首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
There are a large number of Rho guanine nucleotide exchange factors, most of which have no known functions. Here, we carried out a short hairpin RNA-based functional screen of Rho-GEFs for their roles in leukocyte chemotaxis and identified Arhgef5 as an important factor in chemotaxis of a macrophage phage-like RAW264.7 cell line. Arhgef5 can strongly activate RhoA and RhoB and weakly RhoC and RhoG, but not Rac1, RhoQ, RhoD, or RhoV, in transfected human embryonic kidney 293 cells. In addition, Gβγ interacts with Arhgef5 and can stimulate Arhgef5-mediated activation of RhoA in an in vitro assay. In vivo roles of Arhgef5 were investigated using an Arhgef-5-null mouse line. Arhgef5 deficiency did not affect chemotaxis of mouse macrophages, T and B lymphocytes, and bone marrow-derived mature dendritic cells (DC), but it abrogated MIP1α-induced chemotaxis of immature DCs and impaired migration of DCs from the skin to lymph node. In addition, Arhgef5 deficiency attenuated allergic airway inflammation. Therefore, this study provides new insights into signaling mechanisms for DC migration regulation.Leukocyte chemotaxis underlies leukocyte migration, infiltration, trafficking, and homing that are not only important for normal leukocyte functions, but also have a important role in inflammation-related diseases. Leukocyte chemotaxis is regulated by leukocyte chemoattractants that include bacterial by-products such as formylmethionylleucylphenylalanine, complement proteolytic fragments such as C5a, and the superfamily of chemotactic cytokines, chemokines. These chemoattractants bind to their specific cell G protein-coupled receptors and are primarily coupled to the Gi family of G proteins to regulate leukocyte chemotaxis. Previous studies have established that the Rho family of small GTPases regulates leukocyte migration (1, 2). Rac, Cdc42, and RhoA are the three best studied Rho small GTPases. In myeloid cells, Cdc42 regulates directionality by directing where F-actin and lamellipodia are formed, and Rac regulates F-actin formation in the lamellipodia, which provides a driving force for cell motility (36). On the other hand, RhoA regulates the formation and contractility of the actomyosin structure at the back that provides a pushing force (5, 7). Rho guanine nucleotide exchange factors (GEF)3 are key regulators for the activity of these small GTPases. GEFs activate small GTPases by promoting the loading of GTP to the small GTPases, a rate-limiting step in GTPase regulation (811). Previous biochemical and genetic studies have revealed how Cdc42 and Rac may be regulated by chemokine receptors in leukocytes. Chemokine receptors can regulate Cdc42 via a Rho-GEF PIXα, which is regulated by Gβγ from the Gi proteins via the interactions between Gβγ and Pak1 and between Pak1 and PIXα in myeloid cells 12. On the other hand, in neutrophils chemokine receptors regulate Rac2 via another Rho-GEF P-Rex1, which is directly regulated by Gβγ (1315). Two Rho-GEFs have been implicated in regulation of RhoA in neutrophils. GEF115 was found in the leading edges of polarized mouse neutrophils, whereas PDZ Rho-GEF was found in the uropods of differentiated HL-60 cells. Both Rho-GEFs were believed to mediate pertussis toxin-resistant activation of RhoA in these cells. However, a significant portion of RhoA activity in leukocytes are pertussis toxin-sensitive, which is presumably regulated by the α and/or βγ subunits from the Gi proteins. The signaling mechanism for this pertussis toxin-sensitive RhoA regulation by chemokine receptors remains largely elusive.Molecular cloning and genomic sequencing have identified more than 70 Rho-GEFs in mammals (1620). Many of these Rho-GEFs have been shown to activate RhoA in in vitro and overexpression assays (1620). However, it is not known if any of them regulate RhoA in vivo, we have found that PIXα is a specific GEF for Cdcd42 in neutrophils (12) despite its potent activity on Rac in in vitro and overexpression assays (21, 22). Therefore, we used a siRNA-based loss of function screen in an attempt to identify the GEFs that regulate myeloid cell migration and RhoA activity. One of the candidates, Arhgef5, was found to be directly activated by Gβγ to regulate RhoA and has an important role in immature DC migration. In addition, Arhgef5 deficiency attenuated allergic airway inflammation in a mouse model.  相似文献   

2.
