1
|
Lane BS, Heller B, Hollenberg MD, Wells CD. The RGS-RhoGEFs control the amplitude of YAP1 activation by serum. Sci Rep 2021; 11:2348. [PMID: 33504879 PMCID: PMC7841162 DOI: 10.1038/s41598-021-82027-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/14/2021] [Indexed: 12/16/2022] Open
Abstract
Actin-dependent mechanisms drive the nuclear translocation of Yap1 to enable its co-activation of transcription factors that induce pro-growth and survival programs. While Rho GTPases are necessary for the nuclear import of YAP1, the relevant Guanine Exchange Factors (GEFs) and GTPase Activating Proteins (GAPs) that connect this process to upstream signaling are not well defined. To this end, we measured the impact of expressing sixty-seven RhoGEFs and RhoGAPs on the YAP1 dependent activity of a TEAD element transcriptional reporter. Robust effects by all three members of the regulator of G-protein signaling (RGS) domain containing RhoGEFs (ArhGEF1, ArhGEF11 and ArhGEF12) prompted studies relating their known roles in serum signaling onto the regulation of Yap1. Under all conditions examined, ArhGEF12 preferentially mediated the activation of YAP1/TEAD by serum versus ArhGEF1 or ArhGEF11. Conversely, ArhGEF1 in multiple contexts inhibited both basal and serum elevated YAP1 activity through its GAP activity for Gα13. The sensitivity of such inhibition to cellular density and to low states of serum signaling supports that ArhGEF1 is a context dependent regulator of YAP1. Taken together, the relative activities of the RGS-RhoGEFs were found to dictate the degree to which serum signaling promotes YAP1 activity.
Collapse
Affiliation(s)
- Brandon S Lane
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Brigitte Heller
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Clark D Wells
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Indiana University School of Medicine, John D. Van Nuys Medical Science Building. 635 Barnhill Dr., Rm. 4079A, Indianapolis, IN, USA.
| |
Collapse
|
2
|
Maziarz M, Federico A, Zhao J, Dujmusic L, Zhao Z, Monti S, Varelas X, Garcia-Marcos M. Naturally occurring hotspot cancer mutations in Gα 13 promote oncogenic signaling. J Biol Chem 2020; 295:16897-16904. [PMID: 33109615 PMCID: PMC7864081 DOI: 10.1074/jbc.ac120.014698] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/07/2020] [Indexed: 12/15/2022] Open
Abstract
Heterotrimeric G-proteins are signaling switches broadly divided into four families based on the sequence and functional similarity of their Gα subunits: Gs, Gi/o, Gq/11, and G12/13 Artificial mutations that activate Gα subunits of each of these families have long been known to induce oncogenic transformation in experimental systems. With the advent of next-generation sequencing, activating hotspot mutations in Gs, Gi/o, or Gq/11 proteins have also been identified in patient tumor samples. In contrast, patient tumor-associated G12/13 mutations characterized to date lead to inactivation rather than activation. By using bioinformatic pathway analysis and signaling assays, here we identified cancer-associated hotspot mutations in Arg-200 of Gα13 (encoded by GNA13) as potent activators of oncogenic signaling. First, we found that components of a G12/13-dependent signaling cascade that culminates in activation of the Hippo pathway effectors YAP and TAZ is frequently altered in bladder cancer. Up-regulation of this signaling cascade correlates with increased YAP/TAZ activation transcriptional signatures in this cancer type. Among the G12/13 pathway alterations were mutations in Arg-200 of Gα13, which we validated to promote YAP/TAZ-dependent (TEAD) and MRTF-A/B-dependent (SRE.L) transcriptional activity. We further showed that this mechanism relies on the same RhoGEF-RhoGTPase cascade components that are up-regulated in bladder cancers. Moreover, Gα13 Arg-200 mutants induced oncogenic transformation in vitro as determined by focus formation assays. In summary, our findings on Gα13 mutants establish that naturally occurring hotspot mutations in Gα subunits of any of the four families of heterotrimeric G-proteins are putative cancer drivers.
Collapse
Affiliation(s)
- Marcin Maziarz
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Anthony Federico
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jingyi Zhao
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Lorena Dujmusic
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Zhiming Zhao
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Stefano Monti
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Mikel Garcia-Marcos
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA.
| |
Collapse
|
3
|
Histamine-induced biphasic activation of RhoA allows for persistent RhoA signaling. PLoS Biol 2020; 18:e3000866. [PMID: 32881857 PMCID: PMC7494096 DOI: 10.1371/journal.pbio.3000866] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 09/16/2020] [Accepted: 08/12/2020] [Indexed: 12/30/2022] Open
Abstract
The small GTPase RhoA is a central signaling enzyme that is involved in various cellular processes such as cytoskeletal dynamics, transcription, and cell cycle progression. Many signal transduction pathways activate RhoA—for instance, Gαq-coupled Histamine 1 Receptor signaling via Gαq-dependent activation of RhoGEFs such as p63. Although multiple upstream regulators of RhoA have been identified, the temporal regulation of RhoA and the coordination of different upstream components in its regulation have not been well characterized. In this study, live-cell measurement of RhoA activation revealed a biphasic increase of RhoA activity upon histamine stimulation. We showed that the first and second phase of RhoA activity are dependent on p63 and Ca2+/PKC, respectively, and further identified phosphorylation of serine 240 on p115 RhoGEF by PKC to be the mechanistic link between PKC and RhoA. Combined approaches of computational modeling and quantitative measurement revealed that the second phase of RhoA activation is insensitive to rapid turning off of the receptor and is required for maintaining RhoA-mediated transcription after the termination of the receptor signaling. Thus, two divergent pathways enable both rapid activation and persistent signaling in receptor-mediated RhoA signaling via intricate temporal regulation. The small GTPase RhoA is a central signaling enzyme that is involved in various cellular processes such as cytoskeletal dynamics, transcription, and cell cycle progression. This study shows that histamine induces biphasic activation of RhoA via two divergent signaling pathways, allowing for intricate regulation of cellular processes.
Collapse
|
4
|
Maziarz M, Park JC, Leyme A, Marivin A, Garcia-Lopez A, Patel PP, Garcia-Marcos M. Revealing the Activity of Trimeric G-proteins in Live Cells with a Versatile Biosensor Design. Cell 2020; 182:770-785.e16. [PMID: 32634377 DOI: 10.1016/j.cell.2020.06.020] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/21/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022]
Abstract
Heterotrimeric G-proteins (Gαβγ) are the main transducers of signals from GPCRs, mediating the action of countless natural stimuli and therapeutic agents. However, there are currently no robust approaches to directly measure the activity of endogenous G-proteins in cells. Here, we describe a suite of optical biosensors that detect endogenous active G-proteins with sub-second resolution in live cells. Using a modular design principle, we developed genetically encoded, unimolecular biosensors for endogenous Gα-GTP and free Gβγ: the two active species of heterotrimeric G-proteins. This design was leveraged to generate biosensors with specificity for different heterotrimeric G-proteins or for other G-proteins, such as Rho GTPases. Versatility was further validated by implementing the biosensors in multiple contexts, from characterizing cancer-associated G-protein mutants to neurotransmitter signaling in primary neurons. Overall, the versatile biosensor design introduced here enables studying the activity of endogenous G-proteins in live cells with high fidelity, temporal resolution, and convenience.
Collapse
Affiliation(s)
- Marcin Maziarz
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Jong-Chan Park
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Anthony Leyme
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Arthur Marivin
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Alberto Garcia-Lopez
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Prachi P Patel
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Mikel Garcia-Marcos
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
5
|
Müller PM, Rademacher J, Bagshaw RD, Wortmann C, Barth C, van Unen J, Alp KM, Giudice G, Eccles RL, Heinrich LE, Pascual-Vargas P, Sanchez-Castro M, Brandenburg L, Mbamalu G, Tucholska M, Spatt L, Czajkowski MT, Welke RW, Zhang S, Nguyen V, Rrustemi T, Trnka P, Freitag K, Larsen B, Popp O, Mertins P, Gingras AC, Roth FP, Colwill K, Bakal C, Pertz O, Pawson T, Petsalaki E, Rocks O. Systems analysis of RhoGEF and RhoGAP regulatory proteins reveals spatially organized RAC1 signalling from integrin adhesions. Nat Cell Biol 2020; 22:498-511. [PMID: 32203420 DOI: 10.1038/s41556-020-0488-x] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/18/2020] [Indexed: 02/07/2023]
Abstract
Rho GTPases are central regulators of the cytoskeleton and, in humans, are controlled by 145 multidomain guanine nucleotide exchange factors (RhoGEFs) and GTPase-activating proteins (RhoGAPs). How Rho signalling patterns are established in dynamic cell spaces to control cellular morphogenesis is unclear. Through a family-wide characterization of substrate specificities, interactomes and localization, we reveal at the systems level how RhoGEFs and RhoGAPs contextualize and spatiotemporally control Rho signalling. These proteins are widely autoinhibited to allow local regulation, form complexes to jointly coordinate their networks and provide positional information for signalling. RhoGAPs are more promiscuous than RhoGEFs to confine Rho activity gradients. Our resource enabled us to uncover a multi-RhoGEF complex downstream of G-protein-coupled receptors controlling CDC42-RHOA crosstalk. Moreover, we show that integrin adhesions spatially segregate GEFs and GAPs to shape RAC1 activity zones in response to mechanical cues. This mechanism controls the protrusion and contraction dynamics fundamental to cell motility. Our systems analysis of Rho regulators is key to revealing emergent organization principles of Rho signalling.
Collapse
Affiliation(s)
- Paul M Müller
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | - Richard D Bagshaw
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | | | - Carolin Barth
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Jakobus van Unen
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Keziban M Alp
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Girolamo Giudice
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Louise E Heinrich
- Institute of Cancer Research, Chester Beatty Laboratories, London, UK
| | | | - Marta Sanchez-Castro
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | | | - Geraldine Mbamalu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Monika Tucholska
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Lisa Spatt
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Maciej T Czajkowski
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | | | - Sunqu Zhang
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Vivian Nguyen
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | | | - Philipp Trnka
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Kiara Freitag
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Brett Larsen
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Oliver Popp
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Philipp Mertins
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Frederick P Roth
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Donnelly Centre and Departments of Molecular Genetics and Computer Science, University of Toronto, Toronto, Ontario, Canada
- Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Chris Bakal
- Institute of Cancer Research, Chester Beatty Laboratories, London, UK
| | - Olivier Pertz
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Tony Pawson
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Evangelia Petsalaki
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, UK.
| | - Oliver Rocks
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Wauson EM, Guerra ML, Dyachok J, McGlynn K, Giles J, Ross EM, Cobb MH. Differential Regulation of ERK1/2 and mTORC1 Through T1R1/T1R3 in MIN6 Cells. Mol Endocrinol 2015; 29:1114-22. [PMID: 26168033 DOI: 10.1210/me.2014-1181] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The MAPKs ERK1/2 respond to nutrients and other insulin secretagogues in pancreatic β-cells and mediate nutrient-dependent insulin gene transcription. Nutrients also stimulate the mechanistic target of rapamycin complex 1 (mTORC1) to regulate protein synthesis. We showed previously that activation of both ERK1/2 and mTORC1 in the MIN6 pancreatic β-cell-derived line by extracellular amino acids (AAs) is at least in part mediated by the heterodimeric T1R1/T1R3, a G protein-coupled receptor. We show here that AAs differentially activate these two signaling pathways in MIN6 cells. Pretreatment with pertussis toxin did not prevent the activation of either ERK1/2 or mTORC1 by AAs, indicating that G(I) is not central to either pathway. Although glucagon-like peptide 1, an agonist for a G(s-)coupled receptor, activated ERK1/2 well and mTORC1 to a small extent, AAs had no effect on cytosolic cAMP accumulation. Ca(2+) entry is required for ERK1/2 activation by AAs but is dispensable for AA activation of mTORC1. Pretreatment with UBO-QIC, a selective G(q) inhibitor, reduced the activation of ERK1/2 but had little effect on the activation of mTORC1 by AAs, suggesting a differential requirement for G(q). Inhibition of G(12/13) by the overexpression of the regulator of G protein signaling domain of p115 ρ-guanine nucleotide exchange factor had no effect on mTORC1 activation by AAs, suggesting that these G proteins are also not involved. We conclude that AAs regulate ERK1/2 and mTORC1 through distinct signaling pathways.
