM contraction, the effects of these pathways on therapeutic response to M3R agonists and antagonists, and the utility of modulating G protein signaling bias in obstructive airway diseases remains to be elucidated. Detailed study of the effects of muscarinic antagonists on Gq/11 versus G12/13 signaling may provide insights that can improve disease management for asthma patients, or inform development of novel anticholinergics with enhanced potency, efficacy, and tolerability.

Agonists of the β2AR, which stimulate PKA-dependent activation of MLCP and smooth muscle relaxation, are the standard of care for asthma patients of every age and every stage of disease. Short-acting beta agonists (SABA) are used symptomatically to relieve acute asthma exacerbations, while long-acting beta agonists (LABA) are used as maintenance therapy by patients with persistent asthma [136]. Although agonism of relaxation is a common approach to treat acute smooth muscle constriction (e.g., asthma, premature labor), other chronic diseases are often treated with antagonists (e.g., angiotensin converting enzyme inhibition or angiotensin receptor blocking in hypertension [137]; anticholinergics or alpha-blockers in detrussor hyperreflexia [138, 139]). Activated GS is the least oncogenic Gα subunit [97], and cAMP is generally antiproliferative [140], but chronic, unbiased beta-agonism causes iatrogenic eosinophila, mucus production and MAPK activation [141], which may contribute to the increased risk of fatal asthmatic attacks observed in patients who are treated with LABAs [142, 143]. In principle, antagonism of hyperresponsive constrictive pathways (bronchoprotection) would circumvent the adverse therapeutic effects associated with LABA treatment [144], and anticholinergics are considered safe and effective for the treatment of chronic obstructive pulmonary disease (COPD) [145], but FDA-approved, long-acting muscarinic antagonists (LAMAs) display minimal benefit as add-on treatment for patients whose asthma is not well controlled with standard therapies [146, 147]. Notably, this conclusion is based on acute (bronchoconstriction) rather than chronic (AR) endpoints. Given the chronic activation of oncogenic Gq/11 and G12/13 signaling that is characteristic of AHR, it would be worthwhile to assess whether anticholinergics that are able to suppress both Gq/11 and G12/13 signaling might be effective in reducing AR when co-administered with beta agonists and inhaled corticosteroids.

Unlike bronchorelaxation, which is regulated by a single receptor (β2AR), multiple receptors can evoke constriction of ASM. Inhibiting the downstream G protein (highlighted in green in Fig. 4) through which multiple bronchoprovocational agents signal might provide a unified mechanism for relieving bronchoconstriction, but there are no FDA approved drugs that target G proteins. Toxins, natural products and their derivatives regulate several subfamilies of G proteins [148,149,150,151,152], but no natural product activators or inhibitors that are specific to G12/13 subfamily members have been reported. As essential signaling proteins that are expressed in most tissues, G proteins are typically considered to be poor therapeutic targets, but G12 may be an exception. G12 knockout mice are viable, fertile and display minimal developmental abnormalities [153]. Nevertheless, G12 knockdown inhibits contraction of vascular and airway smooth muscle [59, 63] and GNA12 expression predicts asthma control [72]. Direct antagonism of G12 signaling may have considerable therapeutic value.

To transduce downstream signals, G12/13 relies on protein-protein interactions (PPIs) with RhoGEFs. The avidity that dominates the free energy of PPIs is difficult to outcompete with the affinity of protein-ligand interactions, which is why PPIs are considered challenging pharmacologic targets [154]. However, a variety of emerging approaches have succeeded in generating small molecules that can disrupt PPIs (highlighted in blue in Fig. 4) [155,156,157]. A recent study characterized the PPI between Gi and its GEF, Girdin, as a druggable interaction by performing high-throughput screening. The authors identified a small molecule, NF023, that disrupts GEF activity by binding to a site that overlaps with the Gi-Girdin interface but fails to disrupt formation of the inactive Gαβγ heterotrimer [158]. Analysis of the G12/13 interactome may reveal druggable PPI interfaces and enable discrimination of the distinct biological functions of G12 and G13. Indeed, the G13 interactome has already been validated as a therapeutic target. M3mP6, a peptide derived from the G13 binding ExE motif of the integrin beta3 cytoplasmic domain, has antithrombotic effects without off-target adverse symptoms. In a mouse model, post-ischemic injection of M3mP6 protected the heart from myocardial ischemia-reperfusion injury [159].

Receptor-activated G12/13 signals primarily through the RhoA/ROCK pathway. As such, inhibitors of RhoA or ROCK offer an alternative approach to interdict pro-contractile, pro-inflammatory and/or proliferative G12/13-mediated signaling downstream of multiple, pro-contractile GPCRs (highlighted in purple in Fig. 4). Although RhoA inhibitors (including C3 toxin from Clostridium botulinum) have been described [160, 161], these agents are used as research tools rather than therapeutics. In contrast, ROCK inhibitors are FDA approved for treatment of open-angle glaucoma and ocular hypertension [162] and for graft versus host disease [163], and the ROCK inhibitor fasudil is approved in Japan for treatment of cerebral vasospasm and subarachnoid hemorrhage. ROCK inhibitors have also been investigated in clinical trials as treatments for a variety of indications, including amyotrophic lateral sclerosis [164], Parkinson’s disease [165], solid malignancies [166], psoriasis [167], chronic kidney disease [168] and pulmonary hypertension [169]. Clinical trials in which ROCK inhibitors are administered systemically typically report few significant adverse events [170]. Hypotension and syncope are commonly observed, but these side effects were minimized in pulmonary hypertension trials by delivering the inhibitor (fasudil) to the lung by inhalation [171]. No human clinical trials evaluating the efficacy of ROCK inhibitors for the treatment of asthma have been registered to date, but ROCK inhibitors are effective in reducing inflammation [172, 173] and airway constriction [174, 175] in cellular and animal models of asthma. Clinical trials of ROCK inhibitors as asthma therapeutics are feasible, and their potential benefits to patients are well supported by ample pre-clinical data.

Numerous small molecules can interact with the downstream G12/13 signaling pathway to promote bronchorelaxation. PI3K inhibitors promote anti-inflammatory responses in mouse models, reducing mucus production and airway remodeling [176, 177]. Small molecule inhibitors of class I PI3Ks can act as bronchodilators in human airway smooth muscle cells, highlighting this pathway as a promising target for pharmacologic intervention [178, 179]. In mouse asthma models, the selective PI3K p110δ inhibitor IC87114 attenuated AKT phosphorylation and mitigated airway hyperresponsiveness [180]. Inhibiting class I isoforms directly with p110α knockdown, wortmannin, or the small molecule LY294002 resulted in diminished RhoA and MYPT1 activation, which suppressed MLC phosphorylation [181]. These inhibitors also diminished cell depolarization, a common trait of HASM contraction [182], and reduced glucocorticoid insensitivity in patients with severe asthma by restoring HDAC2 or inhibiting the activation of pro-inflammatory transcription factors [183].

Upstream receptors and downstream kinases are the most tractable targets for suppressing chronic G12/13 signaling. However, the effects of anticholinergics on G12/13 signaling have never been characterized, and the extent to which RhoA/ROCK or PI3K inhibition can mitigate the effects of G12/13 pathway activation is unknown.