Differential inhibitors of Gβγ-effector regions are required to dissect the biological contribution of specific Gβγ-initiated signaling pathways. Here, we characterize PhLP-M1-G149, a Gβγ-interacting construct derived from phosducin-like protein 1 (PhLP) as a differential inhibitor of Gβγ, which, in endothelial cells, prevented sphingosine 1-phosphate-induced phosphorylation of AKT, glycogen synthase kinase 3β, cell migration, and tubulogenesis, while having no effect on ERK phosphorylation or hepatocyte growth factor-dependent responses. This construct attenuated the recruitment of phosphoinositide 3-kinase γ (PI3Kγ) to the plasma membrane and the signaling to AKT in response to Gβγ overexpression. In coimmunoprecipitation experiments, PhLP-M1-G149 interfered with the interaction between PI3Kγ and Gβγ. Other PhLP-derived constructs interacted with Gβγ but were not effective inhibitors of Gβγ signaling to AKT or ERK. Our results indicate that PhLP-M1-G149 is a suitable tool to differentially modulate the Gβγ-initiated pathway linking this heterodimer to AKT, endothelial cell migration, and in vitro angiogenesis. It can be also useful to further characterize the molecular determinants of the Gβγ-PI3Kγ interaction.Heterotrimeric G protein signaling depends on the actions of GTP-loaded Gα and free Gβγ, the two functional components of the heterotrimer, leading to the generation of second messengers and cell specific functional events (1, 2). Differential inhibitors of Gβγ are required to dissect the biological impact of different Gβγ-dependent effectors. Gβγ actions can be blocked by competition with peptides derived from its effectors. For example, the effect of Gβγ on adenylyl cyclase II, G protein-activated inward rectifier K+ channel, G protein-coupled receptor kinase 2, and phospholipase Cβ3, is attenuated by a peptide from adenylyl cyclase II (3). In addition, RACK1 (receptor for activated C kinase 1) selectively inhibits the effect the chemokine receptor CXCR2 on the activation of phospholipase Cβ2 and adenylyl cyclase II in HEK293 cells, without affecting other functions of Gβγ (4). Recently, Smrcka and colleagues characterized the effect of small molecule inhibitors of Gβγ, suggesting their potential application in therapeutic strategies targeting particular Gβγ-dependent pathways (5). Emerging possibilities to target this heterodimer in pathological situations such as inflammation and angiogenesis are based on the role of Gβγ in cell survival and chemotaxis. To the best of our knowledge, no molecular tool is yet available to differentially inhibit Gβγ signaling to AKT.3Gβγ is a key transducer of sphingosine 1-phosphate (S1P)-elicited angiogenic signals promoting endothelial cell migration, proliferation, and survival (612). Multiple Gβγ-dependent effectors are potentially involved in the molecular events required for endothelial cell migration. These include lipid kinases such as PI3Kγ and PI3Kβ (13), and a novel family of Rac guanine nucleotide exchange factors, represented by P-REX1, which is activated by Gβγ and phosphatidylinositol 3,4,5-trisphosphate (1416). Gβγ signaling is frequently attributed to pertussis toxin-sensitive Gi coupled receptors, and it has been consistently revealed by the antagonistic effect of the carboxyl-terminal region of G protein-coupled receptor kinase 2, which sequesters Gβγ thereby inhibiting all its intracellular actions (17). In addition, mutational analysis of Gβ revealed that different residues, all of them mapping to the interface of contact between Gβγ and Gα, are important for the activation of distinct Gβγ effector molecules (18).Phosducin was originally identified as a phosphoprotein restricted to the retina and pineal gland forming a complex with Gβγ (19, 20). It was considered a protein kinase A-sensitive regulator of G protein-mediated signaling (21, 22). Further studies identified a family of phosducin-like proteins (PhLPs) (23, 24). Phosducin and Gα share affinity for the same region of Gβγ, as revealed by the structural analysis of Gβγ in complex with Gα or phosducin and by in vitro binding experiments (25). This area of interaction