Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 136(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36994763

ABSTRACT

Looking back at two decades of research on SPIRE actin nucleator proteins, the first decade was clearly dominated by the discovery of SPIRE proteins as founding members of the novel WH2-domain-based actin nucleators, which initiate actin filament assembly through multiple WH2 actin-binding domains. Through complex formation with formins and class 5 myosins, SPIRE proteins coordinate actin filament assembly and myosin motor-dependent force generation. The discovery of SPIRE-regulated cytoplasmic actin filament meshworks in oocytes initiated the next phase of SPIRE research, which has found that SPIRE proteins are integrated in a diverse range of cell biological processes. In addition to regulating vesicle-based actin filament meshworks, SPIRE proteins function in the organisation of actin structures driving the inward movement of pronuclei of the mouse zygote. Localisation at cortical ring structures and the results of knockdown experiments indicate that SPIRE proteins function in the formation of meiotic cleavage sites in mammalian oocytes and the externalisation of von Willebrand factor from endothelial cells. Alternative splicing targets mammalian SPIRE1 towards mitochondria, where it has a role in fission. In this Review, we summarise the past two decades of SPIRE research by addressing the biochemical and cell biological functions of SPIRE proteins in mammalian reproduction, skin pigmentation and wound healing, as well as in mitochondrial dynamics and host-pathogen interactions.


Subject(s)
Actins , Microfilament Proteins , Animals , Mice , Actins/metabolism , Microfilament Proteins/metabolism , Endothelial Cells/metabolism , Actin Cytoskeleton/metabolism , Formins/metabolism , Mammals/metabolism , Nerve Tissue Proteins/metabolism
2.
Cell Mol Life Sci ; 79(2): 96, 2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35084586

ABSTRACT

Weibel-Palade bodies (WPB) are endothelial cell-specific storage granules that regulate vascular hemostasis by releasing the platelet adhesion receptor von Willebrand factor (VWF) following stimulation. Fusion of WPB with the plasma membrane is accompanied by the formation of actin rings or coats that support the expulsion of large multimeric VWF fibers. However, factor(s) organizing these actin ring structures have remained elusive. We now identify the actin-binding proteins Spire1 and Myosin Vc (MyoVc) as cytosolic factors that associate with WPB and are involved in actin ring formation at WPB-plasma membrane fusion sites. We show that both, Spire1 and MyoVc localize only to mature WPB and that upon Ca2+ evoked exocytosis of WPB, Spire1 and MyoVc together with F-actin concentrate in ring-like structures at the fusion sites. Depletion of Spire1 or MyoVc reduces the number of these actin rings and decreases the amount of VWF externalized to the cell surface after histamine stimulation.


Subject(s)
Calcium/metabolism , Exocytosis , Human Umbilical Vein Endothelial Cells/metabolism , Microfilament Proteins/metabolism , Myosin Type V/metabolism , Nuclear Proteins/metabolism , von Willebrand Factor/metabolism , Blotting, Western , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Microfilament Proteins/genetics , Microscopy, Confocal , Models, Biological , Myosin Type V/genetics , Nuclear Proteins/genetics , RNA Interference , Weibel-Palade Bodies/metabolism
3.
Nat Commun ; 11(1): 3495, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32661310

ABSTRACT

Cell biologists generally consider that microtubules and actin play complementary roles in long- and short-distance transport in animal cells. On the contrary, using melanosomes of melanocytes as a model, we recently discovered that the motor protein myosin-Va works with dynamic actin tracks to drive long-range organelle dispersion in opposition to microtubules. This suggests that in animals, as in yeast and plants, myosin/actin can drive long-range transport. Here, we show that the SPIRE-type actin nucleators (predominantly SPIRE1) are Rab27a effectors that co-operate with formin-1 to generate actin tracks required for myosin-Va-dependent transport in melanocytes. Thus, in addition to melanophilin/myosin-Va, Rab27a can recruit SPIREs to melanosomes, thereby integrating motor and track assembly activity at the organelle membrane. Based on this, we suggest a model in which organelles and force generators (motors and track assemblers) are linked, forming an organelle-based, cell-wide network that allows their collective activity to rapidly disperse the population of organelles long-distance throughout the cytoplasm.