Conclusions

The canonical model for smooth muscle contraction posits that actomyosin cross-bridge cycling is regulated primarily by calcium-dependent activation of myosin light chain kinase downstream of M3R and Gq/11. The intuitive satisfaction of this model benefits from the convergence of two conventional wisdoms from muscle and cell physiology: that muscle contraction is calcium dependent; and that kinase activity is induced while phosphatase activity is constitutive. It’s unclear whether either of these conventional wisdoms holds in airway smooth muscle. The calcium dependence of cardiac and skeletal muscle contraction is largely due to the need to relieve tonic inhibition of the interaction between myosin and actin by troponin, which isn’t expressed in smooth muscle [184]. The need for troponin in cardiac and skeletal muscle and calponin in smooth muscle suggests that the basal activity of MLCK is sufficient to promote constriction in the absence of tonic inhibition. The efficacy of β-agonists in asthma pharmacotherapy, which activate MLCP, indicates that bronchomotor tone may be regulated primarily by phosphatase activity rather than kinase activity. The inability of FR to block M3R-dependent contraction of airway smooth muscle [34, 35] is consistent with this view, and indicates the existence of a signaling pathway besides Gq/11 that is able to transduce signals from M3R through RhoA to inhibit MLCP and promote parasympathetic actomyosin cross-bridge cycling. The preponderance of the evidence strongly indicates that this critical signaling pathway is G12/13 [61,62,63], although additional experiments to resolve inconsistencies between functional and biosensor assays would be worthwhile.

Pathologic constriction of airway smooth muscle is the defining symptom of asthma [185]. Smooth muscle contraction relies on actin polymerization and phosphorylation of myosin light chain, both of which are mediated by hyperresponsive acetylcholine signaling to RhoA and ROCK [37], likely through G12/13 [29]. G12/13 signaling also stimulates production of Th2 cytokines [70], which promote airway inflammation, and activates numerous proliferative pathways (JNK [112], Hippo [84], c-Abl [26]) that may drive AR in hyperresponsive airways. The role of G12/13 signaling in airway constriction, inflammation and remodeling has been underappreciated despite compelling direct evidence for its significance [63]. Consequently, the effect of antagonists of constrictive and inflammatory pathways on G12/13, RhoA and ROCK activation has never been adequately characterized. Therapeutic strategies that antagonize upstream receptors or that inhibit G12/13 or its downstream signaling partners should benefit patients with asthma and likely synergize with current asthma treatments.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

GPCR:

G protein-coupled receptor

cAMP:

cyclic AMP

PKA:

Protein Kinase A

PLCβ:

Phospholipase Cβ

IP3:

Inositol Trisphosphate

DAG:

Diacylglycerol

PKC:

Protein Kinase C

Rho:

Ras Homology

GEF:

Guanine Exchange Factor

ROCK:

Rho-Associated Kinase

LIMK:

Lim Domain Kinase

JNK:

c-Jun NH2-terminal kinase

GAP:

GTPase Accelerating Protein

RGS:

Regulator of G Protein Signaling

CysLTR:

Cysteinyl Leukotriene Receptors

H:

Histamine

LPA:

Lysophosphatidic Acid

PAR:

Protease-Activated Receptor

S1P:

Sphingosine-1-Phosphate

AHR:

Airway Hyperresponsiveness

AR:

Airway Remodeling

β2AR:

Beta-2 Adrenergic Receptor

MAPK:

Mitogen-Activated Protein Kinase

ASM:

Airway Smooth Muscle

MLC:

Myosin Light Chain

MLCK:

Myosin Light Chain Kinase

FR:

Gq/11-selective depsipeptide inhibitor FR900359

G:

Globular

F:

Filamentous

MLCP:

Myosin Light Chain Phosphatase

MYPT:

Myosin Phosphatase Targeting Subunit

GDI:

Guanine Nucleotide Dissociation Inhibitor

LARG:

Leukemia-Associated RhoGEF

ARHGEF:

Rho Guanine Exchange Factor

GNA12:

Gene Encoding Guanine Nucleotide Binding Protein Subunit Alpha 12

GNA13:

Gene Encoding Guanine Nucleotide Binding Protein Subunit Alpha 13

M3R:

Muscarinic 3 Acetylcholine Receptor

HEK:

Human Embryonic Kidney

HASM:

Human Airway Smooth Muscle

siRNA:

Short Interfering RNA

TGF:

Tumor Growth Factor

EMT:

Epithelial-Mesenchymal Transition

IgE:

Immunoglobulin E

IL:

Interleukin

Th2:

T-helper 2

NFkB:

Nuclear Factor kB

Tfh:

T Follicular helper

AP-1:

Activator Protein 1

PRG:

PDZ Domain-Containing RhoGEF

Gep:

Gene from Ewing Sarcoma Plasmid Library

SRE:

Serum Response Element

SRF:

Serum Response Factor

MRTFA:

Myocardin-Related Transcription Factor A

MDM2:

Mouse Double Minute 2

FOXO1:

Forkhead Box Protein O1

Lats1/2:

Large Tumor Suppressors 1 & 2

YAP:

Yes-Associated Protein

TAZ:

Transcriptional Activator with a PDZ Binding Motif

c-SRC:

Cellular Rous Sarcoma Virus Homolog

FAK:

Focal Adhesion Kinase

GRP:

Gastrin-Releasing Peptide

CCK:

Cholecystokinin

c-Abl:

Cellular Abelson Murine Lymphosarcoma Virus Homolog Tyrosine Kinase

Pyk2:

Protein Tyrosine Kinase 2

ERK:

Extracellular Signal-Regulated Kinase

SABA:

Short-Acting Beta Agonist

LABA:

Long-Acting Beta Agonist

COPD:

Chronic Obstructive Pulmonary Disease

FDA:

Food and Drug Administration

LAMA:

Long-Acting Muscarinic Antagonist

PPI:

Protein-Protein Interaction

PI3K:

Phosphoinositide-3 Kinase

AKT:

Protein Kinase B/Gene that Causes Thyomas in the Akr Mouse Strain

HDAC:

Histone Deacetylase

PIP2:

Phosphoinositide Bisphosphate

References

  1. Fredriksson R, Lagerstrom MC, Lundin LG, Schioth HB. The G-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol Pharmacol. 2003;63(6):1256–72.

    Article  CAS  PubMed  Google Scholar 

  2. Pierce KL, Premont RT, Lefkowitz RJ. Seven-transmembrane receptors. Nat Rev Mol Cell Biol. 2002;3(9):639–50.

    Article  CAS  PubMed  Google Scholar 

  3. Yang D, Zhou Q, Labroska V, Qin S, Darbalaei S, Wu Y, et al. G protein-coupled receptors: structure- and function-based drug discovery. Signal Transduct Target Ther. 2021;6(1):7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hurowitz EH, Melnyk JM, Chen YJ, Kouros-Mehr H, Simon MI, Shizuya H. Genomic characterization of the human heterotrimeric G protein alpha, beta, and gamma subunit genes. DNA Res. 2000;7(2):111–20.

    Article  CAS  PubMed  Google Scholar 

  5. Hauser AS, Avet C, Normand C, Mancini A, Inoue A, Bouvier M et al. Common coupling map advances GPCR-G protein selectivity. Elife. 2022;11.

  6. Okashah N, Wan Q, Ghosh S, Sandhu M, Inoue A, Vaidehi N, et al. Variable G protein determinants of GPCR coupling selectivity. Proc Natl Acad Sci U S A. 2019;116(24):12054–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Masuho I, Kise R, Gainza P, Von Moo E, Li X, Tany R, et al. Rules and mechanisms governing G protein coupling selectivity of GPCRs. Cell Rep. 2023;42(10):113173.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Smith JS, Hilibrand AS, Skiba MA, Dates AN, Calvillo-Miranda VG, Kruse AC. The M3 muscarinic acetylcholine receptor can signal through multiple G protein families. Mol Pharmacol. 2024.

  9. Namkung Y, LeGouill C, Kumar S, Cao Y, Teixeira LB, Lukasheva V, et al. Functional selectivity profiling of the angiotensin II type 1 receptor using pathway-wide BRET signaling sensors. Sci Signal. 2018;11:559.

    Article  Google Scholar 

  10. Peng X, Yang L, Liu Z, Lou S, Mei S, Li M, et al. Structural basis for recognition of antihistamine drug by human histamine receptor. Nat Commun. 2022;13(1):6105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Höring C, Conrad M, Söldner CA, Wang J, Sticht H, Strasser A, et al. Specific Engineered G Protein Coupling to histamine receptors revealed from Cellular Assay experiments and accelerated Molecular Dynamics simulations. Int J Mol Sci. 2021;22:18.