includes some of the residues considered necessary for the activation of Gβγ-dependent effectors (18, 26). It was initially postulated that phosducin and related proteins, by interfering with the availability of free Gβγ, exert an inhibitory role on Gβγ signaling. However, recent genetic evidence raised an apparently conflicting situation; the knockout of PhLP in fungi resulted in a phenotype equivalent to the absence of Gβγ, contrary to its expected role as an inhibitor (27). Novel experimental evidence indicated that PhLP has a positive effect on Gβγ signaling due to its participation in the assembly of the heterodimer, helping to stabilize free Gβ subunits leaving the ribosome after synthesis (2831).Despite the positive role of full-length PhLP in the assembly of Gβγ heterodimers, it is still possible that different fragments of this protein, which could retain their interaction with distinct regions of Gβγ, might function as inhibitors of Gβγ signaling. Accordingly, we characterized here the effect of different PhLP-derived constructs on the signaling pathways elicited by S1P or HGF in endothelial cells. In addition, we explored the mechanism by which PhLP-M1-G149 interferes with Gβγ preventing the activation of AKT.  相似文献   

3.
The G protein βγ subunit dimer (Gβγ) and the Gβ5/regulator of G protein signaling (RGS) dimer play fundamental roles in propagating and regulating G protein pathways, respectively. How these complexes form dimers when the individual subunits are unstable is a question that has remained unaddressed for many years. In the case of Gβγ, recent studies have shown that phosducin-like protein 1 (PhLP1) works as a co-chaperone with the cytosolic chaperonin complex (CCT) to fold Gβ and mediate its interaction with Gγ. However, it is not known what fraction of the many Gβγ combinations is assembled this way or whether chaperones influence the specificity of Gβγ dimer formation. Moreover, the mechanism of Gβ5-RGS assembly has yet to be assessed experimentally. The current study was undertaken to directly address these issues. The data show that PhLP1 plays a vital role in the assembly of Gγ2 with all four Gβ1–4 subunits and in the assembly of Gβ2 with all twelve Gγ subunits, without affecting the specificity of the Gβγ interactions. The results also show that Gβ5-RGS7 assembly is dependent on CCT and PhLP1, but the apparent mechanism is different from that of Gβγ. PhLP1 seems to stabilize the interaction of Gβ5 with CCT until Gβ5 is folded, after which it is released to allow Gβ5 to interact with RGS7. These findings point to a general role for PhLP1 in the assembly of all Gβγ combinations and suggest a CCT-dependent mechanism for Gβ5-RGS7 assembly that utilizes the co-chaperone activity of PhLP1 in a unique way.Eukaryotic cells utilize receptors coupled to heterotrimeric GTP-binding proteins (G proteins)3 to mediate a vast array of responses ranging from nutrient-induced migration of single-celled organisms to neurotransmitter-regulated neuronal activity in the human brain (1). Ligand binding to a G protein-coupled receptor (GPCR) initiates GTP exchange on the G protein heterotrimer (composed of Gα, Gβ, and Gγ subunits), which in turn causes the release of Gα-GTP from the Gβγ dimer (24). Both Gα-GTP and Gβγ propagate and amplify the signal by interacting with effector enzymes and ion channels (1, 5). The duration and amplitude of the signal is dictated by receptor phosphorylation coupled with arrestin binding and internalization (6) and by regulators of G protein signaling (RGS) proteins, which serve as GTPase-activating proteins for the GTP-bound Gα subunit (7, 8). The G protein signaling cycle is reset as the inactive Gα-GDP reassembles with the Gβγ dimer and Gαβγ re-associates with the GPCR (5).To fulfill its essential role in signaling, the G protein heterotrimer must be assembled post-translationally from its nascent polypeptides. Significant progress has been made recently regarding the mechanism by which this process occurs. It has been clear for some time that the Gβγ dimer must assemble first, followed by subsequent association of Gα with Gβγ (9). What