Collapse
Affiliation(s)
- Eric M Wauson
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Marcy L Guerra
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Julia Dyachok
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Kathleen McGlynn
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Jennifer Giles
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Elliott M Ross
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Melanie H Cobb
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| |
Collapse
|
7
|
Abstract
The RhoGEF (Rho GTPase guanine-nucleotide-exchange factor) domain of AKAP-Lbc (A-kinase-anchoring protein-Lbc, also known as AKAP13) catalyses nucleotide exchange on RhoA and is involved in the development of cardiac hypertrophy. The RhoGEF activity of AKAP-Lbc has also been implicated in cancer. We have determined the X-ray crystal structure of the complex between RhoA–GDP and the AKAP-Lbc RhoGEF [DH (Dbl-homologous)–PH (pleckstrin homology)] domain to 2.1 Å (1 Å=0.1 nm) resolution. The structure reveals important differences compared with related RhoGEF proteins such as leukaemia-associated RhoGEF. Nucleotide-exchange assays comparing the activity of the DH–PH domain to the DH domain alone showed no role for the PH domain in nucleotide exchange, which is explained by the RhoA–AKAP-Lbc structure. Comparison with a structure of the isolated AKAP-Lbc DH domain revealed a change in conformation of the N-terminal ‘GEF switch’ region upon binding to RhoA. Isothermal titration calorimetry showed that AKAP-Lbc has only micromolar affinity for RhoA, which combined with the presence of potential binding pockets for small molecules on AKAP-Lbc, raises the possibility of targeting AKAP-Lbc with GEF inhibitors. The crystal structure of the RhoGEF domain of AKAP-Lbc in complex with RhoA combined with nucleotide exchange assays explain differences to related RhoGEF proteins and allow the possibility of targeting the AKAP-Lbc RhoGEF domain with small molecules.
Collapse
|
8
|
Lenoir M, Sugawara M, Kaur J, Ball LJ, Overduin M. Structural insights into the activation of the RhoA GTPase by the lymphoid blast crisis (Lbc) oncoprotein. J Biol Chem 2014; 289:23992-4004. [PMID: 24993829 PMCID: PMC4156082 DOI: 10.1074/jbc.m114.561787] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The small GTPase RhoA promotes deregulated signaling upon interaction with lymphoid blast crisis (Lbc), the oncogenic form of A-kinase anchoring protein 13 (AKAP13). The onco-Lbc protein is a hyperactive Rho-specific guanine nucleotide exchange factor (GEF), but its structural mechanism has not been reported despite its involvement in cardiac hypertrophy and cancer causation. The pleckstrin homology (PH) domain of Lbc is located at the C-terminal end of the protein and is shown here to specifically recognize activated RhoA rather than lipids. The isolated dbl homology (DH) domain can function as an independent activator with an enhanced activity. However, the DH domain normally does not act as a solitary Lbc interface with RhoA-GDP. Instead it is negatively controlled by the PH domain. In particular, the DH helical bundle is coupled to the structurally dependent PH domain through a helical linker, which reduces its activity. Together the two domains form a rigid scaffold in solution as evidenced by small angle x-ray scattering and 1H,13C,15N-based NMR spectroscopy. The two domains assume a “chair” shape with its back possessing independent GEF activity and the PH domain providing a broad seat for RhoA-GTP docking rather than membrane recognition. This provides structural and dynamical insights into how DH and PH domains work together in solution to support regulated RhoA activity. Mutational analysis supports the bifunctional PH domain mediation of DH-RhoA interactions and explains why the tandem domain is required for controlled GEF signaling.
Collapse
Affiliation(s)
- Marc Lenoir
- From the School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Masae Sugawara
- From the School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jaswant Kaur
- From the School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Linda J Ball
- Structural Genomics Consortium, University of Oxford, Oxford OX3 7DQ, United Kingdom, and The Leibniz Institute of Molecular Pharmacology, Campus Buch, 13125 Berlin, Germany
| | - Michael Overduin
- From the School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom,
| |
Collapse
|
9
|
Carter AM, Gutowski S, Sternweis PC. Regulated localization is sufficient for hormonal control of regulator of G protein signaling homology Rho guanine nucleotide exchange factors (RH-RhoGEFs). J Biol Chem 2014; 289:19737-46. [PMID: 24855647 DOI: 10.1074/jbc.m114.564930] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The regulator of G protein signaling homology (RH) Rho guanine nucleotide exchange factors (RhoGEFs) (p115RhoGEF, leukemia-associated RhoGEF, and PDZ-RhoGEF) contain an RH domain and are specific GEFs for the monomeric GTPase RhoA. The RH domains interact specifically with the α subunits of G12 heterotrimeric GTPases. Activated Gα13 modestly stimulates the exchange activity of both p115RhoGEF and leukemia-associated RhoGEF but not PDZ-RhoGEF. Because all three RH-RhoGEFs can localize to the plasma membrane upon expression of activated Gα13, cellular localization of these RhoGEFs has been proposed as a mechanism for controlling their activity. We use a small molecule-regulated heterodimerization system to rapidly control the localization of RH-RhoGEFs. Acute localization of the proteins to the plasma membrane activates RhoA within minutes and to levels that are comparable with activation of RhoA by hormonal stimulation of G protein-coupled receptors. The catalytic activity of membrane-localized RhoGEFs is not dependent on activated Gα13. We further show that the conserved RH domains can rewire two different RacGEFs to activate Rac1 in response to a traditional activator of RhoA. Thus, RH domains act as independent detectors for activated Gα13 and are sufficient to modulate the activity of RhoGEFs by hormones via mediating their localization to substrate, membrane-associated RhoA.
Collapse
Affiliation(s)
- Angela M Carter
- From the Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - Stephen Gutowski
- From the Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| | - Paul C Sternweis
- From the Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390
| |
Collapse
|
10
|
Medina F, Carter AM, Dada O, Gutowski S, Hadas J, Chen Z, Sternweis PC. Activated RhoA is a positive feedback regulator of the Lbc family of Rho guanine nucleotide exchange factor proteins. J Biol Chem 2013; 288:11325-33. [PMID: 23493395 DOI: 10.1074/jbc.m113.450056] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The monomeric Rho GTPases are essential for cellular regulation including cell architecture and movement. A direct mechanism for hormonal regulation of the RhoA-type GTPases is their modulation by the G12 and G13 proteins via RH (RGS homology) containing RhoGEFs. In addition to the interaction of the G protein α subunits with the RH domain, activated RhoA also binds to the pleckstrin homology (PH) domain of PDZRhoGEF. The latter interaction is now extended to all seven members of the homologous Lbc family of RhoGEFs which includes the RH-RhoGEFs. This is evinced by direct measurements of binding or through effects on selected signaling pathways in cells. Overexpression of these PH domains alone can block RhoA-dependent signaling in cells to various extents. Whereas activated RhoA does not modulate the intrinsic activity of the RhoGEFs, activated RhoA associated with phospholipid vesicles can facilitate increased activity of soluble RhoGEFs on vesicle-delimited substrate (RhoA-GDP). This demonstrates feasibility of the hypothesis that binding of activated RhoA to the PH domains acts as a positive feedback mechanism. This is supported by cellular studies in which mutation of this binding site on PH strongly attenuates the stimulation of RhoA observed by overexpression of five of the RhoGEF DH-PH domains. This mutation is even more dramatic in the context of full-length p115RhoGEF. The utilization of this mechanism by multiple RhoGEFs suggests that this regulatory paradigm may be a common feature in the broader family of RhoGEFs.
Collapse
Affiliation(s)
- Frank Medina
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Jaiswal M, Dvorsky R, Ahmadian MR. Deciphering the molecular and functional basis of Dbl family proteins: a novel systematic approach toward classification of selective activation of the Rho family proteins. J Biol Chem 2012; 288:4486-500. [PMID: 23255595 DOI: 10.1074/jbc.m112.429746] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The diffuse B-cell lymphoma (Dbl) family of the guanine nucleotide exchange factors is a direct activator of the Rho family proteins. The Rho family proteins are involved in almost every cellular process that ranges from fundamental (e.g. the establishment of cell polarity) to highly specialized processes (e.g. the contraction of vascular smooth muscle cells). Abnormal activation of the Rho proteins is known to play a crucial role in cancer, infectious and cognitive disorders, and cardiovascular diseases. However, the existence of 74 Dbl proteins and 25 Rho-related proteins in humans, which are largely uncharacterized, has led to increasing complexity in identifying specific upstream pathways. Thus, we comprehensively investigated sequence-structure-function-property relationships of 21 representatives of the Dbl protein family regarding their specificities and activities toward 12 Rho family proteins. The meta-analysis approach provides an unprecedented opportunity to broadly profile functional properties of Dbl family proteins, including catalytic efficiency, substrate selectivity, and signaling specificity. Our analysis has provided novel insights into the following: (i) understanding of the relative differences of various Rho protein members in nucleotide exchange; (ii) comparing and defining individual and overall guanine nucleotide exchange factor activities of a large representative set of the Dbl proteins toward 12 Rho proteins; (iii) grouping the Dbl family into functionally distinct categories based on both their catalytic efficiencies and their sequence-structural relationships; (iv) identifying conserved amino acids as fingerprints of the Dbl and Rho protein interaction; and (v) defining amino acid sequences conserved within, but not between, Dbl subfamilies. Therefore, the characteristics of such specificity-determining residues identified the regions or clusters conserved within the Dbl subfamilies.
Collapse
Affiliation(s)
- Mamta Jaiswal
- Institut für Biochemie and Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | | | | |
Collapse
|
12
|
Viaud J, Gaits-Iacovoni F, Payrastre B. Regulation of the DH-PH tandem of guanine nucleotide exchange factor for Rho GTPases by phosphoinositides. Adv Biol Regul 2012; 52:303-14. [PMID: 22781744 DOI: 10.1016/j.jbior.2012.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 04/04/2012] [Indexed: 10/28/2022]
Abstract
Rho GTPases act as molecular switches central in cellular processes such as cytoskeleton dynamics, migration, cell proliferation, growth or survival. Their activation is tightly regulated downstream of cell surface receptors by Guanine nucleotide Exchange Factors (GEFs), that are responsible for the specificity, the accuracy, and the spatial restriction of Rho GTPases response to extracellular cues. Because there is about four time more RhoGEFs that Rho GTPases, and GEFs do not always show a strict specificity for GTPases, it is clear that their regulation depends on specific interactions with the subcellular environment. RhoGEFs bear a peculiar structure, highly conserved though evolution, consisting of a DH-PH tandem, the DH (Dbl homology) domain being responsible for the exchange activity. The function of the PH (Pleckstrin homology) domain known to bind phosphoinositides, however, remains elusive, and reports are in many cases rather confusing. This review summarizes data on the regulation of RhoGEFs activity through interaction of the PH-associated DH domain with phosphoinositides which are considered as critical players in the spatial organization of major signaling pathways.