    Article  Google Scholar 

  12. Avet C, Sturino C, Grastilleur S, Gouill CL, Semache M, Gross F, et al. The PAR2 inhibitor I-287 selectively targets Galphaq and Galpha12/13 signaling and has anti-inflammatory effects. Commun Biol. 2020;3(1):719.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Weis WI, Kobilka BK. The molecular basis of G protein-coupled receptor activation. Annu Rev Biochem. 2018;87:897–919.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Knight KM, Ghosh S, Campbell SL, Lefevre TJ, Olsen RHJ, Smrcka AV, et al. A universal allosteric mechanism for G protein activation. Mol Cell. 2021;81(7):1384–96. e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gilman AG. G proteins and dual control of adenylate cyclase. Cell. 1984;36(3):577–9.

    Article  CAS  PubMed  Google Scholar 

  16. Rhee SG. Regulation of phosphoinositide-specific phospholipase C. Annu Rev Biochem. 2001;70:281–312.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Strathmann MP, Simon MI. G alpha 12 and G alpha 13 subunits define a fourth class of G protein alpha subunits. Proc Natl Acad Sci U S A. 1991;88(13):5582–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Suzuki N, Nakamura S, Mano H, Kozasa T. Galpha 12 activates rho GTPase through tyrosine-phosphorylated leukemia-associated RhoGEF. Proc Natl Acad Sci U S A. 2003;100(2):733–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hart MJ, Jiang X, Kozasa T, Roscoe W, Singer WD, Gilman AG, et al. Direct stimulation of the guanine nucleotide exchange activity of p115 RhoGEF by Galpha13. Science. 1998;280(5372):2112–4.

    Article  CAS  PubMed  Google Scholar 

  20. Spiering D, Hodgson L. Dynamics of the rho-family small GTPases in actin regulation and motility. Cell Adh Migr. 2011;5(2):170–80.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Somlyo AP, Somlyo AV. Ca2 + sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev. 2003;83(4):1325–58.

    Article  CAS  PubMed  Google Scholar 

  22. Ross EM, Wilkie TM. GTPase-activating proteins for heterotrimeric G proteins: regulators of G protein signaling (RGS) and RGS-like proteins. Annu Rev Biochem. 2000;69:795–827.

    Article  CAS  PubMed  Google Scholar 

  23. Chiba Y, Misawa M. Increased expression of G12 and G13 proteins in bronchial smooth muscle of airway hyperresponsive rats. Inflamm Res. 2001;50(6):333–6.

    Article  CAS  PubMed  Google Scholar 

  24. Voyno-Yasenetskaya TA, Faure MP, Ahn NG, Bourne HR. Galpha12 and Galpha13 regulate extracellular signal-regulated kinase and c-Jun kinase pathways by different mechanisms in COS-7 cells. J Biol Chem. 1996;271(35):21081–7.

    Article  CAS  PubMed  Google Scholar 

  25. Feng X, Liu P, Zhou X, Li MT, Li FL, Wang Z, et al. Thromboxane A2 activates YAP/TAZ protein to induce vascular smooth muscle cell proliferation and Migration. J Biol Chem. 2016;291(36):18947–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang L, Wang D, Xing B, Tan YC, Huang J, Liu B, et al. G-Protein Galpha13 functions with Abl Kinase to regulate actin cytoskeletal reorganization. J Mol Biol. 2017;429(24):3836–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang Z, Tan X, Luo J, Cui B, Lei S, Si Z, et al. GNA13 promotes tumor growth and angiogenesis by upregulating CXC chemokines via the NF-kappaB signaling pathway in colorectal cancer cells. Cancer Med. 2018;7(11):5611–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hot B, Valnohova J, Arthofer E, Simon K, Shin J, Uhlen M, et al. FZD10-Galpha13 signalling axis points to a role of FZD10 in CNS angiogenesis. Cell Signal. 2017;32:93–103.

    Article  CAS  PubMed  Google Scholar 

  29. Chikumi H, Fukuhara S, Gutkind JS. Regulation of G protein-linked guanine nucleotide exchange factors for rho, PDZ-RhoGEF, and LARG by tyrosine phosphorylation: evidence of a role for focal adhesion kinase. J Biol Chem. 2002;277(14):12463–73.

    Article  CAS  PubMed  Google Scholar 

  30. Prakash YS. Airway smooth muscle in airway reactivity and remodeling: what have we learned? Am J Physiol Lung Cell Mol Physiol. 2013;305(12):L912–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Billington CK, Penn RB. m3 muscarinic acetylcholine receptor regulation in the airway. Am J Respir Cell Mol Biol. 2002;26(3):269–72.

    Article  CAS  PubMed  Google Scholar 

  32. Billington CK, Penn RB. Signaling and regulation of G protein-coupled receptors in airway smooth muscle. Respir Res. 2003;4(1):2.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Winder SJ, Walsh MP. Smooth muscle calponin. Inhibition of actomyosin MgATPase and regulation by phosphorylation. J Biol Chem. 1990;265(17):10148–55.

    Article  CAS  PubMed  Google Scholar 

  34. Bradley SJ, Wiegman CH, Iglesias MM, Kong KC, Butcher AJ, Plouffe B, et al. Mapping physiological G protein-coupled receptor signaling pathways reveals a role for receptor phosphorylation in airway contraction. Proc Natl Acad Sci U S A. 2016;113(16):4524–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Carr R 3rd, Koziol-White C, Zhang J, Lam H, An SS, Tall GG, et al. Interdicting Gq activation in Airway Disease by receptor-dependent and receptor-independent mechanisms. Mol Pharmacol. 2016;89(1):94–104.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cipolla MJ, Gokina NI, Osol G. Pressure-induced actin polymerization in vascular smooth muscle as a mechanism underlying myogenic behavior. FASEB J. 2002;16(1):72–6.

    Article  CAS  PubMed  Google Scholar 

  37. Zhang W, Du L, Gunst SJ. The effects of the small GTPase RhoA on the muscarinic contraction of airway smooth muscle result from its role in regulating actin polymerization. Am J Physiol Cell Physiol. 2010;299(2):C298–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. An SS, Laudadio RE, Lai J, Rogers RA, Fredberg JJ. Stiffness changes in cultured airway smooth muscle cells. Am J Physiol Cell Physiol. 2002;283(3):C792–801.

    Article  CAS  PubMed  Google Scholar 

  39. Nobes CD, Hall A. Rho, rac, and cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell. 1995;81(1):53–62.

    Article  CAS  PubMed  Google Scholar 

  40. Hirshman CA, Emala CW. Actin reorganization in airway smooth muscle cells involves Gq and Gi-2 activation of rho. Am J Physiol. 1999;277(3):L653–61.

    CAS  PubMed  Google Scholar 

  41. Fujii T, Namba K. Structure of actomyosin rigour complex at 5.2 Å resolution and insights into the ATPase cycle mechanism. Nat Commun. 2017;8:13969.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gohla A, Harhammer R, Schultz G. The G-protein G13 but not G12 mediates signaling from lysophosphatidic acid receptor via epidermal growth factor receptor to rho. J Biol Chem. 1998;273(8):4653–9.

    Article  CAS  PubMed  Google Scholar 

  43. Kitazawa T, Masuo M, Somlyo AP. G protein-mediated inhibition of myosin light-chain phosphatase in vascular smooth muscle. Proc Natl Acad Sci U S A. 1991;88(20):9307–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kitazawa T, Eto M, Woodsome TP, Brautigan DL. Agonists trigger G protein-mediated activation of the CPI-17 inhibitor phosphoprotein of myosin light chain phosphatase to enhance vascular smooth muscle contractility. J Biol Chem. 2000;275(14):9897–900.