has not been clear was how Gβγ assembly would occur given the fact that neither Gβ nor Gγ is structurally stable without the other. An important breakthrough was the finding that phosducin-like protein 1 (PhLP1) functions as a co-chaperone with the chaperonin containing tailless complex polypeptide 1 (CCT) in the folding of nascent Gβ and its association with Gγ (1015). CCT is an important chaperone that assists in the folding of actin and tubulin and many other cytosolic proteins, including many β propeller proteins like Gβ (16). PhLP1 has been known for some time to interact with Gβγ and was initially believed to inhibit Gβγ function (17). However, several recent studies have demonstrated that PhLP1 and CCT work together in a highly orchestrated manner to form the Gβγ dimer (1015).Studies on the mechanism of PhLP1-mediated Gβγ assembly have focused on the most common dimer Gβ1γ2 (10, 13, 14), leaving open questions about the role of PhLP1 in the assembly of the other Gβγ combinations. These are important considerations given that humans possess 5 Gβ genes and 12 Gγ genes with some important splice variants (18, 19), resulting in more than 60 possible combinations of Gβγ dimers. Gβ1–4 share between 80 and 90% sequence identity and are broadly expressed (18, 19). Gβ5, the more atypical isoform, shares only ∼53% identity with Gβ1, carries a longer N-terminal domain, and is only expressed in the central nervous system and retina (20). The Gγ protein family is more heterogeneous than the Gβ family. The sequence identity of the 12 Gγ isoforms extends from 10 to 70% (21), and the Gγ family can be separated into 5 subfamilies (2123). All Gγ proteins carry C-terminal isoprenyl modifications, which contribute to their association with the cell membrane, GPCRs, Gαs, and effectors (9). Subfamily I Gγ isoforms are post-translationally farnesylated, whereas all others are geranylgeranylated (22, 24).There is some inherent selectivity in the assembly of different Gβγ combinations, but in general Gβ1–4 can form dimers with most Gγ subunits (25). The physiological purpose of this large number of Gβγ combinations has intrigued researchers in the field for many years, and a large body of research indicates that GPCRs and effectors couple to a preferred subset of Gβγ combinations based somewhat on specific sequence complementarity, but even more so on cellular expression patterns, subcellular localization, and post-translational modifications (18).In contrast to Gβ1–4, Gβ5 does not interact with Gγ subunits in vivo, but it instead forms irreversible dimers with RGS proteins of the R7 family, which includes RGS proteins 6, 7, 9, and 11 (26). All R7 family proteins contain an N-terminal DEP (disheveled, Egl-10, pleckstrin) domain, a central Gγ-like (GGL) domain, and a C-terminal RGS domain (8, 26). The DEP domain interacts with the membrane anchoring/nuclear shuttling R7-binding protein, and the GGL domain binds to Gβ5 in a manner similar to other Gβγ associations (27, 28). Like Gβγs, Gβ5 and R7 RGS proteins form obligate dimers required for their mutual stability (26). Without their partner, Gβ5 and R7 RGS proteins are rapidly degraded in cells (26, 29). Gβ5-R7 RGS complexes act as important GTPase-accelerating proteins for Gi/oα and Gqα subunits in neuronal cells and some immune cells (26).It has been recently shown that all Gβ isoforms are able to interact with the CCT complex, but to varying degrees (15). Gβ4 and Gβ1 bind CCT better than Gβ2 and Gβ3, whereas Gβ5 binds CCT poorly (15). These results suggest that Gβ1 and Gβ4 might be more dependent on PhLP1 than the other Gβs, given the co-chaperone role of PhLP1 with CCT in Gβ1γ2 assembly. However, another report has indicated that Gγ2 assembly with Gβ1 and Gβ2 is more PhLP1-dependent than with Gβ3 and Gβ4 (30). Thus, it is not clear from current information whether PhLP1 and CCT participate in assembly of all Gβγ combinations or whether they contribute to the specificity of Gβγ dimer formation, nor is it clear whether they or other chaperones are involved in Gβ5-R7 RGS dimer formation. This report was designed to address these issues.  相似文献   

4.