Subject(s)
Actins/metabolism , rab27 GTP-Binding Proteins/metabolism , Cell Biology , Cytoskeleton/metabolism , HEK293 Cells , Humans , Microtubules/metabolism , Organelles , Phylogeny , rab27 GTP-Binding Proteins/genetics
4.
Small GTPases ; 10(2): 111-121, 2019 03.
Article in English | MEDLINE | ID: mdl-28394692

ABSTRACT

Spir actin nucleators and myosin V motor proteins were recently discovered to coexist in a protein complex. The direct interaction allows the coordinated activation of actin motor proteins and actin filament track generation at vesicle membranes. By now the cooperation of myosin V (MyoV) motors and Spir actin nucleation function has only been shown in the exocytic transport of Rab11 vesicles in metaphase mouse oocytes. Next to Rab11, myosin V motors however interact with a variety of Rab GTPases including Rab3, Rab8 and Rab10. As a common theme most of the MyoV interacting Rab GTPases function at different steps along the exocytic transport routes. We here summarize the different transport functions of class V myosins and provide as proof of principle data showing a colocalization of Spir actin nucleators and MyoVa at Rab8a vesicles. This suggests that besides Rab11/MyoV transport also the Rab8/MyoV and possibly other MyoV transport processes recruit Spir actin filament nucleation function.


Subject(s)
Actins/metabolism , Myosin Type V/metabolism , Biological Transport , Humans , rho GTP-Binding Proteins/metabolism
5.
Brain Pathol ; 28(5): 695-709, 2018 09.
Article in English | MEDLINE | ID: mdl-29222813

ABSTRACT

Glioblastoma (GBM) represents the most common and most malignant type of primary brain tumor and significantly contributes to cancer morbidity and mortality. Invasion into the healthy brain parenchyma is a major feature of glioblastoma aggressiveness. Reelin (RELN) is a large secreted extracellular matrix glycoprotein that regulates neuronal migration and positioning in the developing brain and sustains functionality in the adult brain. We here show that both RELN and its main downstream effector DAB1 are silenced in glioblastoma as compared to non-neoplastic tissue and mRNA expression is inversely correlated with malignancy grade. Furthermore, RELN expression is positively correlated with patient survival in two large, independent clinically annotated datasets. RELN silencing occurs via promoter hypermethylation as shown by both database mining and bisulfite sequencing of the RELN promoter. Consequently, treatment with 5'-Azacytidine and trichostatin A induced RELN expression in vitro. On the functional level, we found RELN to regulate glioblastoma cell migration both in a DAB1 (tyrosine phosphorylation)-dependent and -independent fashion, depending on the substrate provided. Moreover, stimulation of RELN signaling strongly reduced proliferation in glioblastoma cells. This phenotype depends on DAB1 stimulation by RELN, as a mutant that lacks all RELN induced tyrosine phosphorylation sites (DAB1-5F) failed to induce a growth arrest. Proteomic analyzes revealed that these effects are mediated by a reduction in E2F targets and dephosphorylation of ERK1/2. Taken together, our data establish a relevance of RELN signaling in glioblastoma pathology and thereby might unearth novel, yet unrecognized treatment options.


Subject(s)
Brain Neoplasms/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/physiology , Extracellular Matrix Proteins/metabolism , Glioblastoma/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/metabolism , Brain/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Computer Simulation , Extracellular Matrix Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice , Nerve Tissue Proteins/genetics , Proteome , RNA, Messenger/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Signal Transduction , Young Adult
6.
J Cell Sci ; 130(20): 3427-3435, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-29032357

ABSTRACT

The actin cytoskeleton and associated motor proteins provide the driving forces for establishing the astonishing morphological diversity and dynamics of mammalian cells. Aside from functions in protruding and contracting cell membranes for motility, differentiation or cell division, the actin cytoskeleton provides forces to shape and move intracellular membranes of organelles and vesicles. To establish the many different actin assembly functions required in time and space, actin nucleators are targeted to specific subcellular compartments, thereby restricting the generation of specific actin filament structures to those sites. Recent research has revealed that targeting and activation of actin filament nucleators, elongators and myosin motors are tightly coordinated by conserved protein complexes to orchestrate force generation. In this Cell Science at a Glance article and the accompanying poster, we summarize and discuss the current knowledge on the corresponding protein complexes and their modes of action in actin nucleation, elongation and force generation.