    Article  CAS  PubMed  Google Scholar 

  45. Maekawa M, Ishizaki T, Boku S, Watanabe N, Fujita A, Iwamatsu A, et al. Signaling from rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science. 1999;285(5429):895–8.

    Article  CAS  PubMed  Google Scholar 

  46. Agnew BJ, Minamide LS, Bamburg JR. Reactivation of phosphorylated actin depolymerizing factor and identification of the regulatory site. J Biol Chem. 1995;270(29):17582–7.

    Article  CAS  PubMed  Google Scholar 

  47. Carlier MF, Laurent V, Santolini J, Melki R, Didry D, Xia GX, et al. Actin depolymerizing factor (ADF/cofilin) enhances the rate of filament turnover: implication in actin-based motility. J Cell Biol. 1997;136(6):1307–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kiuchi T, Ohashi K, Kurita S, Mizuno K. Cofilin promotes stimulus-induced lamellipodium formation by generating an abundant supply of actin monomers. J Cell Biol. 2007;177(3):465–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lan B, Wang L, Zhang J, Pascoe CD, Norris BA, Liu JC, et al. Rho-kinase mediated cytoskeletal stiffness in skinned smooth muscle. J Appl Physiol (1985). 2013;115(10):1540–52.

    Article  CAS  PubMed  Google Scholar 

  50. Iizuka K, Shimizu Y, Tsukagoshi H, Yoshii A, Harada T, Dobashi K, et al. Evaluation of Y-27632, a rho-kinase inhibitor, as a bronchodilator in guinea pigs. Eur J Pharmacol. 2000;406(2):273–9.

    Article  CAS  PubMed  Google Scholar 

  51. Iizuka K, Yoshii A, Samizo K, Tsukagoshi H, Ishizuka T, Dobashi K, et al. A major role for the rho-associated coiled coil forming protein kinase in G-protein-mediated Ca2 + sensitization through inhibition of myosin phosphatase in rabbit trachea. Br J Pharmacol. 1999;128(4):925–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Yoshii A, Iizuka K, Dobashi K, Horie T, Harada T, Nakazawa T, et al. Relaxation of contracted rabbit tracheal and human bronchial smooth muscle by Y-27632 through inhibition of Ca2 + sensitization. Am J Respir Cell Mol Biol. 1999;20(6):1190–200.

    Article  CAS  PubMed  Google Scholar 

  53. Fukumoto Y, Kaibuchi K, Hori Y, Fujioka H, Araki S, Ueda T, et al. Molecular cloning and characterization of a novel type of regulatory protein (GDI) for the rho proteins, ras p21-like small GTP-binding proteins. Oncogene. 1990;5(9):1321–8.

    CAS  PubMed  Google Scholar 

  54. Ueda T, Kikuchi A, Ohga N, Yamamoto J, Takai Y. Purification and characterization from bovine brain cytosol of a novel regulatory protein inhibiting the dissociation of GDP from and the subsequent binding of GTP to rhoB p20, a ras p21-like GTP-binding protein. J Biol Chem. 1990;265(16):9373–80.

    Article  CAS  PubMed  Google Scholar 

  55. Gohla A, Schultz G, Offermanns S. Role for G(12)/G(13) in agonist-induced vascular smooth muscle cell contraction. Circ Res. 2000;87(3):221–7.

    Article  CAS  PubMed  Google Scholar 

  56. Wirth A, Wang S, Takefuji M, Tang C, Althoff TF, Schweda F, et al. Age-dependent blood pressure elevation is due to increased vascular smooth muscle tone mediated by G-protein signalling. Cardiovasc Res. 2016;109(1):131–40.

    Article  CAS  PubMed  Google Scholar 

  57. Wirth A, Benyo Z, Lukasova M, Leutgeb B, Wettschureck N, Gorbey S, et al. G12-G13-LARG-mediated signaling in vascular smooth muscle is required for salt-induced hypertension. Nat Med. 2008;14(1):64–8.

    Article  CAS  PubMed  Google Scholar 

  58. Chennupati R, Wirth A, Favre J, Li R, Bonnavion R, Jin YJ et al. Myogenic vasoconstriction requires G12/G13 and LARG to maintain local and systemic vascular resistance. Elife. 2019;8.

  59. Gao J, Denys I, Shahien A, Sutphen J, Kapusta DR. Downregulation of Brain Galpha12 attenuates angiotensin II-Dependent hypertension. Am J Hypertens. 2020;33(2):198–204.

    Article  CAS  PubMed  Google Scholar 

  60. Rumenapp U, Asmus M, Schablowski H, Woznicki M, Han L, Jakobs KH, et al. The M3 muscarinic acetylcholine receptor expressed in HEK-293 cells signals to phospholipase D via G12 but not Gq-type G proteins: regulators of G proteins as tools to dissect pertussis toxin-resistant G proteins in receptor-effector coupling. J Biol Chem. 2001;276(4):2474–9.

    Article  CAS  PubMed  Google Scholar 

  61. Yuan J, Slice LW, Rozengurt E. Activation of protein kinase D by signaling through rho and the alpha subunit of the heterotrimeric G protein G13. J Biol Chem. 2001;276(42):38619–27.

    Article  CAS  PubMed  Google Scholar 

  62. Lee SJ, Lee WH, Ki SH, Kim YM, Lee SJ, Lee CH, et al. Galpha13 regulates methacholine-induced contraction of bronchial smooth muscle via phosphorylation of MLC20. Biochem Pharmacol. 2009;77(9):1497–505.

    Article  CAS  PubMed  Google Scholar 

  63. Yoo EJ, Cao G, Koziol-White CJ, Ojiaku CA, Sunder K, Jude JA, et al. Galpha12 facilitates shortening in human airway smooth muscle by modulating phosphoinositide 3-kinase-mediated activation in a RhoA-dependent manner. Br J Pharmacol. 2017;174(23):4383–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ojiaku CA, Cao G, Zhu W, Yoo EJ, Shumyatcher M, Himes BE, et al. TGF-beta1 evokes human airway smooth muscle cell shortening and hyperresponsiveness via Smad3. Am J Respir Cell Mol Biol. 2018;58(5):575–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Shaifta Y, MacKay CE, Irechukwu N, O’Brien KA, Wright DB, Ward JPT, et al. Transforming growth factor-β enhances rho-kinase activity and contraction in airway smooth muscle via the nucleotide exchange factor ARHGEF1. J Physiol. 2018;596(1):47–66.

    Article  CAS  PubMed  Google Scholar 

  66. Hammad H, Lambrecht BN. The basic immunology of asthma. Cell. 2021;184(9):2521–2.

    Article  CAS  PubMed  Google Scholar 

  67. Wenzel SE. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med. 2012;18(5):716–25.

    Article  CAS  PubMed  Google Scholar 

  68. Deo SS, Mistry KJ, Kakade AM, Niphadkar PV. Role played by Th2 type cytokines in IgE mediated allergy and asthma. Lung India. 2010;27(2):66–71.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Whitsett JA, Alenghat T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat Immunol. 2015;16(1):27–35.

    Article  CAS  PubMed  Google Scholar 

  70. Idzko M, Panther E, Corinti S, Morelli A, Ferrari D, Herouy Y, et al. Sphingosine 1-phosphate induces chemotaxis of immature and modulates cytokine-release in mature human dendritic cells for emergence of Th2 immune responses. FASEB J. 2002;16(6):625–7.

    Article  CAS  PubMed  Google Scholar 

  71. Chen H, Chen K, Huang W, Staudt LM, Cyster JG, Li X. Structure of S1PR2-heterotrimeric G13 signaling complex. Sci Adv. 2022;8(13):eabn0067.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. McGeachie MJ, Dahlin A, Qiu W, Croteau-Chonka DC, Savage J, Wu AC, et al. The metabolomics of asthma control: a promising link between genetics and disease. Immun Inflamm Dis. 2015;3(3):224–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Dusaban SS, Purcell NH, Rockenstein E, Masliah E, Cho MK, Smrcka AV, et al. Phospholipase C epsilon links G protein-coupled receptor activation to inflammatory astrocytic responses. Proc Natl Acad Sci U S A. 2013;110(9):3609–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kuen DS, Park M, Ryu H, Choi G, Moon YH, Kim JO, et al. Critical regulation of follicular helper T cell differentiation and function by Galpha13 signaling. Proc Natl Acad Sci U S A. 2021;118:43.