It is well known that Gαi1(GDP) binds strongly to Gβγ subunits to form the Gαi1(GDP)-Gβγ heterotrimer, and that activation to Gαi1(GTP) results in conformational changes that reduces its affinity for Gβγ subunits. Previous studies of G protein subunit interactions have used stoichiometric amounts of the proteins. Here, we have found that Gαi1(GDP) can bind a second Gβγ subunit with an affinity only 10-fold weaker than the primary site and close to the affinity between activated Gαi1 and Gβγ subunits. Also, we find that phospholipase Cβ2, an effector of Gβγ, does not compete with the second binding site implying that effectors can be bound to the Gαi1(GDP)-(Gβγ)2 complex. Biophysical measurements and molecular docking studies suggest that this second site is distant from the primary one. A synthetic peptide having a sequence identical to the putative second binding site on Gαi1 competes with binding of the second Gβγ subunit. Injection of this peptide into cultured cells expressing eYFP-Gαi1(GDP) and eCFP-Gβγ reduces the overall association of the subunits suggesting this site is operative in cells. We propose that this second binding site serves to promote and stabilize G protein subunit interactions in the presence of competing cellular proteins.The plasma membranes of cells are organized as a series of protein-rich and lipid-rich domains (13). Many of the protein-rich domains, in particular those organized by caveolin proteins, are thought to be complexes of functionally related proteins that transduce extracellular signals (2). There is increasing evidence that heterotrimeric G proteins exist in pre-formed membrane complexes with their receptors and their intracellular effectors (48).The G protein signaling system is initiated when an extracellular agonist binds to its specific G protein-coupled receptor (for review see Refs. 912). The ligand-bound receptor will then catalyze the exchange of GTP for GDP on the Gα subunit in the G protein heterotrimer. In the basal state, Gα(GDP) binds strongly to Gβγ, but in the GTP-bound state this affinity is reduced, allowing Gα(GTP) and Gβγ subunits to individually bind to a host of specific intracellular enzymes and change their catalytic activity.Although the interactions between G protein subunits have been studied extensively in vitro, their behavior in cells may differ. For example, in pure or semi-pure systems, activation of Gα(GDP) sufficiently weakens its affinity for Gβγ resulting in dissociation (13). However, in cells separation of the heterotrimer is observed under some circumstances, but not others (7, 1417). The reason for these differences in behavior is not clear. There are four families of Gα subunits that each contain several members, and, additionally, there are many subtypes of Gβγ subunits (18). It is possible that differences in dissociation behavior reflect differences in affinity between G protein subunit subtypes (19), the presence of various protein partners, and/or differences in post-synthetic modifications of the subunits (20).The mechanism that allows activated G proteins to remain bound is not apparent from the crystal structure (21, 22). If G protein subunits do not dissociate in cells, then their interaction must change in such a manner as to expose the effector interaction site(s). We have found that phospholipase Cβ1 (PLCβ1),4 an important effector of Gαq (23), is bound to Gαq prior to activation and throughout the activation cycle (6) implying that Gαq(GDP) interacts with PLCβ1 in a non-functional manner.We have evidence that signaling complexes are stabilized by a series of secondary interactions. Using purified proteins and model membranes, we have found that membranes of the Gαq-Gβγ/PLCβ1/RGS4 signaling system have secondary, weaker binding sites to members of this signaling system in addition to their high affinity site(s) to their functional partner(s). We speculate that secondary contacts allow for self-scaffolding of signaling proteins. To understand the nature of these secondary contacts, we have studied the ability of the Gαi1(GDP)-Gβγ heterotrimer to remain complexed through the activation cycle (24). Here, we present evidence that Gαi1(GDP) has two distinct Gβγ binding sites that only differ in affinity by an order of magnitude and may allow for continued association between the subunits upon activation. We also find that this site plays an important role in stabilizing G protein associations in cells and provides a mechanism of self-scaffolding.  相似文献   

5.
