Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS One ; 12(3): e0168638, 2017.
Article in English | MEDLINE | ID: mdl-28253301

ABSTRACT

Although techniques for cell-specific gene expression via viral transfer have advanced, many challenges (e.g., viral vector design, transduction of genes into specific target cells) still remain. We investigated a novel, simple methodology for using adenovirus transfer to target specific cells of the kidney tubules for the expression of exogenous proteins. We selected genes encoding sodium-dependent phosphate transporter type 2a (NPT2a) in the proximal tubule, sodium-potassium-2-chloride cotransporter (NKCC2) in the thick ascending limb of Henle (TALH), and aquaporin 2 (AQP2) in the collecting duct. The promoters of the three genes were linked to a GFP-coding fragment, the final constructs were then incorporated into an adenovirus vector, and this was then used to generate gene-manipulated viruses. After flushing circulating blood, viruses were directly injected into the renal arteries of rats and were allowed to site-specifically expression in tubule cells, and rats were then euthanized to obtain kidney tissues for immunohistochemistry. Double staining with adenovirus-derived EGFP and endogenous proteins were examined to verify orthotopic expression, i.e. "adenovirus driven NPT2a-EGFP and endogenous NHE3 protein", "adenovirus driven NKCC2-EGFP and endogenous NKCC2 protein" and "adenovirus driven AQP2-EGFP and endogenous AQP2 protein". Owing to a lack of finding good working anti-NPT2a antibody, an antibody against a different protein (sodium-hydrogen exchanger 3 or NHE3) that is also specifically expressed in the proximal tubule was used. Kidney structures were well-preserved, and other organ tissues did not show EGFP staining. Our gene transfer method is easier than using genetically engineered animals, and it confers the advantage of allowing the manipulation of gene transfer after birth. This is the first method to successfully target gene expression to specific cells in the kidney tubules. This study may serve as the first step for safe and effective gene therapy in the kidney tubule diseases.


Subject(s)
Adenoviridae/genetics , Kidney Tubules/cytology , Kidney Tubules/metabolism , Promoter Regions, Genetic/genetics , Transduction, Genetic/methods , Gene Expression , Genetic Vectors/genetics , HEK293 Cells , Humans , Safety
2.
Cell Signal ; 28(1): 43-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26483157

ABSTRACT

Rho GTPases are integral to the regulation of actin cytoskeleton-dependent processes, including mitosis. Rho and leukemia-associated Rho guanine-nucleotide exchange factor (LARG), also known as ARHGEF12, are involved in mitosis as well as diseases such as cancer and heart disease. Since LARG has a role in mitosis and diverse signaling functions beyond mitosis, it is important to understand the regulation of the protein through modifications such as phosphorylation. Here we report that LARG undergoes a mitotic-dependent and cyclin-dependent kinase 1 (Cdk1) inhibitor-sensitive phosphorylation. Additionally, LARG is phosphorylated at the onset of mitosis and dephosphorylated as cells exit mitosis, concomitant with Cdk1 activity. Furthermore, using an in vitro kinase assay, we show that LARG can be directly phosphorylated by Cdk1. Through expression of phosphonull mutants that contain non-phosphorylatable alanine mutations at potential Cdk1 S/TP sites, we demonstrate that LARG phosphorylation occurs in both termini. Using phosphospecific antibodies, we confirm that two sites, serine 190 and serine 1176, are phosphorylated during mitosis in a Cdk1-dependent manner. In addition, these phosphospecific antibodies show phosphorylated LARG at specific mitotic locations, namely the mitotic organizing centers and flanking the midbody. Lastly, RhoA activity assays reveal that phosphonull LARG is more active in cells than phosphomimetic LARG. Our data thus identifies LARG as a phosphoregulated RhoGEF during mitosis.


Subject(s)
CDC2 Protein Kinase/metabolism , Leukemia/metabolism , Mitosis/physiology , Rho Guanine Nucleotide Exchange Factors/metabolism , Actin Cytoskeleton/metabolism , HeLa Cells , Humans , Phosphorylation , Rho Guanine Nucleotide Exchange Factors/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
3.
Mol Pharmacol ; 86(3): 252-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24958816