    Article  Google Scholar 

  75. Grimm M, Tischner D, Troidl K, Albarran Juarez J, Sivaraj KK, Ferreiros Bouzas N, et al. S1P2/G12/13 signaling negatively regulates macrophage activation and indirectly shapes the atheroprotective B1-Cell Population. Arterioscler Thromb Vasc Biol. 2016;36(1):37–48.

    Article  CAS  PubMed  Google Scholar 

  76. Yu H, Liu Z. GNA12 regulates C5a-induced migration by downregulating C5aR1-PLCbeta2-PI3K-AKT-ERK1/2 signaling. Biophys Rep. 2023;9(1):33–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. van den Bos E, Ambrosy B, Horsthemke M, Walbaum S, Bachg AC, Wettschureck N, et al. Knockout mouse models reveal the contributions of G protein subunits to complement C5a receptor-mediated chemotaxis. J Biol Chem. 2020;295(22):7726–42.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Mikelis CM, Palmby TR, Simaan M, Li W, Szabo R, Lyons R, et al. PDZ-RhoGEF and LARG are essential for embryonic development and provide a link between thrombin and LPA receptors and rho activation. J Biol Chem. 2013;288(17):12232–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Bouafia A, Lofek S, Bruneau J, Chentout L, Lamrini H, Trinquand A, et al. Loss of ARHGEF1 causes a human primary antibody deficiency. J Clin Invest. 2019;129(3):1047–60.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Bettegazzi B, Bellani S, Cattaneo S, Codazzi F, Grohovaz F, Zacchetti D. Galpha13 contributes to LPS-Induced morphological alterations and affects Migration of Microglia. Mol Neurobiol. 2021;58(12):6397–414.

    Article  CAS  PubMed  Google Scholar 

  81. Zhang Y, Zhao X, Shen B, Bai Y, Chang C, Stojanovic A, et al. Integrin beta(3) directly inhibits the Galpha(13)-p115RhoGEF interaction to regulate G protein signaling and platelet exocytosis. Nat Commun. 2023;14(1):4966.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Kaiser R, Anjum A, Kammerer L, Loew Q, Akhalkatsi A, Rossaro D, et al. Mechanosensing via a GpIIb/Src/14-3-3zeta axis critically regulates platelet migration in vascular inflammation. Blood. 2023;141(24):2973–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Cheng N, Zhang Y, Delaney MK, Wang C, Bai Y, Skidgel RA, et al. Targeting Galpha13-integrin interaction ameliorates systemic inflammation. Nat Commun. 2021;12(1):3185.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Wang L, Luo JY, Li B, Tian XY, Chen LJ, Huang Y, et al. Integrin-YAP/TAZ-JNK cascade mediates atheroprotective effect of unidirectional shear flow. Nature. 2016;540(7634):579–82.

    Article  CAS  PubMed  Google Scholar 

  85. Lim WK, Chai X, Ghosh S, Ray D, Wang M, Rasheed SAK, et al. Galpha-13 induces CXC motif chemokine ligand 5 expression in prostate cancer cells by transactivating NF-kappaB. J Biol Chem. 2019;294(48):18192–206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Wang Z, Guan W, Han Y, Ren H, Tang X, Zhang H, et al. Stimulation of dopamine D3 receptor attenuates renal ischemia-reperfusion Injury via increased linkage with Galpha12. Transplantation. 2015;99(11):2274–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Yu W, Beaudry S, Negoro H, Boucher I, Tran M, Kong T, et al. H2O2 activates G protein, alpha 12 to disrupt the junctional complex and enhance ischemia reperfusion injury. Proc Natl Acad Sci U S A. 2012;109(17):6680–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Yanamadala V, Negoro H, Gunaratnam L, Kong T, Denker BM. Galpha12 stimulates apoptosis in epithelial cells through JNK1-mediated Bcl-2 degradation and up-regulation of IkappaBalpha. J Biol Chem. 2007;282(33):24352–63.

    Article  CAS  PubMed  Google Scholar 

  89. Hough KP, Curtiss ML, Blain TJ, Liu RM, Trevor J, Deshane JS, et al. Airway Remodeling in Asthma. Front Med (Lausanne). 2020;7:191.

    Article  PubMed  Google Scholar 

  90. Ebina M, Takahashi T, Chiba T, Motomiya M. Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis. 1993;148(3):720–6.

    Article  CAS  PubMed  Google Scholar 

  91. Johnson PR, Roth M, Tamm M, Hughes M, Ge Q, King G, et al. Airway smooth muscle cell proliferation is increased in asthma. Am J Respir Crit Care Med. 2001;164(3):474–7.

    Article  CAS  PubMed  Google Scholar 

  92. Cohen L, E X, Tarsi J, Ramkumar T, Horiuchi TK, Cochran R, et al. Epithelial cell proliferation contributes to airway remodeling in severe asthma. Am J Respir Crit Care Med. 2007;176(2):138–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Hernandez-Lara MA, Yadav SK, Shah SD, Okumura M, Yokoyama Y, Penn RB, et al. Regulation of Airway smooth muscle cell proliferation by Diacylglycerol Kinase: relevance to Airway Remodeling in Asthma. Int J Mol Sci. 2022;23:19.

    Article  Google Scholar 

  94. Chan AM, Fleming TP, McGovern ES, Chedid M, Miki T, Aaronson SA. Expression cDNA cloning of a transforming gene encoding the wild-type G alpha 12 gene product. Mol Cell Biol. 1993;13(2):762–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Xu N, Bradley L, Ambdukar I, Gutkind JS. A mutant alpha subunit of G12 potentiates the eicosanoid pathway and is highly oncogenic in NIH 3T3 cells. Proc Natl Acad Sci U S A. 1993;90(14):6741–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Voyno-Yasenetskaya TA, Pace AM, Bourne HR. Mutant alpha subunits of G12 and G13 proteins induce neoplastic transformation of Rat-1 fibroblasts. Oncogene. 1994;9(9):2559–65.

    CAS  PubMed  Google Scholar 

  97. Xu N, Voyno-Yasenetskaya T, Gutkind JS. Potent transforming activity of the G13 alpha subunit defines a novel family of oncogenes. Biochem Biophys Res Commun. 1994;201(2):603–9.

    Article  CAS  PubMed  Google Scholar 

  98. Rasheed SAK, Subramanyan LV, Lim WK, Udayappan UK, Wang M, Casey PJ. The emerging roles of Galpha12/13 proteins on the hallmarks of cancer in solid tumors. Oncogene. 2022;41(2):147–58.

    Article  CAS  PubMed  Google Scholar 

  99. Takefuji M, Kruger M, Sivaraj KK, Kaibuchi K, Offermanns S, Wettschureck N. RhoGEF12 controls cardiac remodeling by integrating G protein- and integrin-dependent signaling cascades. J Exp Med. 2013;210(4):665–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Sivaraj KK, Takefuji M, Schmidt I, Adams RH, Offermanns S, Wettschureck N. G13 controls angiogenesis through regulation of VEGFR-2 expression. Dev Cell. 2013;25(4):427–34.

    Article  CAS  PubMed  Google Scholar 

  101. Takefuji M, Wirth A, Lukasova M, Takefuji S, Boettger T, Braun T, et al. G(13)-mediated signaling pathway is required for pressure overload-induced cardiac remodeling and heart failure. Circulation. 2012;126(16):1972–82.