“Regulator of G-protein signaling” (RGS) proteins facilitate the termination of G protein-coupled receptor (GPCR) signaling via their ability to increase the intrinsic GTP hydrolysis rate of Gα subunits (known as GTPase-accelerating protein or “GAP” activity). RGS2 is unique in its in vitro potency and selectivity as a GAP for Gαq subunits. As many vasoconstrictive hormones signal via Gq heterotrimer-coupled receptors, it is perhaps not surprising that RGS2-deficient mice exhibit constitutive hypertension. However, to date the particular structural features within RGS2 determining its selectivity for Gαq over Gαi/o substrates have not been completely characterized. Here, we examine a trio of point mutations to RGS2 that elicits Gαi-directed binding and GAP activities without perturbing its association with Gαq. Using x-ray crystallography, we determined a model of the triple mutant RGS2 in complex with a transition state mimetic form of Gαi at 2.8-Å resolution. Structural comparison with unliganded, wild type RGS2 and of other RGS domain/Gα complexes highlighted the roles of these residues in wild type RGS2 that weaken Gαi subunit association. Moreover, these three amino acids are seen to be evolutionarily conserved among organisms with modern cardiovascular systems, suggesting that RGS2 arose from the R4-subfamily of RGS proteins to have specialized activity as a potent and selective Gαq GAP that modulates cardiovascular function.G protein-coupled receptors (GPCRs)4 form an interface between extracellular and intracellular physiology, as they convert hormonal signals into changes in intracellular metabolism and ultimately cell phenotype and function (13). GPCRs are coupled to their underlying second messenger systems by heterotrimeric guanine nucleotide-binding protein (“G-proteins”) composed of three subunits: Gα, Gβ, and Gγ. Four general classes of Gα subunits have been defined based on functional couplings (in the GTP-bound state) to various effector proteins. Gs subfamily Gα subunits are stimulatory to membrane-bound adenylyl cyclases that generate the second messenger 3′,5′-cyclic adenosine monophosphate (cAMP); conversely, Gi subfamily Gα subunits are generally inhibitory to adenylyl cyclases (4). G12/13 subfamily Gα subunits activate the small G-protein RhoA through stimulation of the GEF subfamily of RGS proteins, namely p115-RhoGEF, LARG, and PDZ-RhoGEF (5). Gq subfamily Gα subunits are potent activators of phospholipase-Cβ enzymes that generate the second messengers diacylglycerol and inositol triphosphate (6); more recently, two additional Gαq effector proteins have been described: the receptor kinase GRK2 and the RhoA nucleotide exchange factor p63RhoGEF (7, 8).The duration of GPCR signaling is controlled by the time Gα remains bound to GTP before its hydrolysis to GDP. RGS proteins are key modulators of GPCR signaling by virtue of their ability to accelerate the intrinsic GTP hydrolysis activity of Gα subunits (reviewed in Refs. 9 and 10). RGS2/G0S8, one of the first mammalian RGS proteins identified (11) and member of the R4-subfamily (10), has a critical role in the maintenance of normostatic blood pressure both in mouse models (12, 13) and in humans (14, 15); additionally, Rgs2-deficient mice exhibit impaired aggression and increased anxiety (16, 17), behavioral phenotypes with potential human clinical correlates (18, 19).Although many RGS proteins are promiscuous and thus act on multiple Gα substrates in vitro (e.g. Ref. 20), RGS2 exhibits exquisite specificity for Gαq in biochemical binding assays and single turnover GTPase acceleration assays (20, 21). Consistent with this in vitro selectivity,5 mice deficient in RGS2 uniquely exhibit constitutive hypertension and prolonged responses to vasoconstrictors, as would be expected upon loss of a potent negative regulator of Gαq that mediates signaling from various vasoconstrictive hormones such as angiotensin II, endothelin, thrombin, norepinephrine, and vasopressin (22). In addition, RGS2-deficient mice respond to sustained pressure overload with an accelerated time course of maladaptive cardiac remodeling (23), a pathophysiological response that evokes myocardial hypertrophy known to be critically dependent on Gαq signaling (24, 25).To gain insight into the structural basis of the unique Gα substrate selectivity exhibited by RGS2, a series of point mutants in RGS2 were evaluated that enable this protein to bind and accelerate GTP hydrolysis by Gαi; we subsequently delineated the structural determinants of the Gαi/mutant RGS2 interaction using x-ray crystallography. Three key positions, first identified by Heximer and colleagues (21) and highlighted in our structural studies as key determinants of RGS2 substrate selection, were also found to be conserved throughout the evolution of the RGS2 protein in a manner suggestive of specialization toward cardiovascular signaling modulation.  相似文献   

6.