ABSTRACT

Gastrin-releasing peptide receptor (GRPR) is ectopically expressed in over 60% of colon cancers. GRPR expression has been correlated with increased colon cancer cell migration. However, the signaling pathway by which GRPR activation leads to increased cancer cell migration is not well understood. We set out to molecularly dissect the GRPR signaling pathways that control colon cancer cell migration through regulation of small GTPase RhoA. Our results show that GRP stimulation activates RhoA predominantly through G13 heterotrimeric G-protein signaling. We also demonstrate that postsynaptic density 95/disk-large/ZO-1 (PDZ)-RhoGEF (PRG), a member of regulator of G-protein signaling (RGS)-homology domain (RH) containing guanine nucleotide exchange factors (RH-RhoGEFs), is the predominant activator of RhoA downstream of GRPR. We found that PRG is required for GRP-stimulated colon cancer cell migration, through activation of RhoA-Rho-associated kinase (ROCK) signaling axis. In addition, PRG-RhoA-ROCK pathway also contributes to cyclo-oxygenase isoform 2 (Cox-2) expression. Increased Cox-2 expression is correlated with increased production of prostaglandin-E2 (PGE2), and Cox-2-PGE2 signaling contributes to total GRPR-mediated cancer cell migration. Our analysis reveals that PRG is overexpressed in colon cancer cell lines. Overall, our results have uncovered a key mechanism for GRPR-regulated colon cancer cell migration through the Gα13-PRG-RhoA-ROCK pathway.


Subject(s)
Colonic Neoplasms/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Bombesin/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Zonula Occludens-1 Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Caco-2 Cells , Cell Movement , Colonic Neoplasms/pathology , Cyclooxygenase 2/biosynthesis , Dinoprostone/biosynthesis , Disks Large Homolog 4 Protein , HT29 Cells , Humans , Protein Structure, Tertiary , Signal Transduction , rho-Associated Kinases/metabolism
4.
Blood ; 123(3): 442-50, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24081657

ABSTRACT

von Willebrand factor (vWF) secretion by endothelial cells (ECs) is essential for hemostasis and thrombosis; however, the molecular mechanisms are poorly understood. Interestingly, we observed increased bleeding in EC-Gα13(-/-);Gα12(-/-) mice that could be normalized by infusion of human vWF. Blood from Gα12(-/-) mice exhibited significantly reduced vWF levels but normal vWF multimers and impaired laser-induced thrombus formation, indicating that Gα12 plays a prominent role in EC vWF secretion required for hemostasis and thrombosis. In isolated buffer-perfused mouse lungs, basal vWF levels were significantly reduced in Gα12(-/-), whereas thrombin-induced vWF secretion was defective in both EC-Gαq(-/-);Gα11(-/-) and Gα12(-/-) mice. Using siRNA in cultured human umbilical vein ECs and human pulmonary artery ECs, depletion of Gα12 and soluble N-ethylmaleimide-sensitive-fusion factor attachment protein α (α-SNAP), but not Gα13, inhibited both basal and thrombin-induced vWF secretion, whereas overexpression of activated Gα12 promoted vWF secretion. In Gαq, p115 RhoGEF, and RhoA-depleted human umbilical vein ECs, thrombin-induced vWF secretion was reduced by 40%, whereas basal secretion was unchanged. Finally, in vitro binding assays revealed that Gα12 N-terminal residues 10-15 mediated the binding of Gα12 to α-SNAP, and an engineered α-SNAP binding-domain minigene peptide blocked basal and evoked vWF secretion. Discovery of obligatory and complementary roles of Gα12 and Gαq/11 in basal vs evoked EC vWF secretion may provide promising new therapeutic strategies for treatment of thrombotic disease.


Subject(s)
Endothelial Cells/cytology , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , rhoA GTP-Binding Protein/metabolism , von Willebrand Factor/metabolism , Animals , Antibodies, Monoclonal/chemistry , Gene Expression Regulation , Hemostasis , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Knockout , Platelet Adhesiveness , Protein Binding , RNA, Small Interfering/metabolism , Signal Transduction , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/chemistry , Thrombosis
5.
Cell Signal ; 25(11): 2085-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23816534