Subject(s)
Actin Cytoskeleton/physiology , Pseudopodia/physiology , Actin Cytoskeleton/ultrastructure , Actins/physiology , Actins/ultrastructure , Animals , Cell Physiological Phenomena , Cells, Cultured , Humans , Protein Multimerization , Pseudopodia/ultrastructure
7.
PLoS One ; 11(9): e0161965, 2016.
Article in English | MEDLINE | ID: mdl-27627128

ABSTRACT

Salmonella Typhimurium (S. Tm) is a leading cause of diarrhea. The disease is triggered by pathogen invasion into the gut epithelium. Invasion is attributed to the SPI-1 type 3 secretion system (T1). T1 injects effector proteins into epithelial cells and thereby elicits rearrangements of the host cellular actin cytoskeleton and pathogen invasion. The T1 effector proteins SopE, SopB, SopE2 and SipA are contributing to this. However, the host cell factors contributing to invasion are still not completely understood. To address this question comprehensively, we used Hela tissue culture cells, a genome-wide siRNA library, a modified gentamicin protection assay and S. TmSipA, a sopBsopE2sopE mutant which strongly relies on the T1 effector protein SipA to invade host cells. We found that S. TmSipA invasion does not elicit membrane ruffles, nor promote the entry of non-invasive bacteria "in trans". However, SipA-mediated infection involved the SPIRE family of actin nucleators, besides well-established host cell factors (WRC, ARP2/3, RhoGTPases, COPI). Stage-specific follow-up assays and knockout fibroblasts indicated that SPIRE1 and SPIRE2 are involved in different steps of the S. Tm infection process. Whereas SPIRE1 interferes with bacterial binding, SPIRE2 influences intracellular replication of S. Tm. Hence, these two proteins might fulfill non-redundant functions in the pathogen-host interaction. The lack of co-localization hints to a short, direct interaction between S. Tm and SPIRE proteins or to an indirect effect.


Subject(s)
Bacterial Proteins/physiology , Genome-Wide Association Study/methods , Host-Pathogen Interactions/physiology , Microfilament Proteins/physiology , Nuclear Proteins/physiology , Salmonella typhimurium/pathogenicity , Animals , Cell Line , Fluorescent Antibody Technique , HeLa Cells/metabolism , HeLa Cells/microbiology , Humans , Mice , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Salmonella typhimurium/physiology
8.
Elife ; 52016 09 13.
Article in English | MEDLINE | ID: mdl-27623148

ABSTRACT

There is growing evidence for a coupling of actin assembly and myosin motor activity in cells. However, mechanisms for recruitment of actin nucleators and motors on specific membrane compartments remain unclear. Here we report how Spir actin nucleators and myosin V motors coordinate their specific membrane recruitment. The myosin V globular tail domain (MyoV-GTD) interacts directly with an evolutionarily conserved Spir sequence motif. We determined crystal structures of MyoVa-GTD bound either to the Spir-2 motif or to Rab11 and show that a Spir-2:MyoVa:Rab11 complex can form. The ternary complex architecture explains how Rab11 vesicles support coordinated F-actin nucleation and myosin force generation for vesicle transport and tethering. New insights are also provided into how myosin activation can be coupled with the generation of actin tracks. Since MyoV binds several Rab GTPases, synchronized nucleator and motor targeting could provide a common mechanism to control force generation and motility in different cellular processes.