Collapse
Affiliation(s)
- Julien Viaud
- INSERM, UMR1048, Université Paul Sabatier, Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, 1 Avenue Jean Poulhès, BP 84225, 31432 Toulouse Cedex 4, France
| | | | | |
Collapse
|
13
|
Pfreimer M, Vatter P, Langer T, Wieland T, Gierschik P, Moepps B. LARG links histamine-H1-receptor-activated Gq to Rho-GTPase-dependent signaling pathways. Cell Signal 2011; 24:652-63. [PMID: 22100544 DOI: 10.1016/j.cellsig.2011.10.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/25/2011] [Accepted: 10/27/2011] [Indexed: 12/11/2022]
Abstract
Activation of heterotrimeric G proteins, such as G(12/13) and G(q), by cell surface receptors is coupled to the regulation of numerous cellular functions controlled by activated Rho GTPases. Previous studies have implicated the Rho guanine nucleotide exchange factor (RhoGEF) leukemia-associated RhoGEF (LARG) as a regulatory protein receiving stimulatory inputs from activated Gα(12/13) and Gα(q). However, the molecular mechanisms of the Gα(q)-mediated LARG activation are not fully understood and the structural elements of LARG involved in this process have remained unclear. In the present work, the specific coupling of the histamine H1 receptor (HRH1) exogenously expressed in COS-7 cells to G(q), but not to G(12/13), was used to conduct a detailed analysis of receptor- and Gα(q)-mediated LARG activation and to define its structural requirements. The results show that HRH1-mediated activation of the strictly Rho-dependent transcriptional activity of serum response factor requires the PDZ domain of LARG and can be mimicked by activated Gα(q)(Q209L). The functional interaction between activated Gα(q) and LARG requires no more than the catalytic DH-PH tandem of LARG, and is independent of PLCβ activation and distinct from the mechanisms of Gα(q)-mediated p63RhoGEF and PLCβ(3) activation. Activated Gα(q) physically interacts with the relevant portions of LARG in COS-7 cells and histamine causes activation of LARG in native HeLa cells endogenously expressing HRH1, G(q), and LARG. This work is the first positive demonstration of a stimulatory effect of LARG on the ability of a strictly G(q)-coupled receptor to cause activation of a Rho-GTPase-dependent signaling pathway.
Collapse
Affiliation(s)
- Mariana Pfreimer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | | | | | | | | | | |
Collapse
|
14
|
Bai Y, Luo Y, Liu S, Zhang L, Shen K, Dong Y, Walls CD, Quilliam LA, Wells CD, Cao Y, Zhang ZY. PRL-1 protein promotes ERK1/2 and RhoA protein activation through a non-canonical interaction with the Src homology 3 domain of p115 Rho GTPase-activating protein. J Biol Chem 2011; 286:42316-42324. [PMID: 22009749 DOI: 10.1074/jbc.m111.286302] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Phosphatases of the regenerating liver (PRL) play oncogenic roles in cancer development and metastasis. Although previous studies indicate that PRL-1 promotes cell growth and migration by activating both the ERK1/2 and RhoA pathways, the mechanism by which it activates these signaling events remains unclear. We have identified a PRL-1-binding peptide (Peptide 1) that shares high sequence identity with a conserved motif in the Src homology 3 (SH3) domain of p115 Rho GTPase-activating protein (GAP). p115 RhoGAP directly binds PRL-1 in vitro and in cells via its SH3 domain. Structural analyses of the PRL-1·Peptide 1 complex revealed a novel protein-protein interaction whereby a sequence motif within the PxxP ligand-binding site of the p115 RhoGAP SH3 domain occupies a folded groove within PRL-1. This prevents the canonical interaction between the SH3 domain of p115 RhoGAP and MEKK1 and results in activation of ERK1/2. Furthermore, PRL-1 binding activates RhoA signaling by inhibiting the catalytic activity of p115 RhoGAP. The results demonstrate that PRL-1 binding to p115 RhoGAP provides a coordinated mechanism underlying ERK1/2 and RhoA activation.
Collapse
Affiliation(s)
- Yunpeng Bai
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Yong Luo
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Sijiu Liu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Lujuan Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Kui Shen
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, Illinois 60115
| | - Yuanshu Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Chad D Walls
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Lawrence A Quilliam
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Clark D Wells
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Youjia Cao
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202.
| |
Collapse
|
15
|
Kozasa T, Hajicek N, Chow CR, Suzuki N. Signalling mechanisms of RhoGTPase regulation by the heterotrimeric G proteins G12 and G13. J Biochem 2011; 150:357-69. [PMID: 21873336 DOI: 10.1093/jb/mvr105] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
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.
Collapse
Affiliation(s)
- Tohru Kozasa
- Laboratory of Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-8904, Japan.
| | | | | | | |
Collapse
|
16
|
de Godoy MAF, Rattan S. Role of rho kinase in the functional and dysfunctional tonic smooth muscles. Trends Pharmacol Sci 2011; 32:384-93. [PMID: 21497405 PMCID: PMC3128689 DOI: 10.1016/j.tips.2011.03.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 03/05/2011] [Accepted: 03/11/2011] [Indexed: 01/13/2023]
Abstract
Tonic smooth muscles play pivotal roles in the pathophysiology of debilitating diseases of the gastrointestinal and cardiovascular systems. Tonic smooth muscles differ from phasic smooth muscles in the ability to spontaneously develop myogenic tone. This ability has been primarily attributed to the local production of specific neurohumoral substances that can work in conjunction with calcium sensitization via signal transduction events associated with the Ras homolog gene family, member A (RhoA)/Rho-associated, coiled-coil containing protein kinase 2 (ROCK II) pathways. In this article, we discuss the molecular pathways involved in the myogenic properties of tonic smooth muscles, particularly the contribution of protein kinase C vs the RhoA/ROCK II pathway in the genesis of basal tone, pathophysiology and novel therapeutic approaches for certain gastrointestinal and cardiovascular diseases. Emerging evidence suggests that manipulation of RhoA/ROCK II activity through inhibitors or silencing of RNA interface techniques could represent a new therapeutic approach for various gastrointestinal and cardiovascular diseases.
Collapse
Affiliation(s)
- Marcio Augusto Fressatto de Godoy
- Department of Medicine, Division of Gastroenterology & Hepatology, Jefferson Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Satish Rattan
- Department of Medicine, Division of Gastroenterology & Hepatology, Jefferson Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Hajicek N, Kukimoto-Niino M, Mishima-Tsumagari C, Chow CR, Shirouzu M, Terada T, Patel M, Yokoyama S, Kozasa T. Identification of critical residues in G(alpha)13 for stimulation of p115RhoGEF activity and the structure of the G(alpha)13-p115RhoGEF regulator of G protein signaling homology (RH) domain complex. J Biol Chem 2011; 286:20625-36. [PMID: 21507947 PMCID: PMC3121507 DOI: 10.1074/jbc.m110.201392] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 04/08/2011] [Indexed: 01/28/2023] Open
Abstract
RH-RhoGEFs are a family of guanine nucleotide exchange factors that contain a regulator of G protein signaling homology (RH) domain. The heterotrimeric G protein Gα(13) stimulates the guanine nucleotide exchange factor (GEF) activity of RH-RhoGEFs, leading to activation of RhoA. The mechanism by which Gα(13) stimulates the GEF activity of RH-RhoGEFs, such as p115RhoGEF, has not yet been fully elucidated. Here, specific residues in Gα(13) that mediate activation of p115RhoGEF are identified. Mutation of these residues significantly impairs binding of Gα(13) to p115RhoGEF as well as stimulation of GEF activity. These data suggest that the exchange activity of p115RhoGEF is stimulated allosterically by Gα(13) and not through its interaction with a secondary binding site. A crystal structure of Gα(13) bound to the RH domain of p115RhoGEF is also presented, which differs from a previously crystallized complex with a Gα(13)-Gα(i1) chimera. Taken together, these data provide new insight into the mechanism by which p115RhoGEF is activated by Gα(13).
Collapse
Affiliation(s)
- Nicole Hajicek
- Department of Pharmacology, College of Medicine, University of Illinois, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Jaiswal M, Gremer L, Dvorsky R, Haeusler LC, Cirstea IC, Uhlenbrock K, Ahmadian MR. Mechanistic insights into specificity, activity, and regulatory elements of the regulator of G-protein signaling (RGS)-containing Rho-specific guanine nucleotide exchange factors (GEFs) p115, PDZ-RhoGEF (PRG), and leukemia-associated RhoGEF (LARG). J Biol Chem 2011; 286:18202-12. [PMID: 21454492 DOI: 10.1074/jbc.m111.226431] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The multimodular guanine nucleotide exchange factors (GEFs) of the Dbl family mostly share a tandem Dbl homology (DH) and pleckstrin homology (PH) domain organization. The function of these and other domains in the DH-mediated regulation of the GDP/GTP exchange reaction of the Rho proteins is the subject of intensive investigations. This comparative study presents detailed kinetic data on specificity, activity, and regulation of the catalytic DH domains of four GEFs, namely p115, p190, PDZ-RhoGEF (PRG), and leukemia-associated RhoGEF (LARG). We demonstrate that (i) these GEFs are specific guanine nucleotide exchange factors for the Rho isoforms (RhoA, RhoB, and RhoC) and inactive toward other members of the Rho family, including Rac1, Cdc42, and TC10. (ii) The DH domain of LARG exhibits the highest catalytic activity reported for a Dbl protein till now with a maximal acceleration of the nucleotide exchange by 10(7)-fold, which is at least as efficient as reported for GEFs specific for Ran or the bacterial toxin SopE. (iii) A novel regulatory region at the N terminus of the DH domain is involved in its association with GDP-bound RhoA monitored by a fluorescently labeled RhoA. (iv) The tandem PH domains of p115 and PRG efficiently contribute to the DH-mediated nucleotide exchange reaction. (v) In contrast to the isolated DH or DH-PH domains, a p115 fragment encompassing both the regulator of G-protein signaling and the DH domains revealed a significantly reduced GEF activity, supporting the proposed models of an intramolecular autoinhibitory mechanism for p115-like RhoGEFs.