ABSTRACT

p115RhoGEF is a member of a family of Rho-specific guanine nucleotide exchange factors that also contains a regulator of G protein signaling homology domain (RH-RhoGEFs) that serves as a link between Gα13 signaling and RhoA activation. While the mechanism of regulation of p115RhoGEF by Gα13 is becoming well-known, the role of other regulatory mechanisms, such as post-translational modification or autoinhibition, in mediating p115RhoGEF activity is less well-characterized. Here, putative phosphorylation sites on p115RhoGEF are identified and characterized. Mutation of Ser(330) leads to a decrease in serum response element-mediated transcription as well as decreased activation by Gα13 in vitro. Additionally, this study provides the first report of the binding kinetics between full-length p115RhoGEF and RhoA in its various nucleotide states and examines the binding kinetics of phospho-mutant p115RhoGEF to RhoA. These data, together with other recent reports on regulatory mechanisms of p115RhoGEF, suggest that this putative phosphorylation site serves as a means for initiation or relief of autoinhibition of p115RhoGEF, providing further insight into the regulation of its activity.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Expression Regulation , Protein Processing, Post-Translational , Rho Guanine Nucleotide Exchange Factors/metabolism , Serine/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Sequence , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Genes, Reporter , Humans , Kinetics , Luciferases/genetics , Luciferases/metabolism , Molecular Sequence Data , Mutation , Phosphorylation , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Serum Response Element , Signal Transduction , rhoA GTP-Binding Protein/genetics
6.
J Biochem ; 150(4): 357-69, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21873336

ABSTRACT

G protein-mediated signal transduction can transduce signals from a large variety of extracellular stimuli into cells and is the most widely used mechanism for cell communication at the membrane. The RhoGTPase family has been well established as key regulators of cell growth, differentiation and cell shape changes. Among G protein-mediated signal transduction, G12/13-mediated signalling is one mechanism to regulate RhoGTPase activity in response to extracellular stimuli. The alpha subunits of G12 or G13 have been shown to interact with members of the RH domain containing guanine nucleotide exchange factors for Rho (RH-RhoGEF) family of proteins to directly connect G protein-mediated signalling and RhoGTPase signalling. The G12/13-RH-RhoGEF signalling mechanism is well conserved over species and is involved in critical steps for cell physiology and disease conditions, including embryonic development, oncogenesis and cancer metastasis. In this review, we will summarize current progress on this important signalling mechanism.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , Animals , Humans
7.
J Immunol Methods ; 369(1-2): 108-14, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21570402

ABSTRACT

The generation of antibodies against self-antigens or antigens having a high degree of structural homology with self-antigens is a difficult task because of immunological tolerance. CD4(+)CD25(+) regulatory T cells play an important role in maintaining peripheral tolerance. Sakaguchi et al. previously reported that the transfusion of CD25(+) cell-depleted mouse splenocytes into syngeneic nude mice results in a breaking of peripheral tolerance that leads to the development of autoimmunity. In this study, we attempted to apply this mouse model to the generation of antibodies against self-antigens. We depleted CD25(+) cells from BALB/c mouse splenocytes and transferred the rest of the cells into syngeneic nude mice. The animals were immunized with mouse thyroglobulin. We observed a significant increase of the anti-mouse thyroglobulin antibody titer in the group of mice immunized twice within 10 days after the cell transfer (P<0.05). From these mice, we established hybridoma cell lines producing anti-mouse thyroglobulin monoclonal antibodies, including a clone with a dissociation constant of 10(-8)M. Control nude mice which received CD25(+) cell-containing BALB/c splenocytes did not produce anti-mouse thyroglobulin antibodies. When the CD25(-)cell-transferred nude mice were immunized with mouse Gα12, another self-antigen, anti-Gα12 antibodies were produced in the sera. This method should prove highly useful in the generation of antibodies against self-antigens or antigens for which the structure is highly conserved across species.


Subject(s)
Antibodies/immunology , Antibody Formation , Autoantigens/immunology , Immunization/methods , T-Lymphocytes, Regulatory/immunology , Animals , Autoantigens/administration & dosage , Cell Separation , Female , Interleukin-2 Receptor alpha Subunit/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Spleen/immunology
8.
Neurosignals ; 17(1): 55-70, 2009.
Article in English | MEDLINE | ID: mdl-19212140

ABSTRACT

Accumulating data indicate that G12 subfamily (Galpha12/13)-mediated signaling pathways play pivotal roles in a variety of physiological processes, while aberrant regulation of this pathway has been identified in various human diseases. It has been demonstrated that Galpha12/13-mediated signals form networks with other signaling proteins at various levels, from cell surface receptors to transcription factors, to regulate cellular responses. Galpha12/13 have slow rates of nucleotide exchange and GTP hydrolysis, and specifically target RhoGEFs containing an amino-terminal RGS homology domain (RH-RhoGEFs), which uniquely function both as a GAP and an effector for Galpha12/13. In this review, we will focus on the mechanisms regulating the Galpha12/13 signaling system, particularly the Galpha12/13-RH-RhoGEF-Rho pathway, which can regulate a wide variety of cellular functions from migration to transformation.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Signal Transduction , Animals , Cardiovascular Diseases/metabolism , GTP-Binding Protein Regulators/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Immune System/physiology , Neoplasms/metabolism , Spodoptera , rho GTP-Binding Proteins/metabolism
9.
J Biol Chem ; 284(8): 5000-9, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19074425