Subject(s)
Cytoplasmic Vesicles/metabolism , Membranes/metabolism , Microfilament Proteins/metabolism , Myosin Type V/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Crystallography, X-Ray , Mice , Microfilament Proteins/chemistry , Models, Molecular , Myosin Type V/chemistry , Protein Binding , Protein Conformation , Protein Multimerization , rab GTP-Binding Proteins/chemistry
9.
J Biol Chem ; 290(10): 6428-44, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25564607

ABSTRACT

Spir and formin (FMN)-type actin nucleators initiate actin polymerization at vesicular membranes necessary for long range vesicular transport processes. Here we studied in detail the membrane binding properties and protein/protein interactions that govern the assembly of the membrane-associated Spir·FMN complex. Using biomimetic membrane models we show that binding of the C-terminal Spir-2 FYVE-type zinc finger involves both the presence of negatively charged lipids and hydrophobic contributions from the turret loop that intrudes the lipid bilayer. In solution, we uncovered a yet unknown intramolecular interaction between the Spir-2 FYVE-type domain and the N-terminal kinase non-catalytic C-lobe domain (KIND) that could not be detected in the membrane-bound state. Interestingly, we found that the intramolecular Spir-2 FYVE/KIND and the trans-regulatory Fmn-2-FSI/Spir-2-KIND interactions are competitive. We therefore characterized co-expressed Spir-2 and Fmn-2 fluorescent protein fusions in living cells by fluorescence cross-correlation spectroscopy. The data corroborate a model according to which Spir-2 exists in two different states, a cytosolic monomeric conformation and a membrane-bound state in which the KIND domain is released and accessible for subsequent Fmn-2 recruitment. This sequence of interactions mechanistically couples membrane binding of Spir to the recruitment of FMN, a pivotal step for initiating actin nucleation at vesicular membranes.


Subject(s)
Actins/metabolism , Microfilament Proteins/chemistry , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Actins/chemistry , Amino Acid Sequence , Formins , HEK293 Cells , Humans , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Nuclear Proteins/metabolism , Protein Interaction Maps/genetics
10.
Trends Cell Biol ; 24(7): 407-15, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24675420

ABSTRACT

The organization of cells into interconnected structures such as animal tissues requires a sophisticated system directing receptors and adhesion proteins to the cell surface. The Rab11 small G proteins (Rab11a, b, and Rab25) of the Ras superfamily are master regulators of the surface expression of receptors and adhesion proteins. Acting as a molecular switch, Rab11 builds distinct molecular machinery such as motor protein complexes and the exocyst to transport proteins to the cell surface. Recent evidence reveals Rab11 localization at the trans-Golgi network (TGN), post-Golgi vesicles, and the recycling endosome, placing it at the intersection between the endocytic and exocytic trafficking pathways. We review Rab11 in various cellular contexts, and discuss its regulation and mechanisms by which Rab11 couples with effector proteins.


Subject(s)
Membrane Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Endosomes/metabolism , Endosomes/physiology , Golgi Apparatus/metabolism , Golgi Apparatus/physiology , Humans , Protein Transport/physiology , trans-Golgi Network/metabolism , trans-Golgi Network/physiology
11.
Eur J Cell Biol ; 93(5-6): 225-37, 2014.
Article in English | MEDLINE | ID: mdl-24345451

ABSTRACT

Spir proteins nucleate actin filaments at vesicle membranes and facilitate intracellular transport processes. The mammalian genome encodes two Spir proteins, namely Spir-1 and Spir-2. While the mouse spir-2 gene has a rather broad expression pattern, high levels of spir-1 expression are restricted to the nervous system, oocytes, and testis. Spir-1 mutant mice generated by a gene trap method have been employed to address Spir-1 function during mouse development and in adult mouse tissues, with a specific emphasis on viability, reproduction, and the nervous system. The gene trap cassette disrupts Spir-1 expression between the N-terminal KIND domain and the WH2 domain cluster. Spir-1 mutant mice are viable and were born in a Mendelian ratio. In accordance with the redundant function of Spir-1 and Spir-2 in oocyte maturation, spir-1 mutant mice are fertile. The overall brain anatomy of spir-1 mutant mice is not altered and visual and motor functions of the mice remain normal. Microscopic analysis shows a slight reduction in the number of dendritic spines on cortical neurons. Detailed behavioral studies of the spir-1 mutant mice, however, unveiled a very specific and highly significant phenotype in terms of fear learning in male mice. In contextual and cued fear conditioning experiments the male spir-1 mutant mice display increased fear memory when compared to their control littermates. Our data point toward a particular function of the vesicle associated Spir-1 actin organizer in neuronal circuits determining fear behavior.