    Article  PubMed  Google Scholar 

  102. Huang J, Qu Q, Dai Y, Ren D, Qian J, Ge J. Detrimental role of PDZ-RhoGEF in Pathological Cardiac Hypertrophy. Hypertension. 2023;80(2):403–15.

    Article  CAS  PubMed  Google Scholar 

  103. Shan D, Chen L, Wang D, Tan YC, Gu JL, Huang XY. The G protein G alpha(13) is required for growth factor-induced cell migration. Dev Cell. 2006;10(6):707–18.

    Article  CAS  PubMed  Google Scholar 

  104. Kim KM, Han CY, Kim JY, Cho SS, Kim YS, Koo JH, et al. Galpha12 overexpression induced by miR-16 dysregulation contributes to liver fibrosis by promoting autophagy in hepatic stellate cells. J Hepatol. 2018;68(3):493–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Martin JW, Cavagnini KS, Brawley DN, Berkley CY, Smolski WC, Garcia RD, et al. A Galpha12-specific binding domain in AKAP-Lbc and p114RhoGEF. J Mol Signal. 2016;11:3.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Gan X, Wang J, Wang C, Sommer E, Kozasa T, Srinivasula S, et al. PRR5L degradation promotes mTORC2-mediated PKC-delta phosphorylation and cell migration downstream of Galpha12. Nat Cell Biol. 2012;14(7):686–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Juneja J, Cushman I, Casey PJ. G12 signaling through c-Jun NH2-terminal kinase promotes breast cancer cell invasion. PLoS ONE. 2011;6(11):e26085.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Fromm C, Coso OA, Montaner S, Xu N, Gutkind JS. The small GTP-binding protein rho links G protein-coupled receptors and Galpha12 to the serum response element and to cellular transformation. Proc Natl Acad Sci U S A. 1997;94(19):10098–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Medlin MD, Staus DP, Dubash AD, Taylor JM, Mack CP. Sphingosine 1-phosphate receptor 2 signals through leukemia-associated RhoGEF (LARG), to promote smooth muscle cell differentiation. Arterioscler Thromb Vasc Biol. 2010;30(9):1779–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Yu OM, Benitez JA, Plouffe SW, Ryback D, Klein A, Smith J, et al. YAP and MRTF-A, transcriptional co-activators of RhoA-mediated gene expression, are critical for glioblastoma tumorigenicity. Oncogene. 2018;37(41):5492–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Yu OM, Miyamoto S, Brown JH. Myocardin-related transcription factor A and Yes-Associated protein exert Dual Control in G protein-coupled receptor- and RhoA-Mediated transcriptional regulation and cell proliferation. Mol Cell Biol. 2016;36(1):39–49.

    Article  CAS  PubMed  Google Scholar 

  112. Marinissen MJ, Chiariello M, Tanos T, Bernard O, Narumiya S, Gutkind JS. The small GTP-binding protein RhoA regulates c-jun by a ROCK-JNK signaling axis. Mol Cell. 2004;14(1):29–41.

    Article  CAS  PubMed  Google Scholar 

  113. Jung HS, Seo YR, Yang YM, Koo JH, An J, Lee SJ, et al. Galpha12gep oncogene inhibits FOXO1 in hepatocellular carcinoma as a consequence of miR-135b and miR-194 dysregulation. Cell Signal. 2014;26(7):1456–65.

    Article  CAS  PubMed  Google Scholar 

  114. Yang YM, Lee WH, Lee CG, An J, Kim ES, Kim SH, et al. Galpha12 gep oncogene deregulation of p53-responsive microRNAs promotes epithelial-mesenchymal transition of hepatocellular carcinoma. Oncogene. 2015;34(22):2910–21.

    Article  CAS  PubMed  Google Scholar 

  115. Yuan B, Cui J, Wang W, Deng K. Galpha12/13 signaling promotes cervical cancer invasion through the RhoA/ROCK-JNK signaling axis. Biochem Biophys Res Commun. 2016;473(4):1240–6.

    Article  CAS  PubMed  Google Scholar 

  116. Lockman K, Hinson JS, Medlin MD, Morris D, Taylor JM, Mack CP. Sphingosine 1-phosphate stimulates smooth muscle cell differentiation and proliferation by activating separate serum response factor co-factors. J Biol Chem. 2004;279(41):42422–30.

    Article  CAS  PubMed  Google Scholar 

  117. Yu FX, Zhao B, Panupinthu N, Jewell JL, Lian I, Wang LH, et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell. 2012;150(4):780–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Mo JS, Yu FX, Gong R, Brown JH, Guan KL. Regulation of the Hippo-YAP pathway by protease-activated receptors (PARs). Genes Dev. 2012;26(19):2138–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. He H, Sugiyama A, Snyder NW, Teneche MG, Liu X, Maner-Smith KM, et al. Acyl-CoA thioesterase 12 suppresses YAP-mediated hepatocarcinogenesis by limiting glycerolipid biosynthesis. Cancer Lett. 2023;565:216210.

    Article  CAS  PubMed  Google Scholar 

  120. Regue L, Mou F, Avruch J. G protein-coupled receptors engage the mammalian Hippo pathway through F-actin: F-Actin, assembled in response to Galpha12/13 induced RhoA-GTP, promotes dephosphorylation and activation of the YAP oncogene. BioEssays. 2013;35(5):430–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Gong H, Shen B, Flevaris P, Chow C, Lam SC, Voyno-Yasenetskaya TA, et al. G protein subunit Galpha13 binds to integrin alphaIIbbeta3 and mediates integrin outside-in signaling. Science. 2010;327(5963):340–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Bian D, Mahanivong C, Yu J, Frisch SM, Pan ZK, Ye RD, et al. The G12/13-RhoA signaling pathway contributes to efficient lysophosphatidic acid-stimulated cell migration. Oncogene. 2006;25(15):2234–44.

    Article  CAS  PubMed  Google Scholar 

  123. Justus CR, Yang LV. GPR4 decreases B16F10 melanoma cell spreading and regulates focal adhesion dynamics through the G13/Rho signaling pathway. Exp Cell Res. 2015;334(1):100–13.

    Article  CAS  PubMed  Google Scholar 

  124. Masia-Balague M, Izquierdo I, Garrido G, Cordomi A, Perez-Benito L, Miller NL, et al. Gastrin-stimulated Galpha13 activation of rgnef protein (ArhGEF28) in DLD-1 Colon carcinoma cells. J Biol Chem. 2015;290(24):15197–209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Anfinogenova Y, Wang R, Li QF, Spinelli AM, Tang DD. Abl silencing inhibits CAS-mediated process and constriction in resistance arteries. Circ Res. 2007;101(4):420–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Jia L, Tang DD. Abl activation regulates the dissociation of CAS from cytoskeletal vimentin by modulating CAS phosphorylation in smooth muscle. Am J Physiol Cell Physiol. 2010;299(3):C630–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Jia L, Wang R, Tang DD. Abl regulates smooth muscle cell proliferation by modulating actin dynamics and ERK1/2 activation. Am J Physiol Cell Physiol. 2012;302(7):C1026–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Cohen SG. Asthma in antiquity: the Ebers Papyrus. Allergy Proc. 1992;13(3):147–54.

    Article  CAS  PubMed  Google Scholar 

  129. Jackson M. Divine stramonium: the rise and fall of smoking for asthma. Med Hist. 2010;54(2):171–94.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Mein HFG. Ueber die Darstellung Des Atropins in Weissen Kristallen. Annalen Der Pharmacie. 1833;6(1):67–72.