Collapse
Affiliation(s)
- Mamta Jaiswal
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
19
|
Xiaolu D, Jing P, Fang H, Lifen Y, Liwen W, Ciliu Z, Fei Y. Role of p115RhoGEF in lipopolysaccharide-induced mouse brain microvascular endothelial barrier dysfunction. Brain Res 2011; 1387:1-7. [PMID: 21354111 DOI: 10.1016/j.brainres.2011.02.059] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 02/15/2011] [Accepted: 02/18/2011] [Indexed: 12/24/2022]
Abstract
BACKGROUND In endothelial cells, exposure to lipopolysaccharide (LPS) results in barrier dysfunction through a complex signaling mechanism. The RhoA/Rho-kinase pathway plays a significant role in endothelial cell permeability. p115RhoGEF, a specific guanine nucleotide exchange factors (GEFs) activates RhoA, triggering RhoA-dependent cytoskeletal remodeling. However, little is known about the role of p115RhoGEF in LPS-induced brain endothelial barrier breakdown. We hypothesized that suppression of p115RhoGEF may inhibit activation of RhoA and prevent LPS-induced brain microvascular endothelial cell hyperpermeability. METHODS The cultured monolayer of bEnd.3 cells, an immortalized mouse brain endothelial cell line, was used in this study. bEnd.3 cells were pretreated with specific siRNA to knockdown p115RhoGEF or C3 transferase to inhibit RhoA activity, and then incubated with LPS (5μg/ml). The degree of RhoA activation was determined by a Rhotekin-based pull-down assay, and expression of p115RhoGEF, zonula occludens-1 (ZO-1), occludin and claudin-5 proteins were detected by Western blot analysis. The barrier function was measured by transendothelial electrical resistance (TEER). F-actin cytoskeleton was visualized by Rhodamine-phalloidin staining. RESULTS The expression level of p115RhoGEF protein was significantly increased in LPS-treated bEnd.3 cells. The activity of RhoA was enhanced after LPS stimulation and pretreatment with p115RhoGEF siRNA or exoenzyme C3 transferase reduced RhoA activation significantly as shown by the pull-down assay. Furthermore, depletion of p115RhoGEF partially prevented the LPS-induced decrease in TEER, stress fiber formation and tight junction proteins degradation. CONCLUSIONS These results suggest that p115RhoGEF is important for LPS signaling to RhoA and LPS-induced endothelial barrier dysfunction, providing new insight into the function of RhoGEFs in inflammation.
Collapse
Affiliation(s)
- Deng Xiaolu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Kintscher C, Wuertenberger S, Eylenstein R, Uhlendorf T, Groemping Y. Autoinhibition of GEF activity in Intersectin 1 is mediated by the short SH3-DH domain linker. Protein Sci 2011; 19:2164-74. [PMID: 20842712 DOI: 10.1002/pro.500] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Intersectin 1L (ITSN1L) acts as a specific guanine nucleotide exchange factor (GEF) for the small guanine nucleotide binding protein Cdc42 via its C-terminal DH domain. Interestingly, constructs of ITSN1L that comprise additional domains, for instance the five SH3 domains amino-terminal of the DH domain, were shown to be inhibited in their exchange factor activity. Here, we investigate the inhibitory mechanism of ITSN1L in detail and identify a novel short amino acid motif which mediates autoinhibition. We found this motif to be located in the linker region between the SH3 domains and the DH domain, and we show that within this motif W1221 acts as key residue in establishing the inhibitory interaction. This assigns ITSN1L to a growing class of GEFs that are regulated by a short amino acid motif inhibiting GEF activity by an intramolecular interaction. Moreover, we quantify the interaction between the ITSN1L SH3 domains and the Cdc42 effector N-WASP using fluorescence anisotropy binding experiments. As the SH3 domains are not involved in autoinhibition, binding of N-WASP does not release inhibition of nucleotide exchange activity in kinetic experiments, in contrast to earlier observations.
Collapse
Affiliation(s)
- Carsten Kintscher
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Tuebingen D-72076, Germany
| | | | | | | | | |
Collapse
|
21
|
Chen Z, Guo L, Sprang SR, Sternweis PC. Modulation of a GEF switch: autoinhibition of the intrinsic guanine nucleotide exchange activity of p115-RhoGEF. Protein Sci 2011; 20:107-17. [PMID: 21064165 PMCID: PMC3047067 DOI: 10.1002/pro.542] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 10/28/2010] [Accepted: 10/29/2010] [Indexed: 12/13/2022]
Abstract
p115-RhoGEF (p115) belongs to the family of RGS-containing guanine nucleotide exchange factors for Rho GTPases (RGS-RhoGEFs) that are activated by G12 class heterotrimeric G protein α subunits. All RGS-RhoGEFs possess tandemly linked Dbl-homology (DH) and plekstrin-homology (PH) domains, which bind and catalyze the exchange of GDP for GTP on RhoA. We have identified that the linker region connecting the N-terminal RGS-homology (RH) domain and the DH domain inhibits the intrinsic guanine nucleotide exchange (GEF) activity of p115, and determined the crystal structures of the DH/PH domains in the presence or absence of the inhibitory linker region. An N-terminal extension of the canonical DH domain (the GEF switch), which is critical to GEF activity, is well folded in the crystal structure of DH/PH alone, but becomes disordered in the presence of the linker region. The linker region is completely disordered in the crystal structure and partially disordered in the molecular envelope calculated from measurements of small angle x-ray scattering (SAXS). It is possible that Gα subunits activate p115 in part by relieving autoinhibition imposed by the linker region.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Pharmacology, The University of Texas Southwestern Medical CenterDallas, Texas 75390
| | - Liang Guo
- BioCAT, Advanced Photon Source, Argonne National Laboratory9700 South Cass Avenue, Argonne, Illinois 60439
| | - Stephen R Sprang
- Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of MontanaMissoula, Montana 59812
| | - Paul C Sternweis
- Department of Pharmacology, The University of Texas Southwestern Medical CenterDallas, Texas 75390
| |
Collapse
|
22
|
Chen Z, Medina F, Liu MY, Thomas C, Sprang SR, Sternweis PC. Activated RhoA binds to the pleckstrin homology (PH) domain of PDZ-RhoGEF, a potential site for autoregulation. J Biol Chem 2010; 285:21070-81. [PMID: 20430886 PMCID: PMC2898337 DOI: 10.1074/jbc.m110.122549] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 04/15/2010] [Indexed: 01/24/2023] Open
Abstract
Guanine nucleotide exchange factors (GEFs) catalyze exchange of GDP for GTP by stabilizing the nucleotide-free state of the small GTPases through their Dbl homology/pleckstrin homology (DH.PH) domains. Unconventionally, PDZ-RhoGEF (PRG), a member of the RGS-RhoGEFs, binds tightly to both nucleotide-free and activated RhoA (RhoA.GTP). We have characterized the interaction between PRG and activated RhoA and determined the structure of the PRG-DH.PH-RhoA.GTPgammaS (guanosine 5'-O-[gamma-thio]triphosphate) complex. The interface bears striking similarity to a GTPase-effector interface and involves the switch regions in RhoA and a hydrophobic patch in PRG-PH that is conserved among all Lbc RhoGEFs. The two surfaces that bind activated and nucleotide-free RhoA on PRG-DH.PH do not overlap, and a ternary complex of PRG-DH.PH bound to both forms of RhoA can be isolated by size-exclusion chromatography. This novel interaction between activated RhoA and PH could play a key role in regulation of RhoGEF activity in vivo.
Collapse
Affiliation(s)
- Zhe Chen
- From the Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390 and
| | - Frank Medina
- From the Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390 and
| | - Mu-ya Liu
- From the Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390 and
| | - Celestine Thomas
- the Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812
| | - Stephen R. Sprang
- the Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812
| | - Paul C. Sternweis
- From the Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390 and
| |
Collapse
|
23
|
Shankaranarayanan A, Boguth CA, Lutz S, Vettel C, Uhlemann F, Aittaleb M, Wieland T, Tesmer JJG. Galpha q allosterically activates and relieves autoinhibition of p63RhoGEF. Cell Signal 2010; 22:1114-23. [PMID: 20214977 PMCID: PMC2862694 DOI: 10.1016/j.cellsig.2010.03.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 03/01/2010] [Indexed: 01/01/2023]
Abstract
Galpha(q) directly activates p63RhoGEF and closely related catalytic domains found in Trio and Kalirin, thereby linking G(q)-coupled receptors to the activation of RhoA. Although the crystal structure of G alpha(q) in complex with the catalytic domains of p63RhoGEF is available, the molecular mechanism of activation has not yet been defined. In this study, we show that membrane translocation does not appear to play a role in G alpha(q)-mediated activation of p63RhoGEF, as it does in some other RhoGEFs. G alpha(q) instead must act allosterically. We next identify specific structural elements in the PH domain that inhibit basal nucleotide exchange activity, and provide evidence that G alpha(q) overcomes this inhibition by altering the conformation of the alpha 6-alpha N linker that joins the DH and PH domains, a region that forms direct contacts with RhoA. We also identify residues in G alpha(q) that are important for the activation of p63RhoGEF and that contribute to G alpha subfamily selectivity, including a critical residue in the G alpha(q) C-terminal helix, and demonstrate the importance of these residues for RhoA activation in living cells.
Collapse
|
24
|
Aittaleb M, Gao G, Evelyn CR, Neubig RR, Tesmer JJG. A conserved hydrophobic surface of the LARG pleckstrin homology domain is critical for RhoA activation in cells. Cell Signal 2009; 21:1569-78. [PMID: 19560536 PMCID: PMC2735620 DOI: 10.1016/j.cellsig.2009.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 06/16/2009] [Indexed: 01/23/2023]
Abstract
Leukemia associated Rho guanine nucleotide exchange factor (LARG) activates RhoA in response to signals received by specific classes of cell surface receptors. The catalytic core of LARG is a Dbl homology (DH) domain whose activity is modulated by an adjacent pleckstrin homology (PH) domain. In this study, we used a transcriptional assay and confocal microscopy to examine the roles of several novel structural features of the LARG DH/PH domains, including a conserved and exposed hydrophobic patch on the PH domain that mediates protein-protein interactions in crystal structures of LARG and its close homolog PDZ-RhoGEF. Mutation of the hydrophobic patch has no effect on nucleotide exchange activity in vitro, but abolished the ability of LARG to activate RhoA and to induce stress fiber formation in cultured cells. The activity of these mutants could be rescued by fusion with exogenous membrane-targeting domains. However, because membrane recruitment by activated G alpha(13) subunits was not sufficient to rescue activity of a hydrophobic patch mutant, the LARG PH domain cannot solely contribute to membrane targeting. Instead, it seems likely that the domain is involved in regulatory interactions with other proteins near the membrane surface. We also show that the hydrophobic patch of the PH domain is likely important for the activity of all Lbc subfamily RhoGEFs.