Subject(s)
Actins/metabolism , Fear/psychology , Microfilament Proteins/genetics , Nerve Tissue Proteins/genetics , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Conditioning, Classical , Dendrites/metabolism , Dendritic Spines/ultrastructure , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Mutant Strains , Microfilament Proteins/metabolism , Motor Activity , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Transport Vesicles/genetics , Transport Vesicles/metabolism , Visual Perception
12.
Biol Chem ; 394(12): 1649-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23863697

ABSTRACT

The diversity of cellular actin functions is attained by the activation of actin nucleator complexes, which initiate the polymerization of actin monomers into a helical double-stranded filament at defined subcellular compartments. Next to actin functions at the cell membrane, including different forms of membrane protrusions and invaginations, actin dynamics at intracellular membranes has recently become a research focus. Experiments addressing the vesicle-associated Spir WH2 domain containing actin nucleators have provided novel mechanistic insights into the function of actin dynamics at intracellular membranes. Spir proteins are targeted by a modified FYVE zinc finger motif toward endosomal and vesicle membranes, where they interact and cooperate with the distinct nucleators of the FMN subfamily of formins in the nucleation of actin filaments. The function of the Spir/formin actin nucleator complex is closely related to the Rab11 small G protein, which is a key regulator of recycling and exocytic transport processes. Together with the actin motor protein and Rab11 effector myosin Vb, Spir/formin nucleated actin filaments mediate actin-dependent vesicle transport processes. Drosophila and mouse genetic studies as well as cell biology experiments point toward an important role of the Spir/formin complex in oocyte maturation and in the structure and signaling of the nervous system.


Subject(s)
Actins/metabolism , Drosophila Proteins/metabolism , Fetal Proteins/metabolism , Microfilament Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Drosophila Proteins/genetics , Formins , Humans , Microfilament Proteins/genetics , Multiprotein Complexes/metabolism , Nerve Tissue Proteins , Oocytes/physiology , Protein Structure, Tertiary , Signal Transduction , Transport Vesicles/metabolism , rab GTP-Binding Proteins/metabolism
13.
J Biol Chem ; 286(35): 30732-30739, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21705804

ABSTRACT

The distinct actin nucleation factors of the Spir and formin subgroup families cooperate in actin nucleation. The Spir/formin cooperativity has been identified to direct two essential steps in mammalian oocyte maturation, the asymmetric spindle positioning and polar body extrusion during meiosis. Understanding the nature and regulation of the Spir/Fmn cooperation is an important requirement to comprehend mammalian reproduction. Recently we dissected the structural elements of the Spir and Fmn family proteins, which physically link the two actin nucleation factors. The trans-regulatory interaction is mediated by the Spir kinase non-catalytic C-lobe domain (KIND) and the C-terminal formin Spir interaction motif (FSI). The interaction inhibits formin nucleation activity and enhances the Spir activity. To get insights into the molecular mechanism of the Spir/Fmn interaction, we determined the crystal structure of the KIND domain alone and in complex with the C-terminal Fmn-2 FSI peptide. Together they confirm the proposed structural homology of the KIND domain to the protein kinase fold and reveal the basis of the Spir/formin interaction. The complex structure showed a large interface with conserved and positively charged residues of the Fmn FSI peptide mediating major contacts to an acidic groove on the surface of KIND. Protein interaction studies verified the electrostatic nature of the interaction. The data presented here provide the molecular basis of the Spir/formin interaction and give a first structural view into the mechanisms of actin nucleation factor cooperativity.


Subject(s)
Actins/chemistry , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , Catalysis , Crystallization , Crystallography, X-Ray/methods , Formins , HeLa Cells , Humans , Mice , Molecular Sequence Data , Oocytes/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Surface Properties
14.
Curr Biol ; 21(11): 955-60, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21620703