    Article  Google Scholar 

  131. Diamant Z, Boot JD, Virchow JC. Summing up 100 years of asthma. Respir Med. 2007;101(3):378–88.

    Article  PubMed  Google Scholar 

  132. Suen JY, Cotterell A, Lohman RJ, Lim J, Han A, Yau MK, et al. Pathway-selective antagonism of proteinase activated receptor 2. Br J Pharmacol. 2014;171(17):4112–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Hollenberg MD, Mihara K, Polley D, Suen JY, Han A, Fairlie DP, et al. Biased signalling and proteinase-activated receptors (PARs): targeting inflammatory disease. Br J Pharmacol. 2014;171(5):1180–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Sedki D, Cho A, Cao Y, Nikolajev L, Atmuri NDP, Lubell WD, et al. Prostaglandin F2alpha and angiotensin II type 1 receptors exhibit differential cognate G protein coupling regulation. J Biol Chem. 2022;298(9):102294.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Tran K, Sainsily X, Cote J, Coquerel D, Couvineau P, Saibi S, et al. Size-reduced macrocyclic analogues of [Pyr(1)]-apelin-13 showing negative Galpha12 Bias still produce prolonged Cardiac effects. J Med Chem. 2022;65(1):531–51.

    Article  CAS  PubMed  Google Scholar 

  136. Expert Panel Working Group of the National Heart, Prevention Program Coordinating L, Cloutier C, Baptist MM et al. AP,. 2020 Focused Updates to the Asthma Management Guidelines: A Report from the National Asthma Education and Prevention Program Coordinating Committee Expert Panel Working Group. J Allergy Clin Immunol. 2020;146(6):1217-70.

  137. Chen R, Suchard MA, Krumholz HM, Schuemie MJ, Shea S, Duke J, et al. Comparative first-line effectiveness and safety of ACE (angiotensin-Converting enzyme) inhibitors and angiotensin receptor blockers: a multinational cohort study. Hypertension. 2021;78(3):591–603.

    Article  CAS  PubMed  Google Scholar 

  138. Pang R, Zhou XY, Wang X, Wang B, Yin XL, Bo H, et al. Anticholinergics combined with alpha-blockers for treating lower urinary tract symptoms related to benign prostatic obstruction. Cochrane Database Syst Rev. 2021;2(2):CD012336.

    PubMed  Google Scholar 

  139. Stoniute A, Madhuvrata P, Still M, Barron-Millar E, Nabi G, Omar MI. Oral anticholinergic drugs versus placebo or no treatment for managing overactive bladder syndrome in adults. Cochrane Database Syst Rev. 2023;5(5):CD003781.

    PubMed  Google Scholar 

  140. Schmitt JM, Stork PJ. Cyclic AMP-mediated inhibition of cell growth requires the small G protein Rap1. Mol Cell Biol. 2001;21(11):3671–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Nguyen LP, Al-Sawalha NA, Parra S, Pokkunuri I, Omoluabi O, Okulate AA, et al. beta(2)-Adrenoceptor signaling in airway epithelial cells promotes eosinophilic inflammation, mucous metaplasia, and airway contractility. Proc Natl Acad Sci U S A. 2017;114(43):E9163–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Nelson HS, Weiss ST, Bleecker ER, Yancey SW, Dorinsky PM, Group SS. The Salmeterol Multicenter Asthma Research Trial: a comparison of usual pharmacotherapy for asthma or usual pharmacotherapy plus salmeterol. Chest. 2006;129(1):15–26.

    Article  CAS  PubMed  Google Scholar 

  143. Salpeter SR, Buckley NS, Ormiston TM, Salpeter EE. Meta-analysis: effect of long-acting beta-agonists on severe asthma exacerbations and asthma-related deaths. Ann Intern Med. 2006;144(12):904–12.

    Article  CAS  PubMed  Google Scholar 

  144. Yan D, Hamed O, Joshi T, Mostafa MM, Jamieson KC, Joshi R, et al. Analysis of the Indacaterol-regulated transcriptome in human airway epithelial cells implicates Gene expression changes in the adverse and therapeutic effects of beta(2)-Adrenoceptor agonists. J Pharmacol Exp Ther. 2018;366(1):220–36.

    Article  CAS  PubMed  Google Scholar 

  145. Sharafkhaneh A, Majid H, Gross NJ. Safety and tolerability of inhalational anticholinergics in COPD. Drug Healthc Patient Saf. 2013;5:49–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Westby M, Benson M, Gibson P. Anticholinergic agents for chronic asthma in adults. Cochrane Database Syst Rev. 2004;2004(3):CD003269.

    PubMed  PubMed Central  Google Scholar 

  147. Novelli F, Malagrino L, Dente FL, Paggiaro P. Efficacy of anticholinergic drugs in asthma. Expert Rev Respir Med. 2012;6(3):309–19.

    Article  CAS  PubMed  Google Scholar 

  148. Vaughan M, Moss J. Mechanism of action of choleragen. J Supramol Struct. 1978;8(4):473–88.

    Article  CAS  PubMed  Google Scholar 

  149. Shah BH. Enhanced degradation of stimulatory G-protein (gs alpha) by cholera toxin is mediated by ADP-ribosylation of Gs alpha protein but not by increased cyclic AMP levels. Adv Exp Med Biol. 1997;419:93–7.

    Article  CAS  PubMed  Google Scholar 

  150. Murayama T, Ui M. Loss of the inhibitory function of the guanine nucleotide regulatory component of adenylate cyclase due to its ADP ribosylation by islet-activating protein, pertussis toxin, in adipocyte membranes. J Biol Chem. 1983;258(5):3319–26.

    Article  CAS  PubMed  Google Scholar 

  151. Takasaki J, Saito T, Taniguchi M, Kawasaki T, Moritani Y, Hayashi K, et al. A novel Galphaq/11-selective inhibitor. J Biol Chem. 2004;279(46):47438–45.

    Article  CAS  PubMed  Google Scholar 

  152. Schrage R, Schmitz AL, Gaffal E, Annala S, Kehraus S, Wenzel D, et al. The experimental power of FR900359 to study Gq-regulated biological processes. Nat Commun. 2015;6:10156.

    Article  CAS  PubMed  Google Scholar 

  153. Gu JL, Muller S, Mancino V, Offermanns S, Simon MI. Interaction of G alpha(12) with G alpha(13) and G alpha(q) signaling pathways. Proc Natl Acad Sci U S A. 2002;99(14):9352–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Wang ZZ, Shi XX, Huang GY, Hao GF, Yang GF. Fragment-based drug discovery supports drugging ‘undruggable’ protein-protein interactions. Trends Biochem Sci. 2023;48(6):539–52.

    Article  CAS  PubMed  Google Scholar 

  155. Berg T. Small-molecule inhibitors of protein-protein interactions. Curr Opin Drug Discov Devel. 2008;11(5):666–74.

    CAS  PubMed  Google Scholar 

  156. Basse MJ, Betzi S, Bourgeas R, Bouzidi S, Chetrit B, Hamon V, et al. 2P2Idb: a structural database dedicated to orthosteric modulation of protein-protein interactions. Nucleic Acids Res. 2013;41(Database issue):D824–7.

    CAS  PubMed  Google Scholar 

  157. Konstantinidou M, Visser EJ, Vandenboorn E, Chen S, Jaishankar P, Overmans M, et al. Structure-based optimization of Covalent, Small-Molecule stabilizers of the 14-3-3σ/ERα protein-protein Interaction from nonselective fragments. J Am Chem Soc. 2023;145(37):20328–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. DiGiacomo V, de Opakua AI, Papakonstantinou MP, Nguyen LT, Merino N, Blanco-Canosa JB, et al. The Gαi-GIV binding interface is a druggable protein-protein interaction. Sci Rep. 2017;7(1):8575.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Pang A, Cheng N, Cui Y, Bai Y, Hong Z, Delaney MK et al. High-loading Galpha13-binding EXE peptide nanoparticles prevent thrombosis and protect mice from cardiac ischemia/reperfusion injury. Sci Transl Med. 2020;12(552).

  160. Quilliam LA, Lacal JC, Bokoch GM. Identification of rho as a substrate for botulinum toxin C3-catalyzed ADP-ribosylation. FEBS Lett. 1989;247(2):221–6.

