Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 288(2): 1184-99, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23184944

ABSTRACT

ELMO and DOCK180 proteins form an evolutionarily conserved module controlling Rac GTPase signaling during cell migration, phagocytosis, and myoblast fusion. Here, we identified the microtubule and actin-binding spectraplakin ACF7 as a novel ELMO-interacting partner. A C-terminal polyproline segment in ELMO and the last spectrin repeat of ACF7 mediate a direct interaction between these proteins. Co-expression of ELMO1 with ACF7 promoted the formation of long membrane protrusions during integrin-mediated cell spreading. Quantification of membrane dynamics established that coupling of ELMO and ACF7 increases the persistence of the protruding activity. Mechanistically, we uncovered a role for ELMO in the recruitment of ACF7 to the membrane to promote microtubule capture and stability. Functionally, these effects of ELMO and ACF7 on cytoskeletal dynamics required the Rac GEF DOCK180. In conclusion, our findings support a role for ELMO in protrusion stability by acting at the interface between the actin cytoskeleton and the microtubule network.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cytoskeletal Proteins/physiology , Microfilament Proteins/metabolism , Microtubules/metabolism , Cell Line , Cell Membrane/metabolism , Cytoskeleton/metabolism , Fluorescence Resonance Energy Transfer , Humans , Immunoprecipitation
2.
Curr Biol ; 20(22): 2021-7, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21035343

ABSTRACT

Dedicator of cytokinesis (DOCK) proteins are guanine nucleotide exchange factors (GEFs) controlling the activity of Rac1/Cdc42 during migration, phagocytosis, and myoblast fusion [1-4]. Engulfment and cell motility (ELMO) proteins bind a subset of DOCK members and are emerging as critical regulators of Rac signaling [5-10]. Although formation of a DOCK180/ELMO complex is not essential for Rac1 activation, ELMO mutants deficient in binding to DOCK180 are unable to promote cytoskeleton remodeling [11]. How ELMO regulates signaling through DOCK GEFs is poorly understood. Here, we identify an autoinhibitory switch in ELMO presenting homology to a regulatory unit described for Dia formins. One part of the switch, composed of a Ras-binding domain (RBD) and Armadillo repeats, is positioned N-terminally while the other is housed in the C terminus. We demonstrate interaction between these fragments, suggesting autoinhibition of ELMO. Using a bioluminescence resonance energy transfer biosensor, we establish that ELMO undergoes conformational changes upon disruption of autoinhibition. We found that engagement of ELMO to RhoG, or with DOCK180, promotes the relief of autoinhibition in ELMO. Functionally, we found that ELMO mutants with impaired autoregulatory activity promote cell elongation. These results demonstrate an unsuspected level of regulation for Rac1 signaling via autoinhibition of ELMO.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Sequence , Animals , Binding Sites , Biosensing Techniques , Computational Biology , Conserved Sequence , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/chemistry , HEK293 Cells , Humans , Models, Biological , Molecular Sequence Data , Protein Structure, Tertiary , Two-Hybrid System Techniques , rac GTP-Binding Proteins/chemistry , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/physiology
3.
Proc Natl Acad Sci U S A ; 105(40): 15446-51, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18820033

ABSTRACT

Dock1 (also known as Dock180) is a prototypical member of a new family of atypical Rho GTPase activators. Genetic studies in Drosophila and Caenorhabditis elegans have demonstrated that Dock1 orthologues in these organisms have a crucial role in activating Rac GTPase signaling. We generated mutant alleles of the closely related Dock1 and Dock5 genes to study their function in mammals. We report that while Dock5 is dispensable for normal mouse embryogenesis, Dock1 has an essential role in embryonic development. A dramatic reduction of all skeletal muscle tissues is observed in Dock1-null embryos. Mechanistically, this embryonic defect is attributed to a strong deficiency in myoblast fusion, which is detectable both in vitro and in vivo. Furthermore, we have uncovered a contribution of Dock5 toward myofiber development. These studies identify Dock1 and Dock5 as critical regulators of the fusion step during primary myogenesis in mammals and demonstrate that a specific component of the myoblast fusion machinery identified in Drosophila plays an evolutionarily conserved role in higher vertebrates.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Myoblasts/metabolism , Animals , Cell Fusion , Embryo, Mammalian/metabolism , Fluorescent Antibody Technique , Guanine Nucleotide Exchange Factors/genetics , Mice , Mice, Transgenic , Models, Genetic , Muscle Development , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Mutation
4.
Mol Biol Cell ; 19(11): 4837-51, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18768751

ABSTRACT

The mammalian DOCK180 protein belongs to an evolutionarily conserved protein family, which together with ELMO proteins, is essential for activation of Rac GTPase-dependent biological processes. Here, we have analyzed the DOCK180-ELMO1 interaction, and map direct interaction interfaces to the N-terminal 200 amino acids of DOCK180, and to the C-terminal 200 amino acids of ELMO1, comprising the ELMO1 PH domain. Structural and biochemical analysis of this PH domain reveals that it is incapable of phospholipid binding, but instead structurally resembles FERM domains. Moreover, the structure revealed an N-terminal amphiphatic alpha-helix, and point mutants of invariant hydrophobic residues in this helix disrupt ELMO1-DOCK180 complex formation. A secondary interaction between ELMO1 and DOCK180 is conferred by the DOCK180 SH3 domain and proline-rich motifs at the ELMO1 C-terminus. Mutation of both DOCK180-interaction sites on ELMO1 is required to disrupt the DOCK180-ELMO1 complex. Significantly, although this does not affect DOCK180 GEF activity toward Rac in vivo, Rac signaling is impaired, implying additional roles for ELMO in mediating intracellular Rac signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction , rac GTP-Binding Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Conserved Sequence , Enzyme Activation , Evolution, Molecular , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Phosphatidylinositols/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship , Surface Properties , rac GTP-Binding Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...