ABSTRACT

Oocytes mature into eggs by extruding half of their chromosomes in a small cell termed the polar body. Asymmetric oocyte division is essential for fertility [1], but despite its importance, little is known about its mechanism. In mammals, the meiotic spindle initially forms close to the center of the oocyte. Thus, two steps are required for asymmetric meiotic division: first, asymmetric spindle positioning and second, polar body extrusion. Here, we identify Spire1 and Spire2 as new key factors in asymmetric division of mouse oocytes. Spire proteins are novel types of actin nucleators that drive nucleation of actin filaments with their four WH2 actin-binding domains [2-6]. We show that Spire1 and Spire2 first mediate asymmetric spindle positioning by assembling an actin network that serves as a substrate for spindle movement. Second, they drive polar body extrusion by promoting assembly of the cleavage furrow. Our data suggest that Spire1 and Spire2 cooperate with Formin-2 (Fmn2) to nucleate actin filaments in mouse oocytes and that both types of nucleators act as a functional unit. This study not only reveals how Spire1 and Spire2 drive two critical steps of asymmetric oocyte division, but it also uncovers the first physiological function of Spire-type actin nucleators in vertebrates.


Subject(s)
Cell Division/physiology , Microfilament Proteins/physiology , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Oocytes/cytology , Actins/metabolism , Animals , Cell Division/genetics , Cell Polarity , Formins , Mice , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nuclear Proteins/metabolism , Oocytes/metabolism , Oocytes/ultrastructure , RNA Interference , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure
15.
PLoS One ; 6(5): e19931, 2011.
Article in English | MEDLINE | ID: mdl-21603613

ABSTRACT

The actin cytoskeleton is continuously remodeled through cycles of actin filament assembly and disassembly. Filaments are born through nucleation and shaped into supramolecular structures with various essential functions. These range from contractile and protrusive assemblies in muscle and non-muscle cells to actin filament comets propelling vesicles or pathogens through the cytosol. Although nucleation has been extensively studied using purified proteins in vitro, dissection of the process in cells is complicated by the abundance and molecular complexity of actin filament arrays. We here describe the ectopic nucleation of actin filaments on the surface of microtubules, free of endogenous actin and interfering membrane or lipid. All major mechanisms of actin filament nucleation were recapitulated, including filament assembly induced by Arp2/3 complex, formin and Spir. This novel approach allows systematic dissection of actin nucleation in the cytosol of live cells, its genetic re-engineering as well as screening for new modifiers of the process.


Subject(s)
Actin Cytoskeleton/metabolism , Microtubules/metabolism , Actins/metabolism , Animals , Fluorescence Recovery After Photobleaching , Mice , Microscopy , Polymerization
16.
Eur J Cell Biol ; 90(11): 922-5, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21129813

ABSTRACT

The assembly of actin monomers into filaments is a highly regulated basic cellular function. The structural organization of a cell, morphological changes or cell motility is dependent on actin filament dynamics. While within the last decade substantial knowledge has been acquired about actin dynamics at the cell membrane, today only little is known about the actin cytoskeleton and its functions at intracellular endosomal and organelle membranes. The Spir actin nucleators are specifically targeted towards endosomal membranes by a FYVE zinc finger membrane localization domain, and provide an important link to study the role of actin dynamics in the regulation of intracellular membrane transport. Spir proteins are the founding members of a novel class of actin nucleation factors, which initiate actin polymerization by binding of actin monomers to one or multiple Wiskott-Aldrich syndrome protein (WASp) homology 2 (WH2) domains. Although Spir proteins can nucleate actin polymerization in vitro by themselves, they form a regulatory complex with the distinct actin nucleators of the formin subgroup (Fmn) of formins. A cooperative mechanism in actin nucleation has been proposed. Ongoing studies on the function and regulation of the Spir proteins in vesicle transport processes will reveal important insights into actin dynamics at intracellular membranes and how this regulates the highly directed and controlled routes of intracellular membrane trafficking.