Collapse
Affiliation(s)
- Mohamed Aittaleb
- Lie Sciences Institute, University of Michigan, Ann Arbor, MI 48109-2216, USA
| | - Guang Gao
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| | - Chris R. Evelyn
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109-5632, USA
| | - Richard R. Neubig
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109-5632, USA
| | - John J. G. Tesmer
- Lie Sciences Institute, University of Michigan, Ann Arbor, MI 48109-2216, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109-5632, USA
| |
Collapse
|
25
|
Krawetz R, Kelly GM. Coordinate Gα13 and Wnt6-β-catenin signaling in F9 embryonal carcinoma cells is required for primitive endoderm differentiation. Biochem Cell Biol 2009; 87:567-80. [DOI: 10.1139/o09-014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The mouse F9 embryonal carcinoma cell line is ideally suited to study the epithelial-to-mesenchymal transition accompanying the differentiation of primitive to parietal extraembryonic endoderm. In F9 cells, the application of exogenous agents including retinoic acid or activation of signal transduction cascades downstream of G-proteins triggers widespread changes in gene expression and leads to the formation of primitive endoderm. The epithelial-to-mesenchymal transition is completed and parietal endoderm develops as of result of increasing PKA activity in primitive endoderm cells. Expression of a constitutively active form of Gα13(Q226L) is sufficient to induce F9 cells into parietal endoderm and a model is emerging that a signaling axis linking G-protein signaling to RhoA and the ERM protein moesin is required for differentiation. In this study, we found that expression of either p115RhoGEF or a constitutively active, GTPase-deficient form of RhoA(L63) promoted primitive, but not parietal, endoderm formation. The overexpression of Gα13(Q226L) or p115RhoGEF, but not Rho(L63), caused β-catenin to translocate to the nucleus. Surprisingly, the stimulation of the Wnt-β-catenin pathway was accompanied by nuclear β-catenin and primitive endoderm formation, even when a dominant negative was used to block the signaling axis at the level of p115RhoGEF or when ROCK activity was inhibited using the pharmacological agent Y-27632. Together, results indicate that the coordinate signaling by two independent pathways, one involving canonical Wnt-β-catenin activation of target genes and the other with Gα13 signaling to ERM proteins to modulate cytoarchitectural changes, is required during the retinoic acid induced differentiation of F9 cells to primitive endoderm.
Collapse
Affiliation(s)
- Roman Krawetz
- Department of Biology, Molecular Genetics Unit, University of Western Ontario, London, ON N6A 5B7, Canada
- Child Health Research Institute, University of Western Ontario, London, ON N6A 5B7, Canada
| | - Gregory M. Kelly
- Department of Biology, Molecular Genetics Unit, University of Western Ontario, London, ON N6A 5B7, Canada
- Child Health Research Institute, University of Western Ontario, London, ON N6A 5B7, Canada
| |
Collapse
|
26
|
Bhattacharyya R, Banerjee J, Khalili K, Wedegaertner PB. Differences in Galpha12- and Galpha13-mediated plasma membrane recruitment of p115-RhoGEF. Cell Signal 2009; 21:996-1006. [PMID: 19249348 PMCID: PMC2674241 DOI: 10.1016/j.cellsig.2009.02.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Revised: 02/16/2009] [Accepted: 02/17/2009] [Indexed: 11/22/2022]
Abstract
Regulator of G protein signaling domain-containing Rho guanine-nucleotide exchange factors (RGS-RhoGEFs) directly links activated forms of the G12 family of heterotrimeric G protein alpha subunits to the small GTPase Rho. Stimulation of G(12/13)-coupled GPCRs or expression of constitutively activated forms of alpha(12) and alpha(13) has been shown to induce the translocation of the RGS-RhoGEF, p115-RhoGEF, from the cytoplasm to the plasma membrane (PM). However, little is known regarding the functional importance and mechanisms of this regulated PM recruitment, and thus PM recruitment of p115-RhoGEF is the focus of this report. A constitutively PM-localized mutant of p115-RhoGEF shows a much greater activity compared to wild type p115-RhoGEF in promoting Rho-dependent neurite retraction of NGF-differentiated PC12 cells, providing the first evidence that PM localization can activate p115-RhoGEF signaling. Next, we uncovered the unexpected finding that Rho is required for alpha(13)-induced PM translocation of p115-RhoGEF. However, inhibition of Rho did not prevent alpha(12)-induced PM translocation of p115-RhoGEF. Additional differences between alpha(13) and alpha(12) in promoting PM recruitment of p115-RhoGEF were revealed by analyzing RGS domain mutants of p115-RhoGEF. Activated alpha(12) effectively recruits the isolated RGS domain of p115-RhoGEF to the PM, whereas alpha(13) only weakly does. On the other hand, alpha(13) strongly recruits to the PM a p115-RhoGEF mutant containing amino acid substitutions in an acidic region at the N-terminus of the RGS domain; however, alpha(12) is unable to recruit this p115-RhoGEF mutant to the PM. These studies provide new insight into the function and mechanisms of alpha(12/13)-mediated PM recruitment of p115-RhoGEF.
Collapse
Affiliation(s)
- Raja Bhattacharyya
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadephia, PA 19122
| | - Jayashree Banerjee
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadephia, PA 19122
| | - Philip B. Wedegaertner
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
27
|
Suzuki N, Hajicek N, Kozasa T. Regulation and physiological functions of G12/13-mediated signaling pathways. Neurosignals 2009; 17:55-70. [PMID: 19212140 DOI: 10.1159/000186690] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 10/10/2008] [Indexed: 12/12/2022] Open
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.
Collapse
Affiliation(s)
- Nobuchika Suzuki
- Laboratory of Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.
| | | | | |
Collapse
|
28
|
Suzuki N, Tsumoto K, Hajicek N, Daigo K, Tokita R, Minami S, Kodama T, Hamakubo T, Kozasa T. Activation of leukemia-associated RhoGEF by Galpha13 with significant conformational rearrangements in the interface. J Biol Chem 2008; 284:5000-9. [PMID: 19074425 DOI: 10.1074/jbc.m804073200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
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).
Collapse
Affiliation(s)
- Nobuchika Suzuki
- Laboratory of Systems Biology and Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chen Z, Singer WD, Danesh SM, Sternweis PC, Sprang SR. Recognition of the activated states of Galpha13 by the rgRGS domain of PDZRhoGEF. Structure 2008; 16:1532-43. [PMID: 18940608 PMCID: PMC2586972 DOI: 10.1016/j.str.2008.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 07/29/2008] [Accepted: 07/29/2008] [Indexed: 12/13/2022]
Abstract
G12 class heterotrimeric G proteins stimulate RhoA activation by RGS-RhoGEFs. However, p115RhoGEF is a GTPase Activating Protein (GAP) toward Galpha13, whereas PDZRhoGEF is not. We have characterized the interaction between the PDZRhoGEF rgRGS domain (PRG-rgRGS) and the alpha subunit of G13 and have determined crystal structures of their complexes in both the inactive state bound to GDP and the active states bound to GDP*AlF (transition state) and GTPgammaS (Michaelis complex). PRG-rgRGS interacts extensively with the helical domain and the effector-binding sites on Galpha13 through contacts that are largely conserved in all three nucleotide-bound states, although PRG-rgRGS has highest affinity to the Michaelis complex. An acidic motif in the N terminus of PRG-rgRGS occupies the GAP binding site of Galpha13 and is flexible in the GDP*AlF complex but well ordered in the GTPgammaS complex. Replacement of key residues in this motif with their counterparts in p115RhoGEF confers GAP activity.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, Texas 75390
| | - William D. Singer
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, Texas 75390
| | - Shahab M. Danesh
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, Texas 75390
| | - Paul C. Sternweis
- Department of Pharmacology, The University of Texas Southwestern Medical Center, 6001 Forest Park Road, Dallas, Texas 75390
| | - Stephen R. Sprang
- Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, 32 Campus Drive, MS 1656, Missoula, Montana 59812
| |
Collapse
|
30
|
Hu J, Strauch P, Rubtsov A, Donovan EE, Pelanda R, Torres RM. Lsc activity is controlled by oligomerization and regulates integrin adhesion. Mol Immunol 2008; 45:1825-36. [PMID: 18157933 PMCID: PMC2315659 DOI: 10.1016/j.molimm.2007.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 11/01/2007] [Accepted: 11/04/2007] [Indexed: 01/20/2023]
Abstract
Lsc is a hematopoietic-restricted protein that functions as an effector of G alpha(12/13)-associated G-protein coupled receptors that activates RhoA. In the absence of Lsc leukocytes exhibit impaired migration and B lymphocytes inefficiently resolve integrin-mediated adhesion. Here, we demonstrate that Lsc exists physiologically in primary B lymphocytes as a large molecular weight complex resembling a homo-tetramer. Interfering with the assembly of this large molecular weight Lsc oligomer results in the activation of both Lsc functional activities and leads to cell rounding and inhibition of integrin-mediated adhesion. During cell migration on integrin ligands we find Lsc localizes predominantly toward the rear of migrating cells where we suggest it activates RhoA to resolve integin-mediated adhesion. Together these data demonstrate that Lsc regulates integrin-mediated adhesive events at the trailing edge of migrating cells.
Collapse
Affiliation(s)
- Jiancheng Hu
- Integrated Department of Immunology, University of Colorado Health Sciences Center and National Jewish Medical and Research Center, Denver, CO, 80206, USA
| | - Pamela Strauch
- Integrated Department of Immunology, University of Colorado Health Sciences Center and National Jewish Medical and Research Center, Denver, CO, 80206, USA
| | - Anatoly Rubtsov
- Integrated Department of Immunology, University of Colorado Health Sciences Center and National Jewish Medical and Research Center, Denver, CO, 80206, USA
| | - Erin E. Donovan
- Integrated Department of Immunology, University of Colorado Health Sciences Center and National Jewish Medical and Research Center, Denver, CO, 80206, USA
| | - Roberta Pelanda
- Integrated Department of Immunology, University of Colorado Health Sciences Center and National Jewish Medical and Research Center, Denver, CO, 80206, USA
| | - Raul M. Torres
- Integrated Department of Immunology, University of Colorado Health Sciences Center and National Jewish Medical and Research Center, Denver, CO, 80206, USA
| |
Collapse
|
31
|
Schulze H, Dose M, Korpal M, Meyer I, Italiano JE, Shivdasani RA. RanBP10 is a cytoplasmic guanine nucleotide exchange factor that modulates noncentrosomal microtubules. J Biol Chem 2008; 283:14109-19. [PMID: 18347012 DOI: 10.1074/jbc.m709397200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microtubule spindle assembly in mitosis is stimulated by Ran.GTP, which is generated along condensed chromosomes by the guanine nucleotide exchange factor (GEF) RCC1. This relationship suggests that similar activities might modulate other microtubule structures. Interphase microtubules usually extend from the centrosome, although noncentrosomal microtubules function in some differentiated cells, including megakaryocytes. In these cells, platelet biogenesis requires massive mobilization of microtubules in the cell periphery, where they form proplatelets, the immediate precursors of platelets, in the apparent absence of centrioles. Here we identify a cytoplasmic Ran-binding protein, RanBP10, as a factor that binds beta-tubulin and associates with megakaryocyte microtubules. Unexpectedly, RanBP10 harbors GEF activity toward Ran. A point mutation in the candidate GEF domain abolishes exchange activity, and our results implicate RanBP10 as a localized cytoplasmic Ran-GEF. RNA interference-mediated loss of RanBP10 in cultured megakaryocytes disrupts microtubule organization. These results lead us to propose that spatiotemporally restricted generation of cytoplasmic Ran.GTP may influence organization of the specialized microtubules required in thrombopoiesis and that RanBP10 might serve as a molecular link between Ran and noncentrosomal microtubules.