ABSTRACT

The transient protein-protein interactions induced by guanine nucleotide-dependent conformational changes of G proteins play central roles in G protein-coupled receptor-mediated signaling systems. Leukemia-associated RhoGEF (LARG), a guanine nucleotide exchange factor for Rho, contains an RGS homology (RH) domain and Dbl homology/pleckstrin homology (DH/PH) domains and acts both as a GTPase-activating protein (GAP) and an effector for Galpha(13). However, the molecular mechanism of LARG activation upon Galpha(13) binding is not yet well understood. In this study, we analyzed the Galpha(13)-LARG interaction using cellular and biochemical methods, including a surface plasmon resonance (SPR) analysis. The results obtained using various LARG fragments demonstrated that active Galpha(13) interacts with LARG through the RH domain, DH/PH domains, and C-terminal region. However, an alanine substitution at the RH domain contact position in Galpha(13) resulted in a large decrease in affinity. Thermodynamic analysis revealed that binding of Galpha(13) proceeds with a large negative heat capacity change (DeltaCp degrees ), accompanied by a positive entropy change (DeltaS degrees ). These results likely indicate that the binding of Galpha(13) with the RH domain triggers conformational rearrangements between Galpha(13) and LARG burying an exposed hydrophobic surface to create a large complementary interface, which facilitates complex formation through both GAP and effector interfaces, and activates the RhoGEF. We propose that LARG activation is regulated by an induced-fit mechanism through the GAP interface of Galpha(13).


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Amino Acid Substitution , GTP-Binding Protein alpha Subunits, G12-G13/chemistry , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Mutation, Missense , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Rho Guanine Nucleotide Exchange Factors , Surface Plasmon Resonance , Thermodynamics
11.
Methods Enzymol ; 390: 285-94, 2004.
Article in English | MEDLINE | ID: mdl-15488184

ABSTRACT

Three mammalian Rho guanine nucleotide exchange factors (RhoGEFs), leukemia-associated RhoGEF (LARG), p115RhoGEF, and PDZ-RhoGEF, contain regulator of G-protein signaling (RGS) domains within their amino-terminal regions. These RhoGEFs link signals from heterotrimeric G12/13 protein-coupled receptors to Rho GTPase activation, leading to various cellular responses, such as actin reorganization and gene expression. The activity of these RhoGEFs is regulated by Galpha12/13 through their RGS domains. Because RhoGEFs stimulate guanine nucleotide exchange by Rho GTPases, RhoGEF activation can be measured by monitoring GTP binding to or GDP dissociation from Rho GTPases. This article describes methods used to perform reconstitution assays to measure the activity of RhoGEFs regulated by Galpha12/13.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Guanine Nucleotide Exchange Factors/metabolism , RGS Proteins/metabolism , Animals , Cell Line , GTP-Binding Protein alpha Subunits, G12-G13/isolation & purification , Guanine Nucleotide Exchange Factors/isolation & purification , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine Diphosphate/metabolism , Phosphorylation , Rho Guanine Nucleotide Exchange Factors , rhoA GTP-Binding Protein/isolation & purification , rhoA GTP-Binding Protein/metabolism
12.
Mol Pharmacol ; 66(4): 1029-34, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15258251

ABSTRACT

Heterotrimeric G proteins of the G12 family regulate the Rho GTPase through RhoGEFs that contain an amino-terminal regulator of G protein signaling (RGS) domain (RGS-RhoGEFs). Direct regulation of the activity of RGS-RhoGEFs p115 or leukemia-associated RhoGEF (LARG) by Galpha13 has previously been demonstrated. However, the precise biochemical mechanism by which Galpha13 stimulates the RhoGEF activity of these proteins has not yet been well understood. Based on the crystal structure of Galphai1 in complex with RGS4, we mutated the Galpha13 residue lysine 204 to alanine (Galpha13K204A) and characterized the effect of this mutation in its regulation of RGS-RhoGEFs p115 or LARG. Compared with wild-type Galpha13, Galpha13K204A induced much less serum-response factor activation when expressed in HeLa cells. Recombinant Galpha13K204A exhibits normal function in terms of nucleotide binding, basal GTP hydrolysis, and formation of heterotrimer with betagamma. We found that lysine 204 of Galpha13 is important for interaction with the RGS domain of p115 or LARG and for the GTPase-activating protein activity of these proteins. In addition, the K204A mutation of Galpha13 impaired its regulation of the RhoGEF activity of p115 or LARG. We conclude that lysine 204 of Galpha13 is important for interaction with RGS-RhoGEFs and is critically involved in the regulation of their activity.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Lysine/physiology , Alanine/genetics , Amino Acid Substitution , Animals , COS Cells , Guanine Nucleotide Exchange Factors/genetics , HeLa Cells , Humans , Leukemia/metabolism , Lysine/genetics , Mutation , Rho Guanine Nucleotide Exchange Factors
13.
Nat Cell Biol ; 5(12): 1095-103, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14634662