Subject(s)
Actins/metabolism , Cell Nucleus/metabolism , Fetal Proteins/metabolism , Intracellular Membranes/metabolism , Microfilament Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Formins , Humans , Protein Binding
17.
Gene Expr Patterns ; 10(7-8): 345-50, 2010.
Article in English | MEDLINE | ID: mdl-20723621

ABSTRACT

Spir proteins are the founding members of the novel class of WH2 domain containing actin nucleation factors. They initiate actin polymerization by binding of actin monomers to four WH2 domains in the central part of the proteins. Despite their ability to nucleate actin polymerization in vitro by themselves, Spir proteins form a regulatory complex with the distinct actin nucleators of the formin subgroup of formins. The mammalian genome encodes two spir genes, spir-1 and spir-2. The corresponding proteins have an identical structural array and share a high degree of homology. Here, we have addressed the yet unknown expression of the mouse spir-2 gene. Northern blot analysis revealed that the spir-2 gene is expressed as a single mRNA. During embryogenesis in situ hybridizations show spir-2 to be expressed in the developing nervous system and intestine. In adult mouse tissues highest expression of spir-2 was detected in the epithelial cells of the digestive tract and in neuronal cells of the nervous system. High expression was also detected in testical spermatocytes. In contrast to the restricted expression of the mouse spir-1 gene, which is mainly found in the nervous system, our data presented here show a distinct and broader expression pattern of the spir-2 gene and by this support a more general cell biological function of the novel actin nucleators.


Subject(s)
Actins/metabolism , Central Nervous System/metabolism , Gene Expression , Intestinal Mucosa/metabolism , Microfilament Proteins/genetics , Actin Cytoskeleton/metabolism , Actins/biosynthesis , Actins/genetics , Animals , Blotting, Northern , Central Nervous System/cytology , Central Nervous System/embryology , Embryonic Development , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gastrointestinal Tract/cytology , Gastrointestinal Tract/metabolism , In Situ Hybridization , Intestines/embryology , Male , Mice , Microfilament Proteins/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatocytes/cytology , Spermatocytes/metabolism
18.
J Biol Chem ; 284(37): 25324-33, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19605360

ABSTRACT

The actin nucleation factors Spire and Cappuccino interact with each other and regulate essential cellular events during Drosophila oogenesis in a cooperative fashion. The interaction blocks formin actin nucleation activity and enhances the Spire activity. Analogous to Spire and Cappuccino, the mammalian homologs Spir-1 and formin-2 show a regulatory interaction. To get an understanding of the nature of the Spir-formin cooperation, we have analyzed the interaction biochemically and biophysically. Our data shows that the association of Spir-1 and formin-2 is not significantly mediated by binding of the Spir-1-KIND domain to the formin FH2 core domain. Instead, a short sequence motif C-terminal adjacent to the formin-2-FH2 domain could be characterized that mediates the interaction and is conserved among the members of the Fmn subgroup of formins. In line with this, we found that both mammalian Spir proteins, Spir-1 and Spir-2, interact with mammalian Fmn subgroup proteins formin-1 and formin-2.


Subject(s)
Fetal Proteins/chemistry , Microfilament Proteins/chemistry , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Amino Acid Sequence , Animals , Anisotropy , Cell Proliferation , Formins , HeLa Cells , Humans , Microscopy, Fluorescence/methods , Models, Biological , Molecular Sequence Data , Oogenesis , Protein Structure, Tertiary , Sequence Homology, Amino Acid
20.
J Cell Biol ; 179(1): 117-28, 2007 Oct 08.
Article in English | MEDLINE | ID: mdl-17923532

ABSTRACT

Spire and Cappuccino are actin nucleation factors that are required to establish the polarity of Drosophila melanogaster oocytes. Their mutant phenotypes are nearly identical, and the proteins interact biochemically. We find that the interaction between Spire and Cappuccino family proteins is conserved across metazoan phyla and is mediated by binding of the formin homology 2 (FH2) domain from Cappuccino (or its mammalian homologue formin-2) to the kinase noncatalytic C-lobe domain (KIND) from Spire. In vitro, the KIND domain is a monomeric folded domain. Two KIND monomers bind each FH2 dimer with nanomolar affinity and strongly inhibit actin nucleation by the FH2 domain. In contrast, formation of the Spire-Cappuccino complex enhances actin nucleation by Spire. In Drosophila oocytes, Spire localizes to the cortex early in oogenesis and disappears around stage 10b, coincident with the onset of cytoplasmic streaming.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microfilament Proteins/metabolism , Actins/metabolism , Animals , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microtubules/metabolism , Oogenesis , Protein Interaction Domains and Motifs , Protein Interaction Mapping
SELECTION OF CITATIONS
SEARCH DETAIL
...