Collapse
|
32
|
Sprang SR, Chen Z, Du X. Structural basis of effector regulation and signal termination in heterotrimeric Galpha proteins. ADVANCES IN PROTEIN CHEMISTRY 2007; 74:1-65. [PMID: 17854654 DOI: 10.1016/s0065-3233(07)74001-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This chapter addresses, from a molecular structural perspective gained from examination of x-ray crystallographic and biochemical data, the mechanisms by which GTP-bound Galpha subunits of heterotrimeric G proteins recognize and regulate effectors. The mechanism of GTP hydrolysis by Galpha and rate acceleration by GAPs are also considered. The effector recognition site in all Galpha homologues is formed almost entirely of the residues extending from the C-terminal half of alpha2 (Switch II) together with the alpha3 helix and its junction with the beta5 strand. Effector binding does not induce substantial changes in the structure of Galpha*GTP. Effectors are structurally diverse. Different effectors may recognize distinct subsets of effector-binding residues of the same Galpha protein. Specificity may also be conferred by differences in the main chain conformation of effector-binding regions of Galpha subunits. Several Galpha regulatory mechanisms are operative. In the regulation of GMP phospodiesterase, Galphat sequesters an inhibitory subunit. Galphas is an allosteric activator and inhibitor of adenylyl cyclase, and Galphai is an allosteric inhibitor. Galphaq does not appear to regulate GRK, but is rather sequestered by it. GTP hydrolysis terminates the signaling state of Galpha. The binding energy of GTP that is used to stabilize the Galpha:effector complex is dissipated in this reaction. Chemical steps of GTP hydrolysis, specifically, formation of a dissociative transition state, is rate limiting in Ras, a model G protein GTPase, even in the presence of a GAP; however, the energy of enzyme reorganization to produce a catalytically active conformation appears to be substantial. It is possible that the collapse of the switch regions, associated with Galpha deactivation, also encounters a kinetic barrier, and is coupled to product (Pi) release or an event preceding formation of the GDP*Pi complex. Evidence for a catalytic intermediate, possibly metaphosphate, is discussed. Galpha GAPs, whether exogenous proteins or effector-linked domains, bind to a discrete locus of Galpha that is composed of Switch I and the N-terminus of Switch II. This site is immediately adjacent to, but does not substantially overlap, the Galpha effector binding site. Interactions of effectors and exogenous GAPs with Galpha proteins can be synergistic or antagonistic, mediated by allosteric interactions among the three molecules. Unlike GAPs for small GTPases, Galpha GAPs supply no catalytic residues, but rather appear to reduce the activation energy for catalytic activation of the Galpha catalytic site.
Collapse
Affiliation(s)
- Stephen R Sprang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | |
Collapse
|
33
|
Sternweis PC, Carter AM, Chen Z, Danesh SM, Hsiung YF, Singer WD. Regulation of Rho guanine nucleotide exchange factors by G proteins. ADVANCES IN PROTEIN CHEMISTRY 2007; 74:189-228. [PMID: 17854659 DOI: 10.1016/s0065-3233(07)74006-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Monomeric Rho GTPases regulate cellular dynamics through remodeling of the cytoskeleton, modulation of immediate signaling pathways, and longer-term regulation of gene transcription. One family of guanine nucleotide exchange factors for Rho proteins (RhoGEFs) provides a direct pathway for regulation of RhoA by cell surface receptors coupled to heterotrimeric G proteins. Some of these RhoGEFs also contain RGS domains that can attenuate signaling by the G(12) and G(13) proteins. The regulation provided by these RhoGEFs is defined by their selective regulation by specific G proteins, phosphorylation by kinases, and potential localization with signaling partners. Evidence of their physiological importance is derived from gene knockouts in Drosophila and mice. Current understanding of the basic regulatory mechanisms of these RhoGEFs is discussed. An overview of identified interactions with other signaling proteins suggests the growing spectrum of their involvement in numerous signaling pathways.
Collapse
Affiliation(s)
- Paul C Sternweis
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | | | | | |
Collapse
|
34
|
Herbrand U, Ahmadian MR. p190-RhoGAP as an integral component of the Tiam1/Rac1-induced downregulation of Rho. Biol Chem 2006; 387:311-7. [PMID: 16542153 DOI: 10.1515/bc.2006.041] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractThe Rho family of small GTPases plays a central role in intracellular signal transduction, particularly in reorganization of the actin cytoskeleton. Rho activity induces cell contractility, whereas Rac promotes cellular protrusion, which counteracts Rho signaling. In this regard, the reciprocal balance between these GTPases determines cell morphology and migratory behavior. Here we demonstrate that Tiam1/Rac1 signaling is able to antagonize Rho activity directly at the GTPase level in COS-7 cells. p190-RhoGAP plays a central regulatory role in this signaling pathway. Interfering with its activation by Src-kinase-dependent tyrosine phosphorylation or its recruitment to the membrane through interaction with the SH2 domains of p120-RasGAP blocks the Tiam1-mediated rapid downregulation of Rho. This process is mediated by Rac1, but not by Rac2 or Rac3 isoforms. Our data provide evidence for a biochemical pathway of the reciprocal regulation of two related small GTPases, which are key elements in cell migration.
Collapse
Affiliation(s)
- Ulrike Herbrand
- Max Planck Institute of Molecular Physiology, Department of Structural Biology, Otto-Hahn-Strasse 11, D-44227 Dortmund, Germany.
| | | |
Collapse
|
35
|
Rubtsov A, Strauch P, Digiacomo A, Hu J, Pelanda R, Torres RM. Lsc regulates marginal-zone B cell migration and adhesion and is required for the IgM T-dependent antibody response. Immunity 2005; 23:527-38. [PMID: 16286020 DOI: 10.1016/j.immuni.2005.09.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2004] [Revised: 09/19/2005] [Accepted: 09/22/2005] [Indexed: 11/29/2022]
Abstract
The humoral immune response to protein antigens is composed of a rapid low-affinity IgM antibody response followed by an IgG response exhibiting higher affinity. Here, we demonstrate that Lsc, a protein that regulates G protein-coupled-receptor signaling and RhoA activation, is required by B lymphocytes for the antigen-specific IgM antibody response to a protein antigen. We further show that in lsc(-/-) mice, MZB cells are selectively affected such that naive and in vivo-activated MZB cells migrate toward sphingosine-1-phosphate at increased proportions but release inefficiently from integrin ligands. Consequently, lsc(-/-) MZB cells do not traffick appropriately in an immune response and do not contribute to the TD antibody response. These data demonstrate that Lsc regulates the migration and adhesion of MZB cells, and this regulation appears to be required for these cells to contribute to the antibody response to TD antigens.
Collapse
Affiliation(s)
- Anatoly Rubtsov
- University of Colorado Health Sciences Center, Integrated Department of Immunology, Denver, Colorado 80207, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Dbl homology (DH) domains are almost always followed immediately by pleckstrin homology (PH) domains in Dbl family proteins, and these DH-PH fragments directly activate GDP-bound Rho GTPases by catalyzing the exchange of GDP for GTP. New crystal structures of the DH-PH domains from leukemia-associated Rho guanine nucleotide exchange factor (RhoGEF) and PDZ-RhoGEF bound to RhoA reveal how DH-PH domains cooperate to specifically activate Rho GTPases.
Collapse
Affiliation(s)
- Kent L Rossman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
37
|
Bakal CJ, Finan D, LaRose J, Wells CD, Gish G, Kulkarni S, DeSepulveda P, Wilde A, Rottapel R. The Rho GTP exchange factor Lfc promotes spindle assembly in early mitosis. Proc Natl Acad Sci U S A 2005; 102:9529-34. [PMID: 15976019 PMCID: PMC1157106 DOI: 10.1073/pnas.0504190102] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Rho GTPases regulate reorganization of actin and microtubule cytoskeletal structures during both interphase and mitosis. The timing and subcellular compartment in which Rho GTPases are activated is controlled by the large family of Rho GTP exchange factors (RhoGEFs). Here, we show that the microtubule-associated RhoGEF Lfc is required for the formation of the mitotic spindle during prophase/prometaphase. The inability of cells to assemble a functioning spindle after Lfc inhibition resulted in a delay in mitosis and an accumulation of prometaphase cells. Inhibition of Lfc's primary target Rho GTPase during prophase/prometaphase, or expression of a catalytically inactive mutant of Lfc, also prevented normal spindle assembly and resulted in delays in mitotic progression. Coinjection of constitutively active Rho GTPase rescued the spindle defects caused by Lfc inhibition, suggesting the requirement of RhoGTP in regulating spindle assembly. Lastly, we implicate mDia1 as an important effector of Lfc signaling. These findings demonstrate a role for Lfc, Rho, and mDia1 during mitosis.
Collapse
Affiliation(s)
- Christopher J Bakal
- Department of Medical Biophysics, University of Toronto, 1 King's College Circle, Toronto, ON, Canada M5S 1A8
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ziembicki J, Tandon R, Schelling JR, Sedor JR, Miller RT, Huang C. Mechanical force-activated phospholipase D is mediated by Galpha12/13-Rho and calmodulin-dependent kinase in renal epithelial cells. Am J Physiol Renal Physiol 2005; 289:F826-34. [PMID: 15914773 DOI: 10.1152/ajprenal.00412.2004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renal glomerulus, the site of plasma ultrafiltration, is exposed to mechanical force in vivo arising from capillary blood pressure and fluid flow. Studies of cultured podocytes demonstrate that they respond to stretch by altering the structure of the actin cytoskeleton, but the mechanisms by which physical force triggers this architectural change and the signaling pathways that lead to generation of second messengers are not defined. In the present study, we found that in renal epithelial cells [podocytes and Madin-Darby canine kidney (MDCK) cells], application of mechanical force to the cell surface through fibronectin-coated ferric beads and exposure of the cells to magnetic force lead to Rho translocation and actin cytoskeleton reorganization. This application of force recruited Rho and filamentous actin (F-actin) to bead loci and subsequently stimulated phospholipase D (PLD), a downstream effector of Rho. Using MDCK cells that stably express regulators of G protein-signaling (RGS) proteins [RGS4 attenuates Galpha(i) and Galpha(q), and the p115RhoGEF-RGS domain (p115-RGS) attenuates Galpha(12/13)] to define the signaling pathway, we found that mechanical force induced Galpha(12/13)-Rho activation and increased F-actin to stimulate PLD activity. The activation can be partially prevented by the C(3) exoenzyme. Pretreatment of the cells with chemical inhibitors of several kinases showed that calmodulin-dependent kinase is also involved in stretch-induced PLD activation by a separate pathway. Taken together, our data demonstrate that in cultured podocytes and MDCK cells, mechanical force leads to actin cytoskeleton reorganization and PLD activation. The signaling pathways for PLD activation involve Galpha(12/13)/Rho/F-actin and calmodulin-dependent kinase.