ABSTRACT

G alpha 13 stimulates the guanine nucleotide exchange factors (GEFs) for Rho, such as p115Rho-GEF. Activated Rho induces numerous cellular responses, including actin polymerization, serum response element (SRE)-dependent gene transcription and transformation. p115Rho-GEF contains a Regulator of G protein Signalling domain (RGS box) that confers GTPase activating protein (GAP) activity towards G alpha 12 and G alpha 13 (ref. 3). In contrast, classical RGS proteins (such as RGS16 and RGS4) exhibit RGS domain-dependent GAP activity on G alpha i and G alpha q, but not G alpha 12 or G alpha 13 (ref 4). Here, we show that RGS16 inhibits G alpha 13-mediated, RhoA-dependent reversal of stellation and SRE activation. The RGS16 amino terminus binds G alpha 13 directly, resulting in translocation of G alpha 13 to detergent-resistant membranes (DRMs) and reduced p115Rho-GEF binding. RGS4 does not bind G alpha 13 or attenuate G alpha 13-dependent responses, and neither RGS16 nor RGS4 affects G alpha 12-mediated signalling. These results elucidate a new mechanism whereby a classical RGS protein regulates G alpha 13-mediated signal transduction independently of the RGS box.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Proteins/metabolism , RGS Proteins/metabolism , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , Cell Line, Tumor , Feedback, Physiological/genetics , Gene Expression Regulation/genetics , Genes, Regulator/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Rho Guanine Nucleotide Exchange Factors
14.
Proc Natl Acad Sci U S A ; 100(2): 733-8, 2003 Jan 21.
Article in English | MEDLINE | ID: mdl-12515866

ABSTRACT

Heterotrimeric G proteins, G12 and G13, have been shown to transduce signals from G protein-coupled receptors to activate Rho GTPase in cells. Recently, we identified p115RhoGEF, one of the guanine nucleotide exchange factors (GEFs) for Rho, as a direct link between Galpha13 and Rho [Kozasa, T., et al. (1998) Science 280, 2109-2111; Hart, M. J., et al. (1998) Science 280, 2112-2114]. Activated Galpha13 stimulated the RhoGEF activity of p115 through interaction with the N-terminal RGS domain. However, Galpha12 could not activate Rho through p115, although it interacted with the RGS domain of p115. The biochemical mechanism from Galpha12 to Rho activation remained unknown. In this study, we analyzed the interaction of leukemia-associated RhoGEF (LARG), which also contains RGS domain, with Galpha12 and Galpha13. RGS domain of LARG demonstrated Galpha12- and Galpha13-specific GAP activity. LARG synergistically stimulated SRF activation by Galpha12 and Galpha13 in HeLa cells, and the SRF activation by Galpha12-LARG was further stimulated by coexpression of Tec tyrosine kinase. It was also found that LARG is phosphorylated on tyrosine by Tec. In reconstitution assays, the RhoGEF activity of nonphosphorylated LARG was stimulated by Galpha13 but not Galpha12. However, when LARG was phosphorylated by Tec, Galpha12 effectively stimulated the RhoGEF activity of LARG. These results demonstrate the biochemical mechanism of Rho activation through Galpha12 and that the regulation of RhoGEFs by heterotrimeric G proteins G1213 is further modulated by tyrosine phosphorylation.


Subject(s)
Guanine Nucleotide Exchange Factors/physiology , Heterotrimeric GTP-Binding Proteins/physiology , Tyrosine/metabolism , rho GTP-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Enzyme Activation , GTP-Binding Protein alpha Subunits, G12-G13 , HeLa Cells , Humans , Phosphorylation , Protein-Tyrosine Kinases/physiology , Rho Guanine Nucleotide Exchange Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...