7.
Heterotrimeric G proteins are critical transducers of cellular signaling. Of the four families of G proteins, the physiological function of Gα13 is less well understood. Gα13 gene-deleted mice die at embryonic day ∼9.5. Here, we show that heterozygous Gα13+/− mice display defects in adult angiogenesis. Female Gα13+/− mice showed a higher number of immature follicles and a lower density of blood vessels in the mature corpus luteum compared with Gα13+/+ mice. Furthermore, implanted tumors grew slower in Gα13+/− host mice. These tumor tissues had many fewer blood vessels compared with those from Gα13+/+ host mice. Moreover, bone marrow-derived progenitor cells from Gα13+/+ mice rescued the failed growth of allografted tumors when reconstituted into irradiated Gα13+/− mice. Hence, Gα13 is haploinsufficient for adult angiogenesis in both the female reproductive system and tumor angiogenesis.A structurally diverse repertoire of ligands, from photons to large peptides, activates G protein-coupled receptors to elicit their physiological functions (1). In turn, ligand-bound G protein-coupled receptors function as guanine nucleotide exchange factors, catalyzing the exchange of GDP bound on the Gα subunit with GTP in the presence of Gβγ and causing the dissociation of the Gα subunit from the Gβγ dimer to form two functional units (Gα and Gβγ) (2). Both Gα and Gβγ subunits signal to various cellular pathways. Based on sequence and functional homologies, G proteins are grouped into four families: Gs, Gi, Gq, and G12 (3). Of these four subfamilies of G proteins, the physiological function of the G12 subfamily is less well understood. In this family, there are two members, G12 and G13. Gα12 knock-out mice appear normal (4). Gα13 knock-out mice display embryonic lethality (embryonic day ∼9.5) (5). Gα13−/− mouse embryos have defective vascular systems (5). Endothelial cell-specific deletion of Gα13 also results in vascular defect and embryonic lethality (6). The molecular basis that underlies the vascular defect observed in Gα13−/− mouse embryos has not been defined.Angiogenesis (formation of endothelium-lined blood vessels) is essential for organ growth in the embryo and for repair of wounded tissues in the adult (7, 8). An imbalance in angiogenesis contributes to the pathogenesis of numerous malignant, inflammatory, ischemic, infectious, and immune disorders and cancers (7, 8). Most angiogenesis events take place during embryonic development. In adult tissues, the majority of endothelial cells are quiescent, and angiogenesis occurs only rarely except in a few adult tissues (including ovary) that exhibit periodic and dynamic growth and regression (911). Under pathological conditions such as tumor growth, adult angiogenesis is induced. Tumor angiogenesis is the proliferation of a network of blood vessels that penetrates into cancerous growths (including implanted tumor tissues), supplying nutrients and oxygen and removing waste products. Solid tumors depend on angiogenesis for growth and metastasis in a hostile environment (12). Bone marrow is the origin of endothelial progenitor cells in the adult. Bone marrow-derived endothelial progenitor cells are mobilized into peripheral blood and recruited to the foci of pathophysiological neovascularization and re-endothelialization, thereby contributing to vascular regeneration (13). Vascular endothelial growth factor (VEGF),2 the most critical factor for angiogenesis, is an important factor for the mobilization of endothelial progenitor cells from bone marrow (7, 8). Bone marrow transplantation experiments have demonstrated the incorporation of bone marrow-derived endothelial progenitor cells into foci of pathological neovascularization such as growing tumors, healing wounds, ischemic skeletal and cardiac muscles, and cornea receiving micropocket surgery (1421).Here, we show that heterozygous Gα13+/− mice display defects in adult angiogenesis. We found that female Gα13+/− mice show a higher number of immature follicles and a lower density of blood vessels in the mature corpus luteum compared with Gα13+/+ mice. Furthermore, implanted tumors grew slower in Gα13+/− host mice. These tumor tissues had many fewer blood vessels compared with those from Gα13+/+ host mice. We also down-regulated Gα13 in endothelial cells by RNA interference and show that defective migration and tube formation in response to VEGF likely contribute to the impaired angiogenesis. Moreover, bone marrow-derived cells from Gα13+/+ mice rescued the failed growth of allografted tumors when reconstituted into irradiated Gα13+/− mice. Our results demonstrate that Gα13 is haploinsufficient for adult angiogenesis in both the female reproductive system and tumor angiogenesis. This role in adult angiogenesis provides a suitable system to further investigate the biochemical and physiological functions of Gα13. Moreover, Gα13 inhibition could be explored for anticancer drug development.  相似文献   