    Article  CAS  PubMed  Google Scholar 

  161. Shang X, Marchioni F, Evelyn CR, Sipes N, Zhou X, Seibel W, et al. Small-molecule inhibitors targeting G-protein-coupled rho guanine nucleotide exchange factors. Proc Natl Acad Sci U S A. 2013;110(8):3155–60.

    Article  CAS  PubMed Central  Google Scholar 

  162. Pagano L, Lee JW, Posarelli M, Giannaccare G, Kaye S, Borgia A. ROCK inhibitors in corneal diseases and Glaucoma-A Comprehensive Review of these emerging drugs. J Clin Med. 2023;12(21).

  163. Ali F, Ilyas A. Belumosudil with ROCK-2 inhibition: chemical and therapeutic development to FDA approval for the treatment of chronic graft-versus-host disease. Curr Res Transl Med. 2022;70(3):103343.

    CAS  PubMed  Google Scholar 

  164. Lingor P, Weber M, Camu W, Friede T, Hilgers R, Leha A, et al. ROCK-ALS: protocol for a Randomized, Placebo-Controlled, double-blind phase IIa trial of Safety, Tolerability and Efficacy of the rho kinase (ROCK) inhibitor Fasudil in Amyotrophic lateral sclerosis. Front Neurol. 2019;10:293.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Wolff AW, Bidner H, Remane Y, Zimmer J, Aarsland D, Rascol O, et al. Protocol for a randomized, placebo-controlled, double-blind phase IIa study of the safety, tolerability, and symptomatic efficacy of the ROCK-inhibitor Fasudil in patients with Parkinson’s disease (ROCK-PD). Front Aging Neurosci. 2024;16:1308577.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. McLeod R, Kumar R, Papadatos-Pastos D, Mateo J, Brown JS, Garces AHI, et al. First-in-human study of AT13148, a dual ROCK-AKT inhibitor in patients with solid tumors. Clin Cancer Res. 2020;26(18):4777–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Zanin-Zhorov A, Weiss JM, Trzeciak A, Chen W, Zhang J, Nyuydzefe MS, et al. Cutting Edge: selective oral ROCK2 inhibitor reduces clinical scores in patients with Psoriasis Vulgaris and normalizes skin Pathology via Concurrent Regulation of IL-17 and IL-10. J Immunol. 2017;198(10):3809–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Matoba K, Takeda Y, Nagai Y, Sekiguchi K, Yokota T, Utsunomiya K, et al. The Physiology, Pathology, and therapeutic interventions for ROCK isoforms in Diabetic kidney disease. Front Pharmacol. 2020;11:585633.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Montagnoli TL, da Silva JS, Sudo SZ, Santos AD, Gomide GF, de Sa MPL et al. ROCK inhibition as potential target for treatment of Pulmonary Hypertension. Cells. 2021;10(7).

  170. Barcelo J, Samain R, Sanz-Moreno V. Preclinical to clinical utility of ROCK inhibitors in cancer. Trends Cancer. 2023;9(3):250–63.

    Article  CAS  PubMed  Google Scholar 

  171. Gupta V, Gupta N, Shaik IH, Mehvar R, McMurtry IF, Oka M, et al. Liposomal fasudil, a rho-kinase inhibitor, for prolonged pulmonary preferential vasodilation in pulmonary arterial hypertension. J Control Release. 2013;167(2):189–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Franova S, Molitorisova M, Kalmanova L, Palencarova J, Joskova M, Smiesko L, et al. The anti-asthmatic potential of rho-kinase inhibitor hydroxyfasudil in the model of experimentally induced allergic airway inflammation. Eur J Pharmacol. 2023;938:175450.

    Article  CAS  PubMed  Google Scholar 

  173. Zhang W, Li X, Zhang Y. Rho-kinase inhibitor attenuates airway mucus hypersecretion and inflammation partly by downregulation of IL-13 and the JNK1/2-AP1 signaling pathway. Biochem Biophys Res Commun. 2019;516(2):571–7.

    Article  CAS  PubMed  Google Scholar 

  174. Wang L, Chitano P, Pare PD, Seow CY. Mechanopharmacology and Synergistic Relaxation of Airway Smooth Muscle. J Eng Sci Med Diagn Ther. 2019;2(1):0110041–110047.

    PubMed  PubMed Central  Google Scholar 

  175. Duan Y, Long J, Chen J, Jiang X, Zhu J, Jin Y, et al. Overexpression of soluble ADAM33 promotes a hypercontractile phenotype of the airway smooth muscle cell in rat. Exp Cell Res. 2016;349(1):109–18.

    Article  CAS  PubMed  Google Scholar 

  176. Duan W, Aguinaldo Datiles AM, Leung BP, Vlahos CJ, Wong WS. An anti-inflammatory role for a phosphoinositide 3-kinase inhibitor LY294002 in a mouse asthma model. Int Immunopharmacol. 2005;5(3):495–502.

    Article  CAS  PubMed  Google Scholar 

  177. Takeda M, Ito W, Tanabe M, Ueki S, Kato H, Kihara J, et al. Allergic airway hyperresponsiveness, inflammation, and remodeling do not develop in phosphoinositide 3-kinase gamma-deficient mice. J Allergy Clin Immunol. 2009;123(4):805–12.

    Article  CAS  PubMed  Google Scholar 

  178. Koziol-White CJ, Yoo EJ, Cao G, Zhang J, Papanikolaou E, Pushkarsky I, et al. Inhibition of PI3K promotes dilation of human small airways in a rho kinase-dependent manner. Br J Pharmacol. 2016;173(18):2726–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. McNamara CR, Degterev A. Small-molecule inhibitors of the PI3K signaling network. Future Med Chem. 2011;3(5):549–65.

    Article  CAS  PubMed  Google Scholar 

  180. Lee HJ, Lee EJ, Seo M. Galpha12 protects vascular endothelial cells from serum Withdrawal-Induced apoptosis through regulation of miR-155. Yonsei Med J. 2016;57(1):247–53.

    Article  CAS  PubMed  Google Scholar 

  181. Huang S, Chen LY, Zuraw BL, Ye RD, Pan ZK. Chemoattractant-stimulated NF-kappaB activation is dependent on the low molecular weight GTPase RhoA. J Biol Chem. 2001;276(44):40977–81.

    Article  CAS  PubMed  Google Scholar 

  182. Wang Y, Cheng J, Tandan S, Jiang M, McCloskey DT, Hill JA. Transient-outward K + channel inhibition facilitates L-type Ca2 + current in heart. J Cardiovasc Electrophysiol. 2006;17(3):298–304.

    Article  PubMed  Google Scholar 

  183. Bi J, Min Z, Yuan H, Jiang Z, Mao R, Zhu T, et al. PI3K inhibitor treatment ameliorates the glucocorticoid insensitivity of PBMCs in severe asthma. Clin Transl Med. 2020;9(1):22.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Webb RC. Smooth muscle contraction and relaxation. Adv Physiol Educ. 2003;27(1–4):201–6.

    Article  PubMed  Google Scholar 

  185. Keglowich LF, Borger P. The Three A’s in asthma - Airway Smooth Muscle, Airway Remodeling & Angiogenesis. Open Respir Med J. 2015;9:70–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Publication made possible in part by support from the Thomas Jefferson University Open Access Fund. 

Funding

This work is supported by NIH grants UL1TR003017 and P01HL114471 (awarded to RAP). ELM is supported by NIH grant P01HL114471-S1.

Author information

Authors and Affiliations

Authors

Contributions

ELM wrote the manuscript and generated the figures, with editorial and content suggestions from RAP and CPS.

Corresponding author

Correspondence to Charles P. Scott.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it.The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

McDuffie, E.L., Panettieri, R.A. & Scott, C.P. G12/13 signaling in asthma. Respir Res 25, 295 (2024). https://doi.org/10.1186/s12931-024-02920-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12931-024-02920-0

Keywords