Collapse
Affiliation(s)
- Jenny Ziembicki
- Dept. of Medicine, Case Western Reserve Univ., Louis Stokes Veterans Affairs Medical Ctr., 10701 East Blvd., 151W, Cleveland, OH 44106, USA
| | | | | | | | | | | |
Collapse
|
39
|
Birukova AA, Birukov KG, Smurova K, Adyshev D, Kaibuchi K, Alieva I, Garcia JGN, Verin AD. Novel role of microtubules in thrombin-induced endothelial barrier dysfunction. FASEB J 2005; 18:1879-90. [PMID: 15576491 DOI: 10.1096/fj.04-2328com] [Citation(s) in RCA: 174] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Disturbances in endothelial cell (EC) barrier regulation are critically dependent upon rearrangements of EC actin cytoskeleton. However, the role of microtubule (MT) network in the regulation of EC permeability is not well understood. We examined involvement of MT remodeling in thrombin-induced EC permeability and explored MT regulation by heterotrimeric G12/13 proteins and by small GTPase Rho. Thrombin induced phosphorylation of MT regulatory protein tau at Ser409 and Ser262 and peripheral MT disassembly, which was linked to increased EC permeability. MT stabilization by taxol attenuated thrombin-induced permeability, actin remodeling, and paracellular gap formation and diminished thrombin-induced activation of Rho and Rho-kinase. Expression of activated Galpha12/13 subunits involved in thrombin-mediated signaling or their effector p115RhoGEF involved in Rho activation caused MT disassembly, whereas p115RhoGEF-specific negative regulator RGS preserved MT from thrombin-induced disassembly. Consistent with these results, expression of activated RhoA and Rho-kinase induced MT disassembly. Conversely, thrombin-induced disassembly of peripheral MT network was attenuated by expression of dominant negative RhoA and Rho-kinase mutants or by pharmacological inhibition of Rho-kinase. Collectively, our data demonstrate for the first time a critical involvement of MT disassembly in thrombin-induced EC barrier dysfunction and indicate G-protein-dependent mechanisms of thrombin-induced MT alteration.
Collapse
Affiliation(s)
- Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, 5200 Eastern Ave., MFL Center Tower 660, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Grabocka E, Wedegaertner PB. Functional consequences of G alpha 13 mutations that disrupt interaction with p115RhoGEF. Oncogene 2005; 24:2155-65. [PMID: 15735747 PMCID: PMC1351220 DOI: 10.1038/sj.onc.1208414] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The G-protein alpha subunit, alpha(13), regulates cell growth and differentiation through the monomeric Rho GTPase. Alpha(13) activates Rho through direct stimulation of the guanine nucleotide exchange factor p115RhoGEF, which contains a regulator of G-protein signaling homology domain (RH) in its N-terminus. Through its RH domain, p115RhoGEF also functions as a GAP for G alpha(13). The mechanism for the G alpha(13)/p115RhoGEF interaction is not well understood. Here, we determined specific alpha(13) residues important for its interaction with p115RhoGEF. GST-pulldowns and co-immunoprecipitation assays revealed that individually mutating alpha(13) residues Lys204, Glu229, or Arg232 to opposite charge residues disrupts the interaction of activated alpha(13) with the RH domain of p115RhoGEF or full-length p115RhoGEF. We further demonstrate that mutation of Glu229, and to a lesser extent Lys204 or Arg232, disrupts the ability of activated alpha(13) to induce the recruitment of p115RhoGEF to the plasma membrane (PM) and to activate Rho-mediated serum response element-luciferase gene transcription. Interestingly, an alpha(13) mutant where a conserved Gly was mutated to a Ser (G205S) retained its ability to bind to p115RhoGEF, induce p115RhoGEF recruitment to the PM, and activate Rho-dependent signaling, even though identical Gly to Ser mutations in other alpha disrupt their interaction with regulator of G-protein signaling (RGS) proteins. These results demonstrate that, whereas several features of a typical alpha/RGS interaction are preserved in the alpha(13)/p115RhoGEF interaction, there are also significant differences.
Collapse
Affiliation(s)
| | - Philip B. Wedegaertner
- Corresponding address: Philip Wedegaertner, Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, 233 S. 10 St., 839 BLSB, Philadelphia, PA 19107, tel: 215-503-3137, fax: 215-923-2117, e-mail:
| |
Collapse
|
41
|
Chen Z, Singer WD, Sternweis PC, Sprang SR. Structure of the p115RhoGEF rgRGS domain–Gα13/i1 chimera complex suggests convergent evolution of a GTPase activator. Nat Struct Mol Biol 2005; 12:191-7. [PMID: 15665872 DOI: 10.1038/nsmb888] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2004] [Accepted: 11/23/2004] [Indexed: 11/08/2022]
Abstract
p115RhoGEF, a guanine nucleotide exchange factor (GEF) for Rho GTPase, is also a GTPase-activating protein (GAP) for G12 and G13 heterotrimeric Galpha subunits. The GAP function of p115RhoGEF resides within the N-terminal region of p115RhoGEF (the rgRGS domain), which includes a module that is structurally similar to RGS (regulators of G-protein signaling) domains. We present here the crystal structure of the rgRGS domain of p115RhoGEF in complex with a chimera of Galpha13 and Galphai1. Two distinct surfaces of rgRGS interact with Galpha. The N-terminal betaN-alphaN hairpin of rgRGS, rather than its RGS module, forms intimate contacts with the catalytic site of Galpha. The interface between the RGS module of rgRGS and Galpha is similar to that of a Galpha-effector complex, suggesting a role for the rgRGS domain in the stimulation of the GEF activity of p115RhoGEF by Galpha13.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Biochemistry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, USA
| | | | | | | |
Collapse
|
42
|
Kostenko EV, Mahon GM, Cheng L, Whitehead IP. The Sec14 Homology Domain Regulates the Cellular Distribution and Transforming Activity of the Rho-specific Guanine Nucleotide Exchange Factor Dbs. J Biol Chem 2005; 280:2807-17. [PMID: 15531584 DOI: 10.1074/jbc.m411139200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Dbs is a Rho-specific guanine nucleotide exchange factor that was identified in a screen for proteins whose overexpression cause deregulated growth in murine fibroblasts. Dbs contains multiple recognizable motifs including a centrally located Rho-specific guanine nucleotide exchange factor domain, a COOH-terminal Src homology 3 domain, two spectrin-like repeats, and a recently identified NH(2)-terminal Sec14 homology domain. The transforming potential of Dbs is substantially activated by the removal of inhibitory sequences that lie outside of the core catalytic sequences, and in this current study we mapped this inhibition to the Sec14 domain. Surprisingly removal of the NH(2) terminus did not alter the catalytic activity of Dbs in vivo but rather altered its subcellular distribution. Whereas full-length Dbs was distributed primarily in a perinuclear structure that coincides with a marker for the Golgi apparatus, removal of the Sec14 domain was associated with translocation of Dbs to the cell periphery where it accumulated within membrane ruffles and lamellipodia. However, translocation of Dbs and the concomitant changes in the actin cytoskeleton were not sufficient to fully activate Dbs transformation. The Sec14 domain also forms intramolecular contacts with the pleckstrin homology domain, and these contacts must also be relieved to achieve full transforming activity. Collectively these observations suggest that the Sec14 domain regulates Dbs transformation through at least two distinct mechanisms, neither of which appears to directly influence the in vivo exchange activity of the protein.
Collapse
Affiliation(s)
- Elena V Kostenko
- Department of Microbiology and Molecular Genetics, University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey 07103, USA
| | | | | | | |
Collapse
|
43
|
Kristelly R, Gao G, Tesmer JJG. Structural determinants of RhoA binding and nucleotide exchange in leukemia-associated Rho guanine-nucleotide exchange factor. J Biol Chem 2004; 279:47352-62. [PMID: 15331592 DOI: 10.1074/jbc.m406056200] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Rho guanine-nucleotide exchange factors (RhoGEFs) activate Rho GTPases, and thereby regulate cytoskeletal structure, gene transcription, and cell migration. Leukemia-associated RhoGEF (LARG) belongs to a small subfamily of RhoGEFs that are RhoA-selective and directly activated by the Galpha12/13 family of heterotrimeric G proteins. Herein we describe the atomic structures of the catalytic Dbl homology (DH) and pleckstrin homology (PH) domains of LARG alone and in complex with RhoA. These structures demonstrate that the DH/PH domains of LARG can undergo a dramatic conformational change upon binding RhoA, wherein both the DH and PH domains directly engage RhoA. Through mutational analysis we show that full nucleotide exchange activity requires a novel N-terminal extension on the DH domain that is predicted to exist in a broader family of RhoGEFs that includes p115-RhoGEF, Lbc, Lfc, Net1, and Xpln, and identify regions within the LARG PH domain that contribute to its ability to facilitate nucleotide exchange in vitro. In crystals of the DH/PH-RhoA complex, the active site of RhoA adopts two distinct GDP-excluding conformations among the four unique complexes in the asymmetric unit. Similar changes were previously observed in structures of nucleotide-free Ras and Ef-Tu. A potential protein-docking site on the LARG PH domain is also evident and appears to be conserved throughout the Lbc subfamily of RhoGEFs.
Collapse
Affiliation(s)
- Romana Kristelly
- Department of Chemistry and Biochemistry, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas 78712-0165, USA
| | | | | |
Collapse
|
44
|
Diviani D, Abuin L, Cotecchia S, Pansier L. Anchoring of both PKA and 14-3-3 inhibits the Rho-GEF activity of the AKAP-Lbc signaling complex. EMBO J 2004; 23:2811-20. [PMID: 15229649 PMCID: PMC514948 DOI: 10.1038/sj.emboj.7600287] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2004] [Accepted: 06/03/2004] [Indexed: 01/05/2023] Open
Abstract
A-kinase anchoring proteins (AKAPs) target the cAMP-regulated protein kinase (PKA) to its physiological substrates. We recently identified a novel anchoring protein, called AKAP-Lbc, which functions as a PKA-targeting protein as well as a guanine nucleotide exchange factor (GEF) for RhoA. We demonstrated that AKAP-Lbc Rho-GEF activity is stimulated by the alpha subunit of the heterotrimeric G protein G12. Here, we identified 14-3-3 as a novel regulatory protein interacting with AKAP-Lbc. Elevation of the cellular concentration of cAMP activates the PKA holoenzyme anchored to AKAP-Lbc, which phosphorylates the anchoring protein on the serine 1565. This phosphorylation event induces the recruitment of 14-3-3, which inhibits the Rho-GEF activity of AKAP-Lbc. AKAP-Lbc mutants that fail to interact with PKA or with 14-3-3 show a higher basal Rho-GEF activity as compared to the wild-type protein. This suggests that, under basal conditions, 14-3-3 maintains AKAP-Lbc in an inactive state. Therefore, while it is known that AKAP-Lbc activity can be stimulated by Galpha12, in this study we demonstrated that it is inhibited by the anchoring of both PKA and 14-3-3.