8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
The simultaneous activation of many distinct G protein-coupled receptors (GPCRs) and heterotrimeric G proteins play a major role in various pathological conditions. Pan-inhibition of GPCR signaling by small molecules thus represents a novel strategy to treat various diseases. To better understand such therapeutic approach, we have characterized the biomolecular target of BIM-46187, a small molecule pan-inhibitor of GPCR signaling. Combining bioluminescence and fluorescence resonance energy transfer techniques in living cells as well as in reconstituted receptor-G protein complexes, we observed that, by direct binding to the Gα subunit, BIM-46187 prevents the conformational changes of the receptor-G protein complex associated with GPCR activation. Such a binding prevents the proper interaction of receptors with the G protein heterotrimer and inhibits the agonist-promoted GDP/GTP exchange. These observations bring further evidence that inhibiting G protein activation through direct binding to the Gα subunit is feasible and should constitute a new strategy for therapeutic intervention.G protein-coupled receptors (GPCRs)3 represent the largest superfamily of signaling proteins with a very high impact on drug discovery (1). Approximately 30% of the current drug targets are indeed GPCRs and these latter are involved in all major disease areas (2). The classical drug discovery process selects and optimizes compounds that interact selectively with a specific receptor (1), but recent reports show that certain critical conditions such as cancer (3) or pain (4) are driven by the concomitant activation of many different GPCRs (5). Novel therapeutic strategies could therefore emerge from the simultaneous blockade of the various GPCRs involved in such pathologies. The GPCR signaling downstream cascade triggers several protein/protein interactions that may be blocked or modulated by small molecules (6). Such protein/protein interactions involve the GPCR transmembrane domain and the heterotrimeric G protein complex, composed of an α subunit (Gα) and a βγ dimer (Gβγ), which interact sequentially with several partners (e.g. guanine nucleotides, effectors, and regulatory proteins) (7). This offers multiple possibilities to develop small molecules controlling heterotrimeric G protein signaling (6, 8, 9). For example, Higashijima et al. (10, 11) showed that Mastoparan, a peptide toxin from wasp venom, directly acts on G proteins to mimic the role played by the activated receptors. The anti-helminthic drug Suramin and some analogs represent a second class of compounds that directly interact with G proteins and interfere with nucleotide exchange (1214). Small molecules modulating regulator of G protein signaling proteins have also been proposed for drug development (15). More recently, Bonacci et al. (16) have described fluorescein analogs that display central pain relief activity via binding to the Gβγ subunits. From our own group, we have reported in vivo inhibition of the GPCR signaling pathway by two closely related imidazopirazine containing small molecules, displaying potent antiproliferative activity (BIM-46174) (17) and potent pain relief activity (BIM-46187) (18).Here, we examined the molecular mechanisms underlying the biological activity of BIM-46187 with the various constituents of the GPCR signaling pathways. We report that this small molecule prevents GPCR-G protein signaling through a selective binding to the Gα protein subunit. Our results support the concept of targeting and inhibiting the heterotrimeric G protein complex as an approach to treat certain pathologies involving simultaneous activation of several GPCRs and/or heterotrimeric G proteins.  相似文献   

18.
19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号