Collapse
Affiliation(s)
- Dario Diviani
- Département de Pharmacologie et de Toxicologie, Faculté de Médecine, Lausanne, Switzerland.
| | | | | | | |
Collapse
|
45
|
Huang C, Hujer KM, Wu Z, Miller RT. The Ca2+-sensing receptor couples to Galpha12/13 to activate phospholipase D in Madin-Darby canine kidney cells. Am J Physiol Cell Physiol 2004; 286:C22-30. [PMID: 12954603 DOI: 10.1152/ajpcell.00229.2003] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Ca2+-sensing receptor (CaR) couples to multiple G proteins involved in distinct signaling pathways: Galphai to inhibit the activity of adenylyl cyclase and activate ERK, Galphaq to stimulate phospholipase C and phospholipase A2, and Gbetagamma to stimulate phosphatidylinositol 3-kinase. To determine whether the receptor also couples to Galpha12/13, we investigated the signaling pathway by which the CaR regulates phospholipase D (PLD), a known Galpha12/13 target. We established Madin-Darby canine kidney (MDCK) cell lines that stably overexpress the wild-type CaR (CaRWT) or the nonfunctional mutant CaRR796W as a negative control, prelabeled these cells with [3H]palmitic acid, and measured CaR-stimulated PLD activity as the formation of [3H]phosphatidylethanol (PEt). The formation of [3H]PEt increased in a time-dependent manner in the cells that overexpress the CaRWT but not the CaRR796W. Treatment of the cells with C3 exoenzyme inhibited PLD activity, which indicates that the CaR activates the Rho family of small G proteins, targets of Galpha12/13. To determine which G protein(s) the CaR couples to in order to activate Rho and PLD, we pretreated the cells with pertussis toxin to inactivate Galphai or coexpressed regulators of G protein-signaling (RGS) proteins to attenuate G protein signaling (RGS4 for Galphai and Galphaq, and a p115RhoGEF construct containing the RGS domain for Galpha12/13). Overexpression of p115RhoGEF-RGS in the MDCK cells that overexpress CaRWT inhibited extracellular Ca2+-stimulated PLD activity, but pretreatment of cells with pertussis toxin and overexpression of RGS4 were without effect. The involvement of other signaling components such as protein kinase C, ADP-ribosylation factor, and phosphatidylinositol biphosphate was excluded. These findings demonstrate that the CaR couples to Galpha12/13 to regulate PLD via a Rho-dependent mechanism and does so independently of Galphai and Galphaq. This suggests that the CaR may regulate cytoskeleton via Galpha12/13, Rho, and PLD.
Collapse
Affiliation(s)
- Chunfa Huang
- Division of Nephrology, Department of Medicine, Case Western Reserve University, Louis Stokes Veteran Affairs Medical Center, 10701 East Boulevard 151W, Cleveland, OH 44106, USA.
| | | | | | | |
Collapse
|
46
|
Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev 2003; 83:1325-58. [PMID: 14506307 DOI: 10.1152/physrev.00023.2003] [Citation(s) in RCA: 1553] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Ca2+ sensitivity of smooth muscle and nonmuscle myosin II reflects the ratio of activities of myosin light-chain kinase (MLCK) to myosin light-chain phosphatase (MLCP) and is a major, regulated determinant of numerous cellular processes. We conclude that the majority of phenotypes attributed to the monomeric G protein RhoA and mediated by its effector, Rho-kinase (ROK), reflect Ca2+ sensitization: inhibition of myosin II dephosphorylation in the presence of basal (Ca2+ dependent or independent) or increased MLCK activity. We outline the pathway from receptors through trimeric G proteins (Galphaq, Galpha12, Galpha13) to activation, by guanine nucleotide exchange factors (GEFs), from GDP. RhoA. GDI to GTP. RhoA and hence to ROK through a mechanism involving association of GEF, RhoA, and ROK in multimolecular complexes at the lipid cell membrane. Specific domains of GEFs interact with trimeric G proteins, and some GEFs are activated by Tyr kinases whose inhibition can inhibit Rho signaling. Inhibition of MLCP, directly by ROK or by phosphorylation of the phosphatase inhibitor CPI-17, increases phosphorylation of the myosin II regulatory light chain and thus the activity of smooth muscle and nonmuscle actomyosin ATPase and motility. We summarize relevant effects of p21-activated kinase, LIM-kinase, and focal adhesion kinase. Mechanisms of Ca2+ desensitization are outlined with emphasis on the antagonism between cGMP-activated kinase and the RhoA/ROK pathway. We suggest that the RhoA/ROK pathway is constitutively active in a number of organs under physiological conditions; its aberrations play major roles in several disease states, particularly impacting on Ca2+ sensitization of smooth muscle in hypertension and possibly asthma and on cancer neoangiogenesis and cancer progression. It is a potentially important therapeutic target and a subject for translational research.
Collapse
Affiliation(s)
- Andrew P Somlyo
- Dept. of Molecular Physiology and Biological Physics, Univ. of Virginia, PO Box 800736, Charlottesville, VA 22908-0736.
| | | |
Collapse
|
47
|
Le Page SL, Bi Y, Williams JA. CCK-A receptor activates RhoA through G alpha 12/13 in NIH3T3 cells. Am J Physiol Cell Physiol 2003; 285:C1197-206. [PMID: 12853286 DOI: 10.1152/ajpcell.00083.2003] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cholecystokinin (CCK) is a major regulator of pancreatic acinar cells and was shown previously to be capable of inducing cytoskeletal changes in these cells. In the present study, using NIH3T3 cells stably transfected with CCK-A receptors as a model cell, we demonstrate that CCK can induce actin stress fibers through a G13- and RhoA-dependent mechanism. CCK induced stress fibers within minutes similar to those induced by lysophosphatidic acid (LPA), the active component of serum. The effects of CCK were mimicked by active RhoV14 and blocked by dominant-negative RhoN19, Clostridium botulinum C3 transferase, and the Rho-kinase inhibitor Y-27632. CCK rapidly induced active Rho in cells as shown with a pull-down assay using the Rho binding domain of rhotekin and by a serum response element (SRE)-luciferase reporter assay. To evaluate the G protein mediating the action of CCK, cells were transfected with active alpha-subunits; Galpha13 and Galpha12 but not Galphaq induced stress fibers and in some cases cell rounding. A p115 Rho guanine nucleotide exchange factor (GEF) regulator of G protein signaling (RGS) domain known to interact with G12/13 inhibited active alpha12/13-and CCK-induced stress fibers, whereas RGS2 and RGS4, which are known to inhibit Gq, had no effect. Cotransfection with plasmids coding for the G protein alpha-subunit carboxy-terminal peptide from alpha13 and, to a lesser extent alpha12, also inhibited the effect of CCK, whereas the peptide from alphaq did not. These results show that in NIH3T3 cells bearing CCK-A receptors, CCK activates Rho primarily through G13, leading to rearrangement of the actin cytoskeleton.
Collapse
Affiliation(s)
- Sophie L Le Page
- Department. of Molecular & Integrative Physiology, University of Michigan, 7744 Medical Science II, Ann Arbor, MI 48109-0622, USA
| | | | | |
Collapse
|
48
|
Shin EY, Woo KN, Lee CS, Koo SH, Kim YG, Kim WJ, Bae CD, Chang SI, Kim EG. Basic fibroblast growth factor stimulates activation of Rac1 through a p85 betaPIX phosphorylation-dependent pathway. J Biol Chem 2003; 279:1994-2004. [PMID: 14557270 DOI: 10.1074/jbc.m307330200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In a previous study (Shin, E. Y., Shin, K. S., Lee, C. S., Woo, K. N., Quan, S. H., Soung, N. K., Kim, Y. G., Cha, C. I., Kim, S. R., Park, D., Bokoch, G. M., and Kim, E. G. (2002) J. Biol. Chem. 277, 44417-44430) we reported that phosphorylation of p85 betaPIX, a guanine nucleotide exchange factor (GEF) for Rac1/Cdc42, is a signal for translocation of the PIX complex to neuronal growth cones and is associated with basic fibroblast growth factor (bFGF)-induced neurite outgrowth. However, the issue of whether p85 betaPIX phosphorylation affects GEF activity on Rac1/Cdc42 is yet to be explored. Here we show that Rac1 activation occurs in a p85 betaPIX phosphorylation-dependent manner. A GST-PBD binding assay reveals that Rac1 is activated in a dose- and time-dependent manner in PC12 cells in response to bFGF. Inhibition of ERK or PAK2, the kinases upstream of p85 betaPIX in the bFGF signaling, prevents Rac1 activation, suggesting that phosphorylation of p85 betaPIX functions upstream of Rac1 activation. To directly address this issue, transfection studies with wild-type and mutant p85 betaPIX (S525A/T526A, a non-phosphorylatable form) were performed. Expression of mutant PIX markedly inhibits both bFGF- and nerve growth factor (NGF)-induced activation of Rac1, indicating that phosphorylation of p85 betaPIX is responsible for activation of this G protein. Both wild-type and mutant p85 betaPIX displaying negative GEF activity (L238R/L239S) are similarly recruited to growth cones, suggesting that Rac1 activation is not essential for translocation of the PIX complex (PAK2-p85 betaPIX-Rac1). However, expression of mutant p85 betaPIX (L238R/L239S) results in retraction of the pre-existing neurites. Our results provide evidence that bFGF- and NGF-induced phosphorylation of p85 betaPIX mediates Rac1 activation, which in turn regulates cytoskeletal reorganization at growth cones, but not translocation of the PIX complex.
Collapse
Affiliation(s)
- Eun-Young Shin
- Department of Biochemistry, College of Medicine, Medical Research Institute and Biotechnology Research Institute, College of Natural Sciences, Chungbuk National University, San 48, Gaesin-dong, Heungduk-ku, Cheongju 361-763, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Blood lymphocyte numbers, which are maintained by recirculation through secondary lymphoid organs, are essential for the efficient development of immune responses. Recirculating populations of B and T lymphocytes are regulated by the sphingosine-1-phosphate (S1P) receptor-dependent control of lymphocyte egress. T-cell egress from thymus into blood, egress from lymph node and Peyer's patch into lymph, and B-cell egress into lymph are rapidly and completely inhibited by agonism of S1P receptors. Mesenteric lymph nodes show log-jamming of lymphocytes subjacent to sinus-lining endothelium. Agonism of S1P receptors produces rapid peripheral blood lymphopenia, which is maintained in the presence of receptor agonist. Effector CD4+ and CD8+ T cells, produced by clonal expansion in draining lymph node in response to antigen, are sequestered in lymph node and fail to reach the peripheral blood. The S1P receptor system may represent an early physiological link between the non-specific inflammatory response and the alteration of lymphocyte traffic through draining lymph nodes. Pharmacological subversion of the S1P receptor system, through systemic S1P agonist-induced inhibition of lymphocyte egress, suppresses antigenic responses to peripheral, but not to systemically, delivered antigen. This inhibition induces significant immunosuppression in models of transplantation and autoimmune tissue damage that may prove to be of clinical benefit.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
50
|
Rosen H, Alfonso C, Surh CD, McHeyzer-Williams MG. Rapid induction of medullary thymocyte phenotypic maturation and egress inhibition by nanomolar sphingosine 1-phosphate receptor agonist. Proc Natl Acad Sci U S A 2003; 100:10907-12. [PMID: 12954982 PMCID: PMC196901 DOI: 10.1073/pnas.1832725100] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2003] [Indexed: 11/18/2022] Open
Abstract
Only a small number of T cells generated in the thymus each day are selected to replenish the peripheral T cell pool. Much is known about thymic selection; however, little is known of the mechanisms regulating medullary maturation and the release of mature T cells into the blood. Here we demonstrate a rapid acceleration of medullary thymocyte phenotypic maturation through loss of CD69 induced by sphingosine 1-phosphate (S1P) receptor agonist. Low nanomolar agonist concentrations selectively induce changes in CD69(int) CD62L(high) single positive T cells, resulting in down-modulation of CD69 within 2 h. While CD69 loss is accelerated, egress of mature T cells into blood is inhibited >95% within 2 h. Both processes exhibit parallel sensitivities and dose-responses. Together, these data reveal a potent means for rapidly regulating thymic export where S1P receptor agonism alters both phenotypic maturation and egress of thymocytes into blood during late thymic maturation. The S1P system is now shown to acutely regulate both thymic and lymph node egress. Inhibition of lymphocyte egress from thymus and lymph node can contribute synergistically to clinically useful immunosupression by disrupting recirculation of peripheral